Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
PLoS Pathog ; 11(12): e1005311, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26633895

RESUMEN

For many emerging and re-emerging infectious diseases, definitive solutions via sterilizing adaptive immunity may require years or decades to develop, if they are even possible. The innate immune system offers alternative mechanisms that do not require antigen-specific recognition or a priori knowledge of the causative agent. However, it is unclear whether effective stable innate immune system activation can be achieved without triggering harmful autoimmunity or other chronic inflammatory sequelae. Here, we show that transgenic expression of a picornavirus RNA-dependent RNA polymerase (RdRP), in the absence of other viral proteins, can profoundly reconfigure mammalian innate antiviral immunity by exposing the normally membrane-sequestered RdRP activity to sustained innate immune detection. RdRP-transgenic mice have life-long, quantitatively dramatic upregulation of 80 interferon-stimulated genes (ISGs) and show profound resistance to normally lethal viral challenge. Multiple crosses with defined knockout mice (Rag1, Mda5, Mavs, Ifnar1, Ifngr1, and Tlr3) established that the mechanism operates via MDA5 and MAVS and is fully independent of the adaptive immune system. Human cell models recapitulated the key features with striking fidelity, with the RdRP inducing an analogous ISG network and a strict block to HIV-1 infection. This RdRP-mediated antiviral mechanism does not depend on secondary structure within the RdRP mRNA but operates at the protein level and requires RdRP catalysis. Importantly, despite lifelong massive ISG elevations, RdRP mice are entirely healthy, with normal longevity. Our data reveal that a powerfully augmented MDA5-mediated activation state can be a well-tolerated mammalian innate immune system configuration. These results provide a foundation for augmenting innate immunity to achieve broad-spectrum antiviral protection.


Asunto(s)
Genes Virales/inmunología , Inmunidad Innata/inmunología , ARN Polimerasa Dependiente del ARN/inmunología , Proteínas Virales/inmunología , Animales , Western Blotting , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunidad Innata/genética , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Picornaviridae/genética , Picornaviridae/inmunología , ARN Polimerasa Dependiente del ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virosis/inmunología , Virosis/prevención & control
2.
Neuroimage ; 105: 181-8, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25451479

RESUMEN

Thalamic deep brain stimulation (DBS) is an FDA-approved neurosurgical treatment for medication-refractory essential tremor. Its therapeutic benefit is highly dependent upon stimulation frequency and voltage parameters. We investigated these stimulation parameter-dependent effects on neural network activation by performing functional magnetic resonance imaging (fMRI) during DBS of the ventral lateral (VL) thalamus and comparing the blood oxygenation level-dependent (BOLD) signals induced by multiple stimulation parameter combinations in a within-subject study of swine. Low (10 Hz) and high (130 Hz) frequency stimulation was applied at 3, 5, and 7 V in the VL thalamus of normal swine (n = 5). We found that stimulation frequency and voltage combinations differentially modulated the brain network activity in the sensorimotor cortex, the basal ganglia, and the cerebellum in a parameter-dependent manner. Notably, in the motor cortex, high frequency stimulation generated a negative BOLD response, while low frequency stimulation increased the positive BOLD response. These frequency-dependent differential effects suggest that the VL thalamus is an exemplary target for investigating functional network connectivity associated with therapeutic DBS.


Asunto(s)
Estimulación Encefálica Profunda , Corteza Motora/fisiología , Vías Nerviosas/fisiología , Núcleos Talámicos Ventrales/fisiología , Animales , Ganglios Basales/fisiología , Imagen por Resonancia Magnética , Masculino , Corteza Sensoriomotora/fisiología , Porcinos
3.
BMC Genomics ; 14: 678, 2013 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-24090483

RESUMEN

BACKGROUND: Blood-brain barrier (BBB) disruption is an integral feature of numerous neurological disorders. However, there is a relative lack of knowledge regarding the underlying molecular mechanisms of immune-mediated BBB disruption. We have previously shown that CD8 T cells and perforin play critical roles in initiating altered permeability of the BBB in the peptide-induced fatal syndrome (PIFS) model developed by our laboratory. Additionally, despite having indistinguishable CD8 T cell responses, C57BL/6J (B6) mice are highly susceptible to PIFS, exhibiting functional motor deficits, increased astrocyte activation, and severe CNS vascular permeability, while 129S1/SvImJ (129S1) mice remain resistant. Therefore, to investigate the potential role of genetic factors, we performed a comprehensive genetic analysis of (B6 x 129S1) F2 progeny to define quantitative trait loci (QTL) linked to the phenotypic characteristics stated above that mediate CD8 T cell-initiated BBB disruption. RESULTS: Using single nucleotide polymorphism (SNP) markers and a 95% confidence interval, we identified one QTL (PIFS1) on chromosome 12 linked to deficits in motor function (SNP markers rs6292954, rs13481303, rs3655057, and rs13481324, LOD score = 3.3). In addition we identified a second QTL (PIFS2) on chromosome 17 linked to changes in CNS vascular permeability (SNP markers rs6196216 and rs3672065, LOD score = 3.7). CONCLUSIONS: The QTL critical intervals discovered have allowed for compilation of a list of candidate genes implicated in regulating functional deficit and CNS vascular permeability. These genes encode for factors that may be potential targets for therapeutic approaches to treat disorders characterized by CD8 T cell-mediated BBB disruption.


Asunto(s)
Barrera Hematoencefálica/patología , Barrera Hematoencefálica/fisiopatología , Linfocitos T CD8-positivos/inmunología , Permeabilidad Capilar/genética , Estudios de Asociación Genética , Sitios de Carácter Cuantitativo/genética , Animales , Astrocitos/patología , Barrera Hematoencefálica/inmunología , Permeabilidad Capilar/inmunología , Distribución de Chi-Cuadrado , Ratones , Ratones Endogámicos C57BL , Actividad Motora , Carácter Cuantitativo Heredable , Síndrome
4.
J Clin Immunol ; 33 Suppl 1: S50-6, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22990667

RESUMEN

We have discovered a role for natural autoantibodies in central nervous system repair, remyelination and axon protection. These natural human antibodies are of the immunoglobulin M (IgM) isotype, and they bind to the surface of neural cells. The epitope of the antibody includes sialic acid because treatment with sialidase disrupts the binding. A fully human recombinant form of one of these IgMs, rHIgM12, has the same properties as the serum-derived IgM. rHIgM12 enhanced polarized axonal outgrowth from primary neurons when presented as a substrate in vitro and improved motor functions in chronically Theiler's virus-infected SJL mice, a model of MS. rHIgM12 bound to neuronal surfaces and induced cholesterol and ganglioside (GM1) clustering, indicating that rHIgM12 functions through a mechanism of axonal membrane stabilization. Our work demonstrates that a natural human neuron-binding IgM can regulate membrane domain dynamics. This antibody has the potential to improve neurologic disease.


Asunto(s)
Inmunoglobulina M/inmunología , Microdominios de Membrana/metabolismo , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Neuronas/inmunología , Animales , Axones/efectos de los fármacos , Axones/inmunología , Axones/metabolismo , Modelos Animales de Enfermedad , Humanos , Inmunoglobulina M/metabolismo , Inmunoglobulina M/farmacología , Microdominios de Membrana/efectos de los fármacos , Ratones , Esclerosis Múltiple/tratamiento farmacológico , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/inmunología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología
5.
Proc Natl Acad Sci U S A ; 107(2): 792-7, 2010 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-20080754

RESUMEN

The potential for endogenous remyelination and axonal protection can be an important factor in determining disease outcome in demyelinating diseases like multiple sclerosis. In many multiple sclerosis (MS) patients CNS repair fails or is incomplete whereas in others the disease is accompanied by extensive repair of demyelinated lesions. We have described significant differences in the ability of two strains of mice to repair CNS damage following Theiler's virus-induced demyelination: FVB/NJ (FVB) mice repair damaged myelin spontaneously and completely, whereas B10.D1-H2(q)/SgJ (B10.Q) mice are deficient in the repair process. A QTL analysis was performed to identify genetic loci that differentially regulate CNS repair following chronic demyelination in these strains and two QTL were detected: one on chromosome 3 with a LOD score of 9.3 and a second on chromosome 9 with a LOD score of 14.0. The mouse genes for epidermal growth factor (EGF) and Tyk2 are encoded within the QTL on chromosomes 3 and 9, respectively. Sequence polymorphisms between the FVB and B10.Q strains at both the EGF and Tyk2 loci define functional variations consistent with roles for these genes in regulating myelin repair. EGF is a key regulator of cell growth and development and we show a sevenfold increase in EGF expression in FVB compared to B10.Q mice. Tyk2 is a Janus kinase that plays a central role in controlling the T(H)1 immune response and we show that attenuation of Tyk2 function correlates with enhanced CNS repair.


Asunto(s)
Enfermedades Desmielinizantes/genética , Factor de Crecimiento Epidérmico/genética , Variación Genética , Ratones Endogámicos/genética , Vaina de Mielina/genética , TYK2 Quinasa/genética , Alelos , Animales , Cruzamientos Genéticos , Daño del ADN , Reparación del ADN , Ratones , Sitios de Carácter Cuantitativo/genética , Receptores de Eritropoyetina/genética
6.
J Neurochem ; 119(1): 100-12, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21824142

RESUMEN

Mouse and human IgMs support neurite extension from primary cerebellar granule neurons. In this study using primary hippocampal and cortical neurons, we demonstrate that a recombinant human IgM, rHIgM12, promotes axon outgrowth by coupling membrane domains (lipid rafts) to microtubules. rHIgM12 binds to the surface of neuron and induces clustering of cholesterol and ganglioside GM1. After cell binding and membrane fractionation, rHIgM12 gets segregated into two pools, one associated with lipid raft fractions and the other with the detergent-insoluble cytoskeleton-containing pellet. Membrane-bound rHIgM12 co-localized with microtubules and co-immuno precipitated with ß3-tubulin. rHIgM12-membrane interaction also enhanced the tyrosination of α-tubulin indicating a stabilization of new neurites. When presented as a substrate, rHIgM12 induced axon outgrowth from primary neurons. We now demonstrate that a recombinant human mAb can induce signals in neurons that regulate membrane lipids and microtubule dynamics required for axon extension. We propose that the pentameric structure of the IgM is critical to cross-link membrane lipids and proteins resulting in signaling cascades.


Asunto(s)
Axones/fisiología , Inmunoglobulina M/fisiología , Microdominios de Membrana/fisiología , Microtúbulos/fisiología , Animales , Caveolina 1/metabolismo , Células Cultivadas , Centrifugación por Gradiente de Densidad , Colesterol/metabolismo , Gangliósido G(M1)/metabolismo , Hipocampo/citología , Hipocampo/efectos de los fármacos , Humanos , Inmunohistoquímica , Inmunoprecipitación , Ratones , Neurogénesis/fisiología , Proteínas Recombinantes/farmacología , Transducción de Señal/fisiología , Tubulina (Proteína)/metabolismo , Tirosina/metabolismo
7.
Neurobiol Dis ; 43(2): 330-7, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21514385

RESUMEN

Platinum-based compounds are widely used and effective chemotherapeutic agents; however, sensory peripheral neuropathy is a dose-limiting and long term side effect for 20-30% of patients. A critical question is whether the mechanisms of cell death underlying clinical efficacy can be separated from the effects on neurons in order to develop strategies that prevent platinum-induced neuropathy. In rodent dorsal root ganglion neurons (DRG), cisplatin has been shown to bind and damage neuronal DNA, inducing apoptosis; however genetic manipulation in order to study mechanisms of this phenomenon in the rodent model system is costly and time-consuming. Drosophila melanogaster are commonly used to study neurological disorders, have DNA damage-apoptosis mechanisms homologous to mammalian systems, and have readily-available, inexpensive tools for rapid genetic manipulation. We therefore sought to develop adult Drosophila as a new model to study cisplatin-induced neurotoxicity. Adult Drosophila were exposed to 10, 25, 50, 100, 200 and 400 µg/ml cisplatin for 3 days and observed for fly survival and geotactic climbing behavior, cisplatin-DNA binding and cellular apoptosis. On day 3, 50 µg/ml cisplatin reduced the number of flies able to climb above 2 cm to 43% while fly survival was maintained at 92%. 100% lethality was observed at 400 µg/ml cisplatin. Whole fly platinum-genomic DNA adducts were measured and found to be comparable to adduct levels previously measured in rat DRG neurons. Brain, ovaries, kidney and heart harvested from cisplatin treated flies were stained for active caspase 3. Apoptosis was found in ovaries and brain but not in heart and kidney. Brain apoptosis was confirmed by transmission electron microscopy. Expression of the anti-apoptotic baculoviral protein, p35, in neurons using the GAL4-UAS system prevented cisplatin-induced apoptosis in the brain and restored climbing behavior. In conclusion, cisplatin-induced behavioral and apoptotic changes in Drosophila resemble those seen in mammals. Furthermore, the use of lethality and climbing assays combined with powerful gene manipulation, make Drosophila a suitable model to study mechanisms of cisplatin neurotoxicity.


Asunto(s)
Cisplatino/toxicidad , Modelos Animales de Enfermedad , Drosophila melanogaster/efectos de los fármacos , Degeneración Nerviosa/inducido químicamente , Neurotoxinas/toxicidad , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Femenino , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/virología , Proteínas Virales/biosíntesis , Proteínas Virales/genética
8.
J Autoimmun ; 37(2): 144-50, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21624820

RESUMEN

Immunoglobulins (Ig) or antibodies are heavy plasma proteins, with sugar chains added to amino acid residues by N-linked glycosylation and occasionally by O-linked glycosylation. The versatility of antibodies is demonstrated by the various functions that they mediate such as neutralization, agglutination, fixation with activation of complement and activation of effector cells. In addition to this plethora of functions, some antibodies express enzymatic activity. Antibodies endowed with enzymatic properties have been described in human autoimmune manifestations in a variety of disorders such as autoimmune thyroiditis, systemic erythematosus (SLE), scleroderma, rheumatoid arthritis (RA), multiple sclerosis (MS) and acquired hemophilia (AH). Antibodies isolated from these conditions were able to specifically hydrolyze thyroglobulin, DNA, RNA, myelin basic protein (MBP), and factor VIII (FVIII) or factor IX (FIX), respectively. The therapeutic relevance of these findings is discussed.


Asunto(s)
Anticuerpos Catalíticos/metabolismo , Autoanticuerpos/metabolismo , Autoantígenos/metabolismo , Enfermedades Autoinmunes/enzimología , Enfermedades Autoinmunes/inmunología , Animales , Anticuerpos Catalíticos/inmunología , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Enfermedades Autoinmunes/tratamiento farmacológico , Humanos , Hidrólisis , Inmunoterapia/tendencias
9.
Mol Pain ; 6: 15, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20205720

RESUMEN

BACKGROUND: Cisplatin is primarily used for treatment of ovarian and testicular cancer. Oxaliplatin is the only effective treatment for metastatic colorectal cancer. Both are known to cause dose related, cumulative toxic effects on the peripheral nervous system and thirty to forty percent of cancer patients receiving these agents experience painful peripheral neuropathy. The mechanisms underlying painful platinum-induced neuropathy remain poorly understood. Previous studies have demonstrated important roles for TRPV1, TRPM8, and TRPA1 in inflammation and nerve injury induced pain. RESULTS: In this study, using real-time, reverse transcriptase, polymerase chain reaction (RT-PCR), we analyzed the expression of TRPV1, TRPM8, and TRPA1 induced by cisplatin or oxaliplatin in vitro and in vivo. For in vitro studies, cultured E15 rat dorsal root ganglion (DRG) neurons were treated for up to 48 hours with cisplatin or oxaliplatin. For in vivo studies, trigeminal ganglia (TG) were isolated from mice treated with platinum drugs for three weeks. We show that cisplatin and oxaliplatin-treated DRG neurons had significantly increased in TRPV1, TRPA1, and TRPM8 mRNA expression. TG neurons from cisplatin treated mice had significant increases in TRPV1 and TRPA1 mRNA expression while oxaliplatin strongly induced only TRPA1. Furthermore, compared to the cisplatin-treated wild-type mice, cisplatin-treated TRPV1-null mice developed mechanical allodynia but did not exhibit enhancement of noxious heat- evoked pain responses. Immunohistochemistry studies showed that cisplatin-treated mice had no change in the proportion of the TRPV1 immunopositive TG neurons. CONCLUSION: These results indicate that TRPV1 and TRPA1 could contribute to the development of thermal hyperalgesia and mechanical allodynia following cisplatin-induced painful neuropathy but that TRPV1 has a crucial role in cisplatin-induced thermal hyperalgesia in vivo.


Asunto(s)
Cisplatino/toxicidad , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Antineoplásicos/toxicidad , Células Cultivadas , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Hiperalgesia/fisiopatología , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nociceptores/efectos de los fármacos , Nociceptores/metabolismo , Compuestos Organoplatinos/toxicidad , Oxaliplatino , Enfermedades del Sistema Nervioso Periférico/fisiopatología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Canal Catiónico TRPA1 , Canales Catiónicos TRPM/efectos de los fármacos , Canales Catiónicos TRPM/metabolismo , Canales Catiónicos TRPV/efectos de los fármacos , Canales Catiónicos TRPV/genética , Canales de Potencial de Receptor Transitorio/efectos de los fármacos , Canales de Potencial de Receptor Transitorio/metabolismo
10.
J Virol ; 83(23): 12279-89, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19759133

RESUMEN

The RNA-dependent RNA polymerase 3D(pol) is required for the elongation of positive- and negative-stranded picornavirus RNA. During the course of investigating the effect of the transgenic expression of viral genes on the host immune response, we evaluated the viral load present in the host after infection. To our surprise, we found that 3D transgenic expression in genetically susceptible FVB mice led to substantially lower viral loads after infection with Theiler's murine encephalomyelitis virus (TMEV). As a result, spinal cord damage caused by chronic viral infection in the central nervous system was reduced in FVB mice that expressed 3D. This led to the preservation of large-diameter axons and motor function in these mice. The 3D transgene also lowered early viral loads when expressed in FVB-D(b) mice resistant to persistent TMEV infection. The protective effect of 3D transgenic expression was not altered in FVB-Rag(-/-).3D mice that are deficient in T and B cells, thus ruling out a mechanism by which the overexpression of 3D enhanced the adaptive immune clearance of the virus. Understanding how endogenously overexpressed 3D polymerase inhibits viral replication may lead to new strategies for targeting therapies to all picornaviruses.


Asunto(s)
Enfermedades Desmielinizantes/inmunología , ARN Polimerasa Dependiente del ARN/biosíntesis , Theilovirus/inmunología , Animales , Ratones , Ratones Noqueados , Ratones Transgénicos , Actividad Motora , ARN Polimerasa Dependiente del ARN/genética , Médula Espinal/patología , Carga Viral
11.
J Mol Neurosci ; 31(3): 297-305, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17726234

RESUMEN

In the mammalian brain, the hippocampus is involved in memory formation and storage and has an enriched level of Ca2+/calmodulin-dependent protein kinase type II (CaM kinase II). CaM kinase II has a number of downstream targets and is shown to play a role in memory development, axonal transport, and signaling across the synapse. The shiverer mutant mouse is a knockout lacking myelin basic protein. As a result, the axons of the central nervous system (CNS) of the shiverer have no or very thin myelin sheath, neurons in their CNS have distorted shapes, and synaptic signaling is impaired. shiverer mice develop symptoms similar to those experienced by patients with multiple sclerosis. In this study, proteins from the hippocampus, cerebellum, pons, medulla, and olfactory bulbs of shiverer and wild-type mice were extracted. Western blot analysis was used to compare the expression levels of CaM kinase II in these regions of the two types of mice. Analysis shows that at least two (50 and 58-59 kDa) of the four CaM kinase II isoforms are expressed in the brain, with one isoform (50 kDa) expressed in all regions examined. shiverer brain contains a decreased level of the two isoforms of CaM kinase II, an indication that the cognitive function of these mice might also be impaired.


Asunto(s)
Encéfalo/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Isoenzimas/metabolismo , Ratones Endogámicos , Animales , Encéfalo/anatomía & histología , Mediciones Luminiscentes , Ratones , Ratones Noqueados , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/metabolismo , Subunidades de Proteína/metabolismo
12.
J Neuropathol Exp Neurol ; 64(1): 46-57, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15715084

RESUMEN

For reasons that are not well understood, central nervous system repair in multiple sclerosis is often minimal. We present evidence, in a murine model of chronic progressive multiple sclerosis, that genetic factors can substantially influence remyelination, axonal integrity, and neurologic function. Four inbred mouse strains, SJL, B10.D1-H2(q), FVB, and SWR, developed extensive inflammatory demyelination by 3 months after infection with Theiler's murine encephalomyelitis virus. Demyelination continued lifelong in SJL and B10.D1-H2(q) mice, accompanied by axonal injury, minimal remyelination, and progressive motor dysfunction. In contrast, FVB and SWR mice showed less axonal injury, progressive remyelination, and stabilization of motor function. Genetic dominance of the reparative traits was demonstrated by crossing remyelinating strains (FVB and SWR) with nonremyelinating strains (SJL and B10.D1-H2(q)). All F1 mice developed a phenotype identical to FVB and SWR, showing extensive remyelination, partial preservation of axons, and preserved motor function. Analyses of viral RNA and antigen, immune cell infiltration, and antiviral antibody titers did not predict the phenotypic differences between strains. These results highlight the significant extent to which hereditary factors can control disease course and demonstrate that the switch from a pathogenic to a reparative phenotype can occur even after prolonged inflammatory demyelination.


Asunto(s)
Genes Dominantes , Esclerosis Múltiple Crónica Progresiva/genética , Esclerosis Múltiple Crónica Progresiva/fisiopatología , Médula Espinal/patología , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple Crónica Progresiva/virología , Vaina de Mielina/patología , Vaina de Mielina/virología , Fibras Nerviosas Mielínicas/patología , Fibras Nerviosas Mielínicas/virología , Especificidad de la Especie , Theilovirus/genética
13.
FASEB J ; 18(13): 1577-9, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15319372

RESUMEN

The human monoclonal antibody rHIgM22 enhances remyelination following spinal cord demyelination in a virus-induced murine model of multiple sclerosis. Using three-dimensional T2-weighted in vivo spinal cord magnetic resonance imaging (MRI), we have therefore assessed the extent of spinal cord demyelination, before and after 5 weeks of treatment with rHIgM22, to determine whether antibody enhanced remyelination can be detected by MRI. A significant decrease was seen in T2 high signal lesion volume following antibody treatment. Histologic examination of the spinal cord tissue reveals that this decrease in lesion volume correlates with antibody promoted remyelination. To show that rHIgM22 enters the spinal cord and colocalizes with demyelinating lesions, we used ultrasmall superparamagnetic iron oxide particle (USPIO)-labeled antibodies. This may be considered as additional evidence to the hypothesis that rHIgM22 promotes remyelination by local effects in the lesions, likely by binding to CNS cells. The reduction in high signal T2-weighted lesion volume may be an important outcome measure in future clinical trials in humans.


Asunto(s)
Anticuerpos/uso terapéutico , Modelos Animales de Enfermedad , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/patología , Vaina de Mielina/metabolismo , Médula Espinal/metabolismo , Médula Espinal/patología , Animales , Anticuerpos/administración & dosificación , Anticuerpos/farmacología , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Humanos , Imagen por Resonancia Magnética , Ratones , Esclerosis Múltiple/diagnóstico , Esclerosis Múltiple/virología , Médula Espinal/efectos de los fármacos , Factores de Tiempo
14.
FASEB J ; 16(10): 1325-7, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12154009

RESUMEN

Certain human sera from patients with monoclonal gammopathies contain factors that induce myelin repair in animals with demyelinating disease. We hypothesize that antibodies functionally distinguish the serum of one patient from another. However, pooled normal polyclonal human IgM antibodies also induce remyelination. Definitive proof that specific antibodies are the biologically active components of serum is missing because unquestionably pure preparations of antibody molecules cannot be generated by fractionation. To demonstrate definitively that antibody is the biologically active component of patient serum, recombinant antibody was generated for evaluation in bioassays. The induction of remyelination in vivo requires milligram quantities of antibody. Consequently, an expression system was engineered to express high-titer, recombinant human IgM antibodies in vitro. A resulting recombinant antibody (rHIgM22) was evaluated for its ability to induce remyelination in the Theiler's virus mouse model of chronic-progressive demyelinating disease. We demonstrate that a single recombinant monoclonal antibody recapitulates the key characteristics of patient serum, including binding specificity, the induction of calcium signals in oligodendrocytes in vitro, and the induction of myelin repair within demyelinated plaques in vivo. The rHIgM22 antibody provides a new venue for the analysis of mechanisms governing remyelination and may prove useful in the treatment of demyelinating diseases.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Enfermedades Autoinmunes Desmielinizantes SNC/terapia , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/farmacología , Astrocitos/metabolismo , Encéfalo/citología , Señalización del Calcio , Células Cultivadas , Enfermedad Crónica , Enfermedades Autoinmunes Desmielinizantes SNC/patología , Progresión de la Enfermedad , Técnicas de Transferencia de Gen , Humanos , Sueros Inmunes/inmunología , Sueros Inmunes/farmacología , Inmunoglobulina M/genética , Inmunoglobulina M/farmacología , Inmunoglobulina M/uso terapéutico , Ratones , Modelos Biológicos , Vaina de Mielina/patología , Oligodendroglía/inmunología , Oligodendroglía/metabolismo , Proteínas Recombinantes/farmacología
15.
J Neuropathol Exp Neurol ; 63(5): 461-73, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15198125

RESUMEN

Two human IgMs (sHIgM12 and sHIgM42) were identified that supported in vitro central nervous system (CNS) neurite extension equal to the potent neurite stimulatory molecule laminin. Both IgMs bound to multiple cell types in unfixed CNS tissue and to the surface of neurons in culture. Both monoclonal antibodies (mAbs) overrode the inhibitory effect of CNS mouse myelin on granule cell neurite extension. Neither mAb bound to the surface of mature oligodendrocytes or strictly colocalized with myelin proteins. Sialidase treatment eliminated the neuronal surface binding of both mAbs, whereas blocking sphingolipid synthesis with Fumonisin B1 or removing GPI-linked proteins with PIPLC did not. When used as substrates for mixed neuron/glia aggregates, sHIgM12 and sHIgM42 supported robust neurite extension while astrocytes remained in the aggregates. In contrast, laminin supported astrocyte migration and spreading. Human mAbs that support neurite extension are novel factors that may be of use in encouraging axon repair following injury while minimizing glial cell infiltration. Both human mAbs were isolated from individuals with monoclonal gammopathy. Each individual has carried high mAb titers in circulation for years without detriment. sHIgM12 and sHIgM42 are therefore unlikely to be systemically pathogenic.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Sistema Nervioso Central/crecimiento & desarrollo , Sistema Nervioso Central/inmunología , Inmunoglobulina M/metabolismo , Regeneración Nerviosa/inmunología , Neuritas/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Células Cultivadas , Sistema Nervioso Central/citología , Gliosis/inmunología , Gliosis/prevención & control , Conos de Crecimiento/efectos de los fármacos , Conos de Crecimiento/inmunología , Conos de Crecimiento/metabolismo , Humanos , Inmunoglobulina M/inmunología , Inmunoglobulina M/farmacología , Laminina/inmunología , Laminina/metabolismo , Laminina/farmacología , Ratones , Proteínas de la Mielina/metabolismo , Factores de Crecimiento Nervioso/inmunología , Factores de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/farmacología , Regeneración Nerviosa/efectos de los fármacos , Neuraminidasa/metabolismo , Neuritas/efectos de los fármacos , Neuritas/ultraestructura , Neuroglía/efectos de los fármacos , Neuroglía/inmunología , Neuroglía/metabolismo , Unión Proteica/inmunología , Ratas , Ratas Sprague-Dawley , Esfingolípidos/antagonistas & inhibidores , Esfingolípidos/metabolismo
16.
Brain Pathol ; 13(4): 608-16, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14655764

RESUMEN

The human monoclonal IgM antibody sHIgM22 and mouse IgM monoclonal antibody 94.03 bind to oligodendrocytes, induce calcium signals in cultured glial cells, and promote remyelination in mouse models of multiple sclerosis. In order to address the mechanisms employed by these antibodies to promote CNS repair, bivalent monomers, F(ab')2 fragments, and monovalent forms of these antibodies were investigated to determine whether they exhibit the same remyelinating potential as the intact IgMs. The two antibodies displayed different structural requirements for retention of function. Antibody sHIgM22 remained functional even when reduced to a bivalent F(ab')2 fragment, while disruption of the pentameric structure of antibody 94.03 destroyed its functional properties. Competition studies demonstrated that the two antibodies recognize different entities on the surface of glial cells. These results indicate that the constant region and pentameric structure of IgM is not always necessary for the stimulation of myelin repair, eliminating the requirement for IgM immune effector functions in this process. The ability of the antibodies to cross-link cell surface determinants on oligodendrocytes appears to be an essential aspect of the mechanism of cellular activation. The finding that two antibodies, which induce similar in vivo effects, bind to different structures, and have different cross-linking requirements suggests that activation of glial cells involves the rearrangement of a complex membrane compartment.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Enfermedades Desmielinizantes/terapia , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina M/uso terapéutico , Animales , Anticuerpos Monoclonales/inmunología , Calcio/metabolismo , Línea Celular , Sistema Nervioso Central/inmunología , Interacciones Farmacológicas , Electroforesis en Gel de Poliacrilamida/métodos , Femenino , Humanos , Hibridomas/metabolismo , Fragmentos Fab de Inmunoglobulinas/metabolismo , Inmunoglobulina G/uso terapéutico , Inmunoglobulina M/inmunología , Inmunohistoquímica , Técnicas In Vitro , Infecciones , Ratones , Ratones Endogámicos , Vaina de Mielina/inmunología , Regeneración Nerviosa/efectos de los fármacos , Oligodendroglía/metabolismo , Ratas , Médula Espinal/metabolismo , Médula Espinal/patología , Macroglobulinemia de Waldenström/inmunología , Cicatrización de Heridas
17.
J Neurosci Methods ; 227: 29-34, 2014 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-24486877

RESUMEN

BACKGROUND: Systemic delivery of pharmacologic agents has led to many significant advances in the treatment of neurologic and psychiatric conditions. However, this approach has several limitations, including difficulty penetrating the blood-brain barrier and enzymatic degradation prior to reaching its intended target. Here, we describe the testing of a system allowing intraparenchymal (IPa) infusion of therapeutic agents directly to the appropriate anatomical targets, in a swine model. NEW METHOD: Five male pigs underwent 3.0T magnetic resonance (MR) guided placement of an IPa catheter into the dorso-medial putamen, using a combined system of the Leksell stereotactic arc, a Mayo-developed MRI-compatible pig head frame, and a custom-designed Fred Haer Company (FHC) delivery system. RESULTS: Our results show hemi-lateral coverage of the pig putamen is achievable from a single infusion point and that the volume of the bolus detected in each animal is uniform (1544±420mm(3)). COMPARISON WITH EXISTING METHOD: The IPa infusion system is designed to isolate the intracranial catheter from bodily-induced forces while delivering drugs and molecules into the brain tissue by convection-enhanced delivery, with minimal-to-no catheter track backflow. CONCLUSION: This study presents an innovative IPa drug delivery system, which includes a sophisticated catheter and implantable pump designed to deliver drugs and various molecules in a precise and controlled manner with limited backflow. It also demonstrates the efficacy of the delivery system, which has the potential to radically impact the treatment of a wide range of neurologic conditions. Lastly, the swine model used here has certain advantages for translation into clinical applications.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Lateralidad Funcional , Bombas de Infusión Implantables , Animales , Convección , Sistemas de Liberación de Medicamentos/instrumentación , Gadolinio DTPA/metabolismo , Imagenología Tridimensional , Imagen por Resonancia Magnética , Masculino , Modelos Animales , Putamen/efectos de los fármacos , Putamen/fisiología , Porcinos , Factores de Tiempo
18.
Front Neurosci ; 8: 169, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25009455

RESUMEN

Current strategies for optimizing deep brain stimulation (DBS) therapy involve multiple postoperative visits. During each visit, stimulation parameters are adjusted until desired therapeutic effects are achieved and adverse effects are minimized. However, the efficacy of these therapeutic parameters may decline with time due at least in part to disease progression, interactions between the host environment and the electrode, and lead migration. As such, development of closed-loop control systems that can respond to changing neurochemical environments, tailoring DBS therapy to individual patients, is paramount for improving the therapeutic efficacy of DBS. Evidence obtained using electrophysiology and imaging techniques in both animals and humans suggests that DBS works by modulating neural network activity. Recently, animal studies have shown that stimulation-evoked changes in neurotransmitter release that mirror normal physiology are associated with the therapeutic benefits of DBS. Therefore, to fully understand the neurophysiology of DBS and optimize its efficacy, it may be necessary to look beyond conventional electrophysiological analyses and characterize the neurochemical effects of therapeutic and non-therapeutic stimulation. By combining electrochemical monitoring and mathematical modeling techniques, we can potentially replace the trial-and-error process used in clinical programming with deterministic approaches that help attain optimal and stable neurochemical profiles. In this manuscript, we summarize the current understanding of electrophysiological and electrochemical processing for control of neuromodulation therapies. Additionally, we describe a proof-of-principle closed-loop controller that characterizes DBS-evoked dopamine changes to adjust stimulation parameters in a rodent model of DBS. The work described herein represents the initial steps toward achieving a "smart" neuroprosthetic system for treatment of neurologic and psychiatric disorders.

19.
PLoS One ; 8(1): e54161, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23326593

RESUMEN

BACKGROUND: Chemotherapy-induced neuropathy is the principle dose limiting factor requiring discontinuation of many chemotherapeutic agents, including cisplatin and oxaliplatin. About 30 to 40% of patients receiving chemotherapy develop pain and sensory changes. Given that poly (ADP-ribose) polymerase (PARP) inhibition has been shown to provide neuroprotection, the current study was developed to test whether the novel PARP inhibitor compound 4a (analog of ABT-888) would attenuate pain in cisplatin and oxaliplatin-induced neuropathy in mice. RESULTS: An established chemotherapy-induced painful neuropathy model of two weekly cycles of 10 intraperitoneal (i.p.) injections separated by 5 days rest was used to examine the therapeutic potential of the PARP inhibitor compound 4a. Behavioral testing using von Frey, paw radiant heat, cold plate, and exploratory behaviors were taken at baseline, and followed by testing at 3, 6, and 8 weeks from the beginning of drug treatment. CONCLUSION: Cisplatin-treated mice developed heat hyperalgesia and mechanical allodynia while oxaliplatin-treated mice exhibited cold hyperalgesia and mechanical allodynia. Co-administration of 50 mg/kg or 25 mg/kg compound 4a with platinum regimen, attenuated cisplatin-induced heat hyperalgesia and mechanical allodynia in a dose dependent manner. Similarly, co-administration of 50 mg/kg compound 4a attenuated oxaliplatin-induced cold hyperalgesia and mechanical allodynia. These data indicate that administration of a novel PARP inhibitor may have important applications as a therapeutic agent for human chemotherapy-induced painful neuropathy.


Asunto(s)
Hiperalgesia/inducido químicamente , Neuralgia/tratamiento farmacológico , Fármacos Neuroprotectores/administración & dosificación , Poli(ADP-Ribosa) Polimerasas , Animales , Antineoplásicos/toxicidad , Bencimidazoles/administración & dosificación , Bencimidazoles/síntesis química , Cisplatino/toxicidad , Humanos , Hiperalgesia/tratamiento farmacológico , Masculino , Ratones , Neuralgia/inducido químicamente , Neuralgia/metabolismo , Compuestos Organoplatinos/toxicidad , Oxaliplatino , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/metabolismo
20.
PLoS One ; 8(12): e81443, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24339929

RESUMEN

Restoration of movement following spinal cord injury (SCI) has been achieved using electrical stimulation of peripheral nerves and skeletal muscles. However, practical limitations such as the rapid onset of muscle fatigue hinder clinical application of these technologies. Recently, direct stimulation of alpha motor neurons has shown promise for evoking graded, controlled, and sustained muscle contractions in rodent and feline animal models while overcoming some of these limitations. However, small animal models are not optimal for the development of clinical spinal stimulation techniques for functional restoration of movement. Furthermore, variance in surgical procedure, targeting, and electrode implantation techniques can compromise therapeutic outcomes and impede comparison of results across studies. Herein, we present a protocol and large animal model that allow standardized development, testing, and optimization of novel clinical strategies for restoring motor function following spinal cord injury. We tested this protocol using both epidural and intraspinal stimulation in a porcine model of spinal cord injury, but the protocol is suitable for the development of other novel therapeutic strategies. This protocol will help characterize spinal circuits vital for selective activation of motor neuron pools. In turn, this will expedite the development and validation of high-precision therapeutic targeting strategies and stimulation technologies for optimal restoration of motor function in humans.


Asunto(s)
Terapia por Estimulación Eléctrica/métodos , Recuperación de la Función , Traumatismos de la Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/terapia , Médula Espinal/fisiopatología , Animales , Interfaces Cerebro-Computador , Modelos Animales de Enfermedad , Espacio Epidural , Femenino , Calidad de Vida , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA