Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Neurosci Res ; 91(10): 1292-302, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23907992

RESUMEN

We previously showed that, in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease (PD), vaccination with bacillus Calmette-Guerin (BCG) prior to MPTP exposure limited the loss of striatal dopamine (DA) and dopamine transporter (DAT) and prevented the activation of nigral microglia. Here, we conducted BCG dose studies and investigated the mechanisms underlying BCG vaccination's neuroprotective effects in this model. We found that a dose of 1 × 10(6) cfu BCG led to higher levels of striatal DA and DAT ligand binding (28% and 42%, respectively) in BCG-vaccinated vs. unvaccinated MPTP-treated mice, but without a significant increase in substantia nigra tyrosine hydroxylase-staining neurons. Previous studies showed that BCG can induce regulatory T cells (Tregs) and that Tregs are neuroprotective in models of neurodegenerative diseases. However, MPTP is lymphotoxic, so it was unclear whether Tregs were maintained after MPTP treatment and whether a relationship existed between Tregs and the preservation of striatal DA system integrity. We found that, 21 days post-MPTP treatment, Treg levels in mice that had received BCG prior to MPTP were threefold greater than those in MPTP-only-treated mice and elevated above those in saline-only-treated mice, suggesting that the persistent BCG infection continually promoted Treg responses. Notably, the magnitude of the Treg response correlated positively with both striatal DA levels and DAT ligand binding. Therefore, BCG vaccine-mediated neuroprotection is associated with Treg levels in this mouse model. Our results suggest that BCG-induced Tregs could provide a new adjunctive therapeutic approach to ameliorating pathology associated with PD and other neurodegenerative diseases.


Asunto(s)
Vacuna BCG/inmunología , Fármacos Neuroprotectores/inmunología , Trastornos Parkinsonianos/inmunología , Trastornos Parkinsonianos/prevención & control , Linfocitos T Reguladores/inmunología , Animales , Vacuna BCG/administración & dosificación , Encéfalo/inmunología , Encéfalo/patología , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Citometría de Flujo , Masculino , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/administración & dosificación
2.
J Immunol ; 184(2): 816-23, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20018625

RESUMEN

Studies of mice lacking MHC class I (MHC I)-associated proteins have demonstrated a role for MHC I in neurodevelopment. A central question arising from these observations is whether neuronal recognition of MHC I has specificity for the MHC I allele product and the peptide presented. Using a well-established embryonic retina explant system, we observed that picomolar levels of a recombinant self-MHC I molecule inhibited neurite outgrowth. We then assessed the neurobiological activity of a panel of recombinant soluble MHC Is, consisting of different MHC I heavy chains with a defined self- or nonself-peptide presented, on cultured embryonic retinas from mice with different MHC I haplotypes. We observed that self-MHC I allele products had greater inhibitory neuroactivity than nonself-MHC I molecules, regardless of the nature of the peptide presented, a pattern akin to MHC I recognition by some innate immune system receptors. However, self-MHC I molecules had no effect on retinas from MHC I-deficient mice. These observations suggest that neuronal recognition of MHC I may be coordinated with the inherited MHC I alleles, as occurs in the innate immune system. Consistent with this notion, we show that MHC I and MHC I receptors are coexpressed by precursor cells at the earliest stages of retina development, which could enable such coordination.


Asunto(s)
Alelos , Presentación de Antígeno , Antígenos de Histocompatibilidad Clase I/genética , Neuronas/inmunología , Péptidos/inmunología , Animales , Autoantígenos/inmunología , Células Madre Embrionarias , Inmunidad Innata , Ratones , Neuritas/inmunología , Neuronas/citología , Retina/embriología
3.
Biomedicines ; 10(1)2022 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-35052808

RESUMEN

Sjögren's syndrome (SS) is a chronic autoimmune disease characterized by lymphocytic infiltrates in the salivary and lachrymal glands resulting in oral and ocular dryness. There are no clinically approved therapies to slow the progression of SS. Immune cells possess receptors for the neurotransmitter GABA (GABA-Rs) and their activation has immunoregulatory actions. We tested whether GABA administration has potential for amelioration of SS in NOD.B10-H2b and C57BL/6.NOD-Aec1Aec2 mice, two spontaneous SS models. Oral GABA treatment was initiated (1) after the development of sialadenitis but before the onset of overt symptoms, or (2) after the appearance of overt symptoms. When assessed weeks later, GABA-treated mice had greater saliva and tear production, as well as quicker times to salvia flow, in both SS mouse models. This was especially evident when GABA treatment was initiated after the onset of overt disease. This preservation of exocrine function was not accompanied by significant changes in the number or area of lymphocytic foci in the salivary or lachrymal glands of GABA-treated mice and we discuss the possible reasons for these observations. Given that GABA-treatment preserved saliva and tear production which are the most salient symptoms of SS and is safe for consumption, it may provide a new approach to help ameliorate SS.

4.
J Neurosci Res ; 89(3): 365-72, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21259323

RESUMEN

Mice that are deficient in classical major histocompatibility complex class I (MHCI) have abnormalities in synaptic plasticity and neurodevelopment and have more extensive loss of synapses and reduced axon regeneration after sciatic nerve transection, suggesting that MHCI participates in maintaining synapses and axon regeneration. Little is known about the biological consequences of up-regulating MHCI's expression on neurons. To understand MHCI's neurobiological activity better, and in particular its role in neurorepair after injury, we have studied neurorepair in a transgenic mouse model in which classical MHCI expression is up-regulated only on neurons. Using a well-established spinal cord injury (SCI) model, we observed that transgenic mice with elevated neuronal MHCI expression had significantly better recovery of locomotor abilities after SCI than wild-type mice. Although previous studies have implicated inflammation as both deleterious and beneficial for recovery after SCI, our results point directly to enhanced neuronal MHCI expression as a beneficial factor for promoting recovery of locomotor function after SCI.


Asunto(s)
Regulación de la Expresión Génica/genética , Antígenos de Histocompatibilidad Clase I/genética , Locomoción/fisiología , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Prueba de Esfuerzo/métodos , Lateralidad Funcional , Locomoción/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Fosfopiruvato Hidratasa/genética , Recuperación de la Función/genética , Traumatismos de la Médula Espinal/patología
5.
Viruses ; 13(6)2021 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-34071034

RESUMEN

There is an urgent need for new approaches to limit the severity of coronavirus infections. Many cells of the immune system express receptors for the neurotransmitter γ-aminobutyric acid (GABA), and GABA-receptor (GABA-R) agonists have anti-inflammatory effects. Lung epithelial cells also express GABA-Rs, and GABA-R modulators have been shown to limit acute lung injuries. There is currently, however, no information on whether GABA-R agonists might impact the course of a viral infection. Here, we assessed whether clinically applicable GABA-R agonists could be repurposed for the treatment of a lethal coronavirus (murine hepatitis virus 1, MHV-1) infection in mice. We found that oral GABA administration before, or after the appearance of symptoms, very effectively limited MHV-1-induced pneumonitis, severe illness, and death. GABA treatment also reduced viral load in the lungs, suggesting that GABA-Rs may provide a new druggable target to limit coronavirus replication. Treatment with the GABAA-R-specific agonist homotaurine, but not the GABAB-R-specific agonist baclofen, significantly reduced the severity of pneumonitis and death rates in MHV-1-infected mice, indicating that the therapeutic effects were mediated primarily through GABAA-Rs. Since GABA and homotaurine are safe for human consumption, they are promising candidates to help treat coronavirus infections.


Asunto(s)
Infecciones por Coronavirus/tratamiento farmacológico , Agonistas de Receptores de GABA-A/uso terapéutico , Virus de la Hepatitis Murina/efectos de los fármacos , Neumonía/tratamiento farmacológico , Animales , Infecciones por Coronavirus/mortalidad , Infecciones por Coronavirus/virología , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/virología , Ratones , Virus de la Hepatitis Murina/patogenicidad , Neumonía/mortalidad , Neumonía/virología , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Carga Viral/efectos de los fármacos , Pérdida de Peso/efectos de los fármacos , Ácido gamma-Aminobutírico/uso terapéutico
6.
Biomedicines ; 9(1)2021 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-33418884

RESUMEN

Some immune system cells express type A and/or type B γ-aminobutyric acid receptors (GABAA-Rs and/or GABAB-Rs). Treatment with GABA, which activates both GABAA-Rs and GABAB-Rs), and/or a GABAA-R-specific agonist inhibits disease progression in mouse models of type 1 diabetes (T1D), multiple sclerosis, rheumatoid arthritis, and COVID-19. Little is known about the clinical potential of specifically modulating GABAB-Rs. Here, we tested lesogaberan, a peripherally restricted GABAB-R agonist, as an interventive therapy in diabetic NOD mice. Lesogaberan treatment temporarily restored normoglycemia in most newly diabetic NOD mice. Combined treatment with a suboptimal dose of lesogaberan and proinsulin/alum immunization in newly diabetic NOD mice or a low-dose anti-CD3 in severely hyperglycemic NOD mice greatly increased T1D remission rates relative to each monotherapy. Mice receiving combined lesogaberan and anti-CD3 displayed improved glucose tolerance and, unlike mice that received anti-CD3 alone, had some islets with many insulin+ cells, suggesting that lesogaberan helped to rapidly inhibit ß-cell destruction. Hence, GABAB-R-specific agonists may provide adjunct therapies for T1D. Finally, the analysis of microarray and RNA-Seq databases suggested that the expression of GABAB-Rs and GABAA-Rs, as well as GABA production/secretion-related genes, may be a more common feature of immune cells than currently recognized.

7.
bioRxiv ; 2020 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-33024975

RESUMEN

There is an urgent need for new treatments to prevent and ameliorate severe illness and death induced by SARS-CoV-2 infection in COVID-19 patients. The coronavirus mouse hepatitis virus (MHV)-1 causes pneumonitis in mice which shares many pathological characteristics with human SARS-CoV infection. Previous studies have shown that the amino acid gamma-aminobutyric acid (GABA) has anti-inflammatory effects. We tested whether oral treatment with GABA could modulate the MHV-1 induced pneumonitis in susceptible A/J mice. As expected, MHV-1-inoculated control mice became severely ill (as measured by weight loss, clinical score, and the ratio of lung weight to body weight) and >60% of them succumbed to the infection. In contrast, mice that received GABA immediately after MHV-1 inoculation became only mildly ill and all of them recovered. When GABA treatment was initiated after the appearance of illness (3 days post-MHV-1 infection), we again observed that GABA treatment significantly reduced the severity of illness and greatly increased the frequency of recovery. Therefore, the engagement of GABA receptors (GABA-Rs) prevented the MHV-1 infection-induced severe pneumonitis and death in mice. Given that GABA-R agonists, like GABA and homotaurine, are safe for human consumption, stable, inexpensive, and available worldwide, they are promising candidates to help prevent severe illness stemming from SARS-CoV-2 infection and other coronavirus strains.

8.
Sci Rep ; 7(1): 374, 2017 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-28336918

RESUMEN

A key goal of diabetes research is to develop treatments to safely promote human ß-cell replication. It has recently become appreciated that activation of γ-aminobutyric acid receptors (GABA-Rs) on ß-cells can promote their survival and replication. A number of positive allosteric modulators (PAMs) that enhance GABA's actions on neuronal GABAA-Rs are in clinical use. Repurposing these GABAA-R PAMs to help treat diabetes is theoretically appealing because of their safety and potential to enhance the ability of GABA, secreted from ß-cells, or exogenously administered, to promote ß-cell replication and survival. Here, we show that clinically applicable GABAA-R PAMs can increase significantly INS-1 ß-cell replication, which is enhanced by exogenous GABA application. Furthermore, a GABAA-R PAM promoted human islet cell replication in vitro. This effect was abrogated by a GABAA-R antagonist. The combination of a PAM and low levels of exogenous GABA further increased human islet cell replication. These findings suggest that PAMs may potentiate the actions of GABA secreted by islet ß-cells on GABAA-Rs and provide a new class of drugs for diabetes treatment. Finally, our findings may explain a past clinical observation of a GABAA-R PAM reducing HbA1c levels in diabetic patients.


Asunto(s)
Benzodiazepinas/farmacología , División Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Receptores de GABA-A/fisiología , Ácido gamma-Aminobutírico/biosíntesis , Regulación Alostérica , Alprazolam/farmacología , Animales , Proteínas Portadoras/metabolismo , Clonazepam/farmacología , Moduladores del GABA/farmacología , Antagonistas de Receptores de GABA-A/administración & dosificación , Humanos , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/fisiología , Ratones , Midazolam/farmacología , Ratas , Receptores de GABA-A/metabolismo , Ácido gamma-Aminobutírico/administración & dosificación
9.
J Neuroimmunol ; 247(1-2): 1-8, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22503373

RESUMEN

We studied cultured hippocampal neurons from embryonic wildtype, major histocompatibility complex class I (MHCI) heavy chain-deficient (K(b)D(b)-/-) and NSE-D(b) (which have elevated neuronal MHCI expression) C57BL/6 mice. K(b)D(b)-/- neurons displayed slower neuritogenesis and establishment of polarity, while NSE-D(b) neurons had faster neurite outgrowth, more primary neurites, and tended to have accelerated polarization. Additional studies with ß2M-/- neurons, exogenous ß2M, and a self-MHCI monomer suggest that free heavy chain cis interactions with other surface molecules can promote neuritogenesis while tripartite MHCI interactions with classical MHCI receptors can inhibit axon outgrowth. Together with the results of others, MHCI appears to differentially modulate neuritogenesis and synaptogenesis.


Asunto(s)
Polaridad Celular/fisiología , Antígenos de Histocompatibilidad Clase I/metabolismo , Neurogénesis/fisiología , Neuronas/fisiología , Actinas/metabolismo , Análisis de Varianza , Animales , Axones/fisiología , Polaridad Celular/efectos de los fármacos , Células Cultivadas , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/fisiología , Hipocampo/citología , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Neuritas/efectos de los fármacos , Neuritas/fisiología , Neuronas/efectos de los fármacos , Fosfopiruvato Hidratasa/genética , Proteínas Recombinantes/farmacología , Factores de Tiempo , Tubulina (Proteína)/metabolismo , Microglobulina beta-2/deficiencia
10.
PLoS One ; 6(1): e16610, 2011 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-21304945

RESUMEN

There is a growing interest in using vaccination with CNS antigens to induce autoreactive T cell responses that home to damaged areas in the CNS and ameliorate neurodegenerative disease. Neuroprotective vaccine studies have focused on administering oligodendrocyte antigens or Copaxone® in complete Freund's adjuvant (CFA). Theoretical considerations, however, suggest that vaccination with a neuronal antigen may induce more robust neuroprotective immune responses. We assessed the neuroprotective potential of vaccines containing tyrosine hydroxylase (a neuronal protein involved in dopamine synthesis) or Copaxone® in CFA in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease. Surprisingly, we observed that the main beneficial factor in these vaccines was the CFA. Since the major immunogenic component in CFA is Mycobacterium tuberculosis, which closely related to the bacille Calmette-Guérin (BCG) that is used in human vaccines, we tested BCG vaccination in the MPTP mouse model. We observed that BCG vaccination partially preserved markers of striatal dopamine system integrity and prevented an increase in activated microglia in the substantia nigra of MPTP-treated mice. These results support a new neuroprotective vaccine paradigm in which general (nonself-reactive) immune stimulation in the periphery can limit potentially deleterious microglial responses to a neuronal insult and exert a neurorestorative effect in the CNS. Accordingly, BCG vaccination may provide a new strategy to augment current treatments for a wide range of neuropathological conditions.


Asunto(s)
Vacuna BCG/farmacología , Fármacos Neuroprotectores , Enfermedad de Parkinson/terapia , Animales , Vacuna BCG/uso terapéutico , Cuerpo Estriado , Modelos Animales de Enfermedad , Dopamina , Ratones , Sustancia Negra , Tirosina 3-Monooxigenasa/uso terapéutico , Vacunación
11.
PLoS One ; 6(3): e18439, 2011 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-21483793

RESUMEN

The neurobiological activities of classical major histocompatibility class I (MHCI) molecules are just beginning to be explored. To further examine MHCI's actions during the formation of neuronal connections, we cultured embryonic mouse retina explants a short distance from wildtype thalamic explants, or thalami from transgenic mice (termed "NSE-Db") whose neurons express higher levels of MHCI. While retina neurites extended to form connections with wildtype thalami, we were surprised to find that retina neurite outgrowth was very stunted in regions proximal to NSE-Db thalamic explants, suggesting that a diffusible factor from these thalami inhibited retina neurite outgrowth. It has been long known that MHCI-expressing cells release soluble forms of MHCI (sMHCI) due to the shedding of intact MHCI molecules, as well as the alternative exon splicing of its heavy chain or the action proteases which cleave off it's transmembrane anchor. We show that the diffusible inhibitory factor from the NSE-Db thalami is sMHCI. We also show that COS cells programmed to express murine MHCI release sMHCI that inhibits neurite outgrowth from nearby neurons in vitro. The neuroinhibitory effect of sMHCI could be blocked by lowering cAMP levels, suggesting that the neuronal MHCI receptor's signaling mechanism involves a cyclic nucleotide-dependent pathway. Our results suggest that MHCI may not only have neurobiological activity in its membrane-bound form, it may also influence local neurons as a soluble molecule. We discuss the involvement of complement proteins in generating sMHCI and new theoretical models of MHCI's biological activities in the nervous system.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Neuritas/metabolismo , Neuritas/fisiología , Animales , Células COS , Chlorocebus aethiops , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Ratones , Ratones Endogámicos C57BL , Retina/citología , Retina/embriología , Retina/metabolismo , Tálamo/citología , Tálamo/embriología , Tálamo/metabolismo
12.
Diabetes ; 60(5): 1383-92, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21441442

RESUMEN

OBJECTIVE: ß-Cells that express an imaging reporter have provided powerful tools for studying ß-cell development, islet transplantation, and ß-cell autoimmunity. To further expedite diabetes research, we generated transgenic C57BL/6 "MIP-TF" mice that have a mouse insulin promoter (MIP) driving the expression of a trifusion (TF) protein of three imaging reporters (luciferase/enhanced green fluorescent protein/HSV1-sr39 thymidine kinase) in their ß-cells. This should enable the noninvasive imaging of ß-cells by charge-coupled device (CCD) and micro-positron emission tomography (PET), as well as the identification of ß-cells at the cellular level by fluorescent microscopy. RESEARCH DESIGN AND METHODS: MIP-TF mouse ß-cells were multimodality imaged in models of type 1 and type 2 diabetes. RESULTS: MIP-TF mouse ß-cells were readily identified in pancreatic tissue sections using fluorescent microscopy. We show that MIP-TF ß-cells can be noninvasively imaged using microPET. There was a correlation between CCD and microPET signals from the pancreas region of individual mice. After low-dose streptozotocin administration to induce type 1 diabetes, we observed a progressive reduction in bioluminescence from the pancreas region before the appearance of hyperglycemia. Although there have been reports of hyperglycemia inducing proinsulin expression in extrapancreatic tissues, we did not observe bioluminescent signals from extrapancreatic tissues of diabetic MIP-TF mice. Because MIP-TF mouse ß-cells express a viral thymidine kinase, ganciclovir treatment induced hyperglycemia, providing a new experimental model of type 1 diabetes. Mice fed a high-fat diet to model early type 2 diabetes displayed a progressive increase in their pancreatic bioluminescent signals, which were positively correlated with area under the curve-intraperitoneal glucose tolerance test (AUC-IPGTT). CONCLUSIONS: MIP-TF mice provide a new tool for monitoring ß-cells from the single cell level to noninvasive assessments of ß-cells in models of type 1 diabetes and type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Animales , Citometría de Flujo , Prueba de Tolerancia a la Glucosa , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Insulina/genética , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Tomografía de Emisión de Positrones , Regiones Promotoras Genéticas/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Timidina Quinasa/genética , Timidina Quinasa/metabolismo
13.
Mol Ther ; 14(6): 851-6, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16982215

RESUMEN

Islet transplantation can restore glucose homeostasis in those with type 1 diabetes; however, most recipients eventually lose graft function. A noninvasive method to monitor islets following transplantation would enable assessment of their survival and aid the development of therapeutics to prolong graft survival. Here, we show that recombinant lentivirus can be used to engineer human islets to express a positron emission tomography (PET) reporter gene. Following transplantation into mice, transduced islets could be imaged in vivo using microPET and a radiolabeled probe approved by the FDA for clinical use in humans. The magnitude of signal from engineered islets implanted into the axillary cavity reflected the implanted islet mass. Signals from implanted islets decreased by approximately one-half during the first few weeks following transplantation, which may reflect islet cell death shortly after transplantation. Thereafter, the magnitude of signals from the implanted islets remained fairly constant when the recipients were repetitively reimaged over 90 days. Histological analysis of the implants showed healthy islets with PET reporter-expressing cells distributed throughout the islet architecture. These studies suggest that PET imaging of lentivirus-transduced islets could provide a safe and feasible method for long-term monitoring of islet graft survival.


Asunto(s)
Trasplante de Islotes Pancreáticos/diagnóstico por imagen , Islotes Pancreáticos/metabolismo , Tomografía de Emisión de Positrones/métodos , Animales , Células Cultivadas , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 1/terapia , Vectores Genéticos/genética , Supervivencia de Injerto/genética , Supervivencia de Injerto/fisiología , Herpesvirus Humano 1/enzimología , Humanos , Islotes Pancreáticos/citología , Islotes Pancreáticos/diagnóstico por imagen , Trasplante de Islotes Pancreáticos/métodos , Lentivirus/genética , Masculino , Ratones , Ratones Endogámicos NOD , Mutación/genética , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Factores de Tiempo , Transfección , Trasplante Heterólogo
14.
Proc Natl Acad Sci U S A ; 103(30): 11294-9, 2006 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-16868090

RESUMEN

Islet transplantation offers a potential therapy to restore glucose homeostasis in type 1 diabetes patients. However, islet transplantation is not routinely successful because most islet recipients gradually lose graft function. Furthermore, serological markers of islet function are insensitive to islet loss until the latter stages of islet graft rejection. A noninvasive method of monitoring islet grafts would aid in the assessment of islet graft survival and the evaluation of interventions designed to prolong graft survival. Here, we show that recombinant adenovirus can engineer isolated islets to express a positron-emission tomography (PET) reporter gene and that these islets can be repeatedly imaged by using microPET after transplantation into mice. The magnitude of signal from engineered islets implanted into the axillary cavity was directly related to the implanted islet mass. PET signals attenuated over the following weeks because of the transient nature of adenovirus-mediated gene expression. Because the liver is the preferred site for islet implantation in humans, we also tested whether islets could be imaged after transfusion into the mouse liver. Control studies revealed that both intrahepatic islet transplantation and hyperglycemia altered the biodistribution kinetics of the PET probe systemically. Although transplanted islets were dispersed throughout the liver, clear signals from the liver region of mice receiving PET reporter-expressing islets were detectable for several weeks. Viral transduction, PET reporter expression, and repeated microPET imaging had no apparent deleterious effects on islet function after implantation. These studies lay a foundation for noninvasive quantitative assessments of islet graft survival using PET.


Asunto(s)
Trasplante de Islotes Pancreáticos/métodos , Tomografía de Emisión de Positrones/métodos , Animales , Supervivencia Celular , Genes Reporteros , Supervivencia de Injerto , Humanos , Hígado/patología , Ratones , Ratas , Transducción de Señal
15.
J Immunol ; 175(3): 1991-9, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16034144

RESUMEN

As organ-specific autoimmune diseases do not become manifest until well-advanced, interventive therapies must inhibit late-stage disease processes. Using a panel of immunogenic peptides from various beta cell Ags, we evaluated the factors influencing the efficacy of Ag-based therapies in diabetes-prone NOD mice with advanced disease. The ability of the major beta cell autoantigen target determinants (TDs) to prime Th2 responses declined sharply between 6 and 12 wk of age, whereas the ability of immunogenic ignored determinants (IDs) of beta cell Ags to prime Th2 responses was unaffected by the disease process. The different patterns of TD and ID immunogenicity (even from the same beta cell Ag) may be due to the exhaustion of uncommitted TD-reactive, but not ID-reactive, T cell pools by recruitment into the autoimmune cascade. Therapeutic efficacy was associated with a peptide's immunogenicity and ability to promote Th2 spreading late in the disease process but not its affinity for I-Ag7 or its expression pattern (beta cell specific/nonspecific or rare/abundant). Characterization of some IDs revealed them to be "absolute" cryptic determinants. Such determinants have little impact on T cell selection, leaving large precursor T cell pools available for priming by synthetic peptides. Traditional Ag-based therapeutics using whole autoantigens or their TDs cannot prime responses to such determinants. These findings suggest a new strategy for designing more efficacious Ag-based therapeutics for late-stage autoimmune diseases.


Asunto(s)
Autoantígenos/uso terapéutico , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Epítopos/uso terapéutico , Predisposición Genética a la Enfermedad , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/metabolismo , Secuencia de Aminoácidos , Animales , Autoantígenos/administración & dosificación , Autoantígenos/inmunología , Diabetes Mellitus Tipo 1/genética , Progresión de la Enfermedad , Esquema de Medicación , Epítopos/administración & dosificación , Epítopos/inmunología , Femenino , Glutamato Descarboxilasa/administración & dosificación , Glutamato Descarboxilasa/inmunología , Glutamato Descarboxilasa/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Isoenzimas/administración & dosificación , Isoenzimas/inmunología , Isoenzimas/metabolismo , Ratones , Ratones Endogámicos NOD , Datos de Secuencia Molecular , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Péptidos/administración & dosificación , Péptidos/inmunología , Péptidos/metabolismo , Unión Proteica/inmunología , Células TH1/inmunología , Células TH1/metabolismo , Células Th2/inmunología , Células Th2/metabolismo
16.
Mol Ther ; 9(3): 428-35, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15006610

RESUMEN

Islet transplantation offers a potential therapy to restore glucose homeostasis in type 1 diabetes patients. A method to image transplanted islets noninvasively and repeatedly would greatly assist studies of islet transplantation. Using recombinant adenovirus, we show that isolated rodent and human islets can be genetically engineered to express luciferase and then imaged after implantation into NOD-scid mice using a cooled charge-coupled device. The magnitude of the signal was dependent on the islet dose. Adenovirus-directed luciferase expression, however, rapidly attenuated. We next tested lentivirus vectors that should direct the long-term expression of reporter genes in transduced islets. Transplanted lentivirus-transduced islets restored euglycemia long term in streptozotocin-treated NOD-scid mice. The signal from implanted lentivirus-transduced islets was related directly to the implanted islet mass, and the signal did not attenuate over the observation period. Viral transduction, luciferase expression, and repeated imaging had no apparent long-term deleterious effects on islet function after implantation. These data demonstrate that the introduction of reporter genes into an isolated tissue allows the long-term monitoring of its survival following implantation. Such imaging technologies may allow earlier detection of graft rejection and the adjustment of therapies to prolong graft survival posttransplantation.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética/métodos , Supervivencia de Injerto , Trasplante de Islotes Pancreáticos/métodos , Adenoviridae/genética , Animales , Diabetes Mellitus Tipo 1/terapia , Genes Reporteros , Vectores Genéticos , Humanos , Inmunohistoquímica , Islotes Pancreáticos/metabolismo , Lentivirus/genética , Luciferasas/genética , Luciferasas/metabolismo , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratas , Estreptozocina/farmacología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA