Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
J Neuroinflammation ; 20(1): 7, 2023 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-36611185

RESUMEN

BACKGROUND: Promotion of myelin repair in the context of demyelinating diseases such as multiple sclerosis (MS) still represents a clinical unmet need, given that this disease is not only characterized by autoimmune activities but also by impaired regeneration processes. Hence, this relates to replacement of lost oligodendrocytes and myelin sheaths-the primary targets of autoimmune attacks. Endogenous remyelination is mainly mediated via activation and differentiation of resident oligodendroglial precursor cells (OPCs), whereas its efficiency remains limited and declines with disease progression and aging. Teriflunomide has been approved as a first-line treatment for relapsing remitting MS. Beyond its role in acting via inhibition of de novo pyrimidine synthesis leading to a cytostatic effect on proliferating lymphocyte subsets, this study aims to uncover its potential to foster myelin repair. METHODS: Within the cuprizone mediated de-/remyelination model teriflunomide dependent effects on oligodendroglial homeostasis and maturation, related to cellular processes important for myelin repair were analyzed in vivo. Teriflunomide administration was performed either as pulse or continuously and markers specific for oligodendroglial maturation and mitochondrial integrity were examined by means of gene expression and immunohistochemical analyses. In addition, axon myelination was determined using electron microscopy. RESULTS: Both pulse and constant teriflunomide treatment efficiently boosted myelin repair activities in this model, leading to accelerated generation of oligodendrocytes and restoration of myelin sheaths. Moreover, teriflunomide restored mitochondrial integrity within oligodendroglial cells. CONCLUSIONS: The link between de novo pyrimidine synthesis inhibition, oligodendroglial rescue, and maintenance of mitochondrial homeostasis appears as a key for successful myelin repair and hence for protection of axons from degeneration.


Asunto(s)
Vaina de Mielina , Oligodendroglía , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Crotonatos/farmacología , Crotonatos/uso terapéutico , Hidroxibutiratos/metabolismo , Hidroxibutiratos/farmacología , Diferenciación Celular
2.
Glia ; 70(6): 1100-1116, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35188681

RESUMEN

We have previously shown that targeting endoneurial macrophages with the orally applied CSF-1 receptor specific kinase (c-FMS) inhibitor PLX5622 from the age of 3 months onwards led to a substantial alleviation of the neuropathy in mouse models of Charcot-Marie-Tooth (CMT) 1X and 1B disease, which are genetically-mediated nerve disorders not treatable in humans. The same approach failed in a model of CMT1A (PMP22-overexpressing mice, line C61), representing the most frequent form of CMT. This was unexpected since previous studies identified macrophages contributing to disease severity in the same CMT1A model. Here we re-approached the possibility of alleviating the neuropathy in a model of CMT1A by targeting macrophages at earlier time points. As a proof-of-principle experiment, we genetically inactivated colony-stimulating factor-1 (CSF-1) in CMT1A mice, which resulted in lower endoneurial macrophage numbers and alleviated the neuropathy. Based on these observations, we pharmacologically ablated macrophages in newborn CMT1A mice by feeding their lactating mothers with chow containing PLX5622, followed by treatment of the respective progenies after weaning until the age of 6 months. We found that peripheral neuropathy was substantially alleviated after early postnatal treatment, leading to preserved motor function in CMT1A mice. Moreover, macrophage depletion affected the altered Schwann cell differentiation phenotype. These findings underscore the targetable role of macrophage-mediated inflammation in peripheral nerves of inherited neuropathies, but also emphasize the need for an early treatment start confined to a narrow therapeutic time window in CMT1A models and potentially in respective patients.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth , Lactancia , Animales , Diferenciación Celular , Enfermedad de Charcot-Marie-Tooth/genética , Femenino , Humanos , Macrófagos/metabolismo , Ratones , Nervios Periféricos/metabolismo
3.
Glia ; 68(2): 393-406, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31633850

RESUMEN

Apart from dedicated oligodendroglial progenitor cells, adult neural stem cells (aNSCs) can also give rise to new oligodendrocytes in the adult central nervous system (CNS). This process mainly confers myelinating glial cell replacement in pathological situations and can hence contribute to glial heterogeneity. Our previous studies demonstrated that the p57kip2 gene encodes an intrinsic regulator of glial fate acquisition and we here investigated to what degree its modulation can affect stem cell-dependent oligodendrogenesis in different CNS environments. We therefore transplanted p57kip2 knockdown aNSCs into white and gray matter (WM and GM) regions of the mouse brain, into uninjured spinal cords as well as in the vicinity of spinal cord injuries and evaluated integration and differentiation in vivo. Our experiments revealed that under healthy conditions intrinsic suppression of p57kip2 as well as WM localization promote differentiation toward myelinating oligodendrocytes at the expense of astrocyte generation. Moreover, p57kip2 knockdown conferred a strong benefit on cell survival augmenting net oligodendrocyte generation. In the vicinity of hemisectioned spinal cords, the gene knockdown led to a similar induction of oligodendroglial features; however, newly generated oligodendrocytes appeared to suffer more from the hostile environment. This study contributes to our understanding of mechanisms of adult oligodendrogenesis and glial heterogeneity and further reveals critical factors when considering aNSC mediated cell replacement in injury and disease.


Asunto(s)
Sustancia Gris/metabolismo , Células-Madre Neurales/citología , Oligodendroglía/metabolismo , Sustancia Blanca/metabolismo , Células Madre Adultas/metabolismo , Animales , Astrocitos/metabolismo , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Ratones Endogámicos C57BL , Neuroglía/metabolismo , Ratas
4.
J Neuroinflammation ; 17(1): 323, 2020 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-33115477

RESUMEN

BACKGROUND: The neuronal ceroid lipofuscinoses (CLN diseases) are fatal lysosomal storage diseases causing neurodegeneration in the CNS. We have previously shown that neuroinflammation comprising innate and adaptive immune reactions drives axonal damage and neuron loss in the CNS of palmitoyl protein thioesterase 1-deficient (Ppt1-/-) mice, a model of the infantile form of the diseases (CLN1). Therefore, we here explore whether pharmacological targeting of innate immune cells modifies disease outcome in CLN1 mice. METHODS: We applied treatment with PLX3397 (150 ppm in the chow), a potent inhibitor of the colony stimulating factor-1 receptor (CSF-1R) to target innate immune cells in CLN1 mice. Experimental long-term treatment was non-invasively monitored by longitudinal optical coherence tomography and rotarod analysis, as well as analysis of visual acuity, myoclonic jerks, and survival. Treatment effects regarding neuroinflammation, neural damage, and neurodegeneration were subsequently analyzed by histology and immunohistochemistry. RESULTS: We show that PLX3397 treatment attenuates neuroinflammation in CLN1 mice by depleting pro-inflammatory microglia/macrophages. This leads to a reduction of T lymphocyte recruitment, an amelioration of axon damage and neuron loss in the retinotectal system, as well as reduced thinning of the inner retina and total brain atrophy. Accordingly, long-term treatment with the inhibitor also ameliorates clinical outcomes in CLN1 mice, such as impaired motor coordination, visual acuity, and myoclonic jerks. However, we detected a sex- and region-biased efficacy of CSF-1R inhibition, with male microglia/macrophages showing higher responsiveness toward depletion, especially in the gray matter of the CNS. This results in a better treatment outcome in male Ppt1-/- mice regarding some histopathological and clinical readouts and reflects heterogeneity of innate immune reactions in the diseased CNS. CONCLUSIONS: Our results demonstrate a detrimental impact of innate immune reactions in the CNS of CLN1 mice. These findings provide insights into CLN pathogenesis and may guide in the design of immunomodulatory treatment strategies.


Asunto(s)
Aminopiridinas/uso terapéutico , Encéfalo/efectos de los fármacos , Macrófagos/efectos de los fármacos , Microglía/efectos de los fármacos , Lipofuscinosis Ceroideas Neuronales/tratamiento farmacológico , Pirroles/uso terapéutico , Aminopiridinas/farmacología , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Macrófagos/patología , Masculino , Ratones , Microglía/patología , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/inmunología , Degeneración Nerviosa/patología , Lipofuscinosis Ceroideas Neuronales/inmunología , Lipofuscinosis Ceroideas Neuronales/patología , Neuronas/efectos de los fármacos , Neuronas/patología , Pirroles/farmacología , Retina/efectos de los fármacos , Retina/patología , Factores Sexuales , Linfocitos T/efectos de los fármacos , Linfocitos T/patología , Tomografía de Coherencia Óptica
5.
Int J Mol Sci ; 21(12)2020 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-32570968

RESUMEN

Mesenchymal stem cell (MSC)-secreted factors have been shown to significantly promote oligodendrogenesis from cultured primary adult neural stem cells (aNSCs) and oligodendroglial precursor cells (OPCs). Revealing underlying mechanisms of how aNSCs can be fostered to differentiate into a specific cell lineage could provide important insights for the establishment of novel neuroregenerative treatment approaches aiming at myelin repair. However, the nature of MSC-derived differentiation and maturation factors acting on the oligodendroglial lineage has not been identified thus far. In addition to missing information on active ingredients, the degree to which MSC-dependent lineage instruction is functional in vivo also remains to be established. We here demonstrate that MSC-derived factors can indeed stimulate oligodendrogenesis and myelin sheath generation of aNSCs transplanted into different rodent central nervous system (CNS) regions, and furthermore, we provide insights into the underlying mechanism on the basis of a comparative mass spectrometry secretome analysis. We identified a number of secreted proteins known to act on oligodendroglia lineage differentiation. Among them, the tissue inhibitor of metalloproteinase type 1 (TIMP-1) was revealed to be an active component of the MSC-conditioned medium, thus validating our chosen secretome approach.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células-Madre Neurales/citología , Oligodendroglía/citología , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Células Madre Adultas/citología , Animales , Diferenciación Celular , Células Cultivadas , Medios de Cultivo Condicionados/química , Femenino , Células Madre Mesenquimatosas/metabolismo , Cultivo Primario de Células , Proteómica , Ratas , Trasplante de Células Madre
6.
Glia ; 67(2): 277-290, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30565754

RESUMEN

Genetically caused neurological disorders of the central nervous system (CNS) usually result in poor or even fatal clinical outcome and few or no causative treatments are available. Often, these disorders are associated with disease-amplifying neuroinflammation, a feature shared by progressive forms of multiple sclerosis (PMS), another poorly treatable disorder of the CNS. We have previously generated two mouse lines carrying distinct mutations in the oligodendrocytic PLP1 gene that have initially been identified in patients fulfilling clinical criteria for multiple sclerosis (MS). These mutations cause a loss of function of the gene product resulting in a histopathological and clinical phenotype common to both PMS and genetic CNS disorders, like hereditary spastic paraplegias. Importantly, neuroinflammation comprising adaptive immune reactions promotes disease progression in these PLP1 mutant models, opening the possibility to improve disease outcome of the respective disorders by targeting/modulating inflammation. We here show that PLX3397, a potent inhibitor of the CSF-1R and targeting innate immune cells, attenuates neuroinflammation in our models by reducing numbers of resident microglia and attenuating T-lymphocyte recruitment in the CNS. This leads to an amelioration of demyelination, axonopathic features and neuron loss in the retinotectal system, also reflected by reduced thinning of the inner retinal composite layer in longitudinal studies using noninvasive optical coherence tomography. Our findings identify microglia as important promoters of neuroinflammation-related neural damage and CSF-1R inhibition as a possible therapeutic strategy not only for PMS but also for inflammation-related genetic diseases of the nervous system for which causal treatment options are presently lacking.


Asunto(s)
Enfermedades del Sistema Nervioso Central/complicaciones , Enfermedades del Sistema Nervioso Central/genética , Inflamación , Microglía/metabolismo , Mutación/genética , Proteína Proteolipídica de la Mielina/genética , Aminopiridinas/uso terapéutico , Animales , Antiinflamatorios/uso terapéutico , Citocinas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Humanos , Inflamación/tratamiento farmacológico , Inflamación/etiología , Inflamación/genética , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/patología , Microglía/ultraestructura , Microscopía Electrónica de Transmisión , Proteína Proteolipídica de la Mielina/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Neuronas/ultraestructura , Pirroles/uso terapéutico , Linfocitos T/efectos de los fármacos , Linfocitos T/patología , Tomografía de Coherencia Óptica
7.
Hum Mol Genet ; 25(21): 4686-4702, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28173160

RESUMEN

Progressive forms of multiple sclerosis lead to chronic disability, substantial decline in quality of life and reduced longevity. It is often suggested that they occur independently of inflammation. Here we investigated the disease progression in mouse models carrying PLP1 point mutations previously found in patients displaying clinical features of multiple sclerosis. These mouse models show loss-of-function of PLP1 associated with neuroinflammation; the latter leading to clinically relevant axonal degeneration, neuronal loss and brain atrophy as demonstrated by inactivation of the recombination activating gene 1. Moreover, these pathological hallmarks were substantially amplified when we attenuated immune regulation by inactivation of the programmed cell death-1 gene. Our observations support the view that primary oligodendroglial abnormalities can evoke pathogenically relevant neuroinflammation that drives neurodegeneration, as observed in some forms of multiple sclerosis but also in other, genetically-mediated neurodegenerative disorders of the human nervous system. As many potent immunomodulatory drugs have emerged during the last years, it is tempting to consider immunomodulation as a treatment option not only for multiple sclerosis, but also for so far non-treatable, genetically-mediated disorders of the nervous system accompanied by pathogenic neuroinflammation.


Asunto(s)
Esclerosis Múltiple/genética , Mutación , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Factores Inmunológicos/genética , Factores Inmunológicos/inmunología , Inflamación/genética , Inflamación/metabolismo , Masculino , Ratones , Ratones Transgénicos , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/metabolismo
8.
J Neuroinflammation ; 15(1): 194, 2018 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-29970109

RESUMEN

BACKGROUND: Genetically caused neurological disorders of the central nervous system (CNS) are mostly characterized by poor or even fatal clinical outcome and few or no causative treatments are available. Often, these disorders are associated with low-grade, disease-promoting inflammation, another feature shared by progressive forms of multiple sclerosis (PMS). We previously generated two mouse lines carrying distinct mutations in the oligodendrocytic PLP1 gene that have initially been identified in patients diagnosed with MS. These mutations cause a loss of PLP function leading to a histopathological and clinical phenotype common to both PMS and genetic CNS disorders, like hereditary spastic paraplegias. Importantly, neuroinflammation promotes disease progression in these models, suggesting that pharmacological modulation of inflammation might ameliorate disease outcome. METHODS: We applied teriflunomide, an approved medication for relapsing-remitting MS targeting activated T-lymphocytes, in the drinking water (10 mg/kg body weight/day). Experimental long-term treatment of PLP mutant mice was non-invasively monitored by longitudinal optical coherence tomography and by rotarod analysis. Immunomodulatory effects were subsequently analyzed by flow cytometry and immunohistochemistry and treatment effects regarding neural damage, and neurodegeneration were assessed by histology and immunohistochemistry. RESULTS: Preventive treatment with teriflunomide attenuated the increase in number of CD8+ cytotoxic effector T cells and fostered the proliferation of CD8+ CD122+ PD-1+ regulatory T cells in the CNS. This led to an amelioration of axonopathic features and neuron loss in the retinotectal system, also reflected by reduced thinning of the innermost retinal composite layer in longitudinal studies and ameliorated clinical outcome upon preventive long-term treatment. Treatment of immune-incompetent PLP mutants did not provide evidence for a direct, neuroprotective effect of the medication. When treatment was terminated, no rebound of neuroinflammation occurred and histopathological improvement was preserved for at least 75 days without treatment. After disease onset, teriflunomide halted ongoing axonal perturbation and enabled a recovery of dendritic arborization by surviving ganglion cells. However, neither neuron loss nor clinical features were ameliorated, likely due to already advanced neurodegeneration before treatment onset. CONCLUSIONS: We identify teriflunomide as a possible medication not only for PMS but also for inflammation-related genetic diseases of the nervous system for which causal treatment options are presently lacking.


Asunto(s)
Antiinflamatorios/uso terapéutico , Crotonatos/uso terapéutico , Inflamación , Leucocitos/patología , Esclerosis Múltiple , Toluidinas/uso terapéutico , Animales , Antígenos CD/metabolismo , Axones/efectos de los fármacos , Axones/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hidroxibutiratos , Inflamación/tratamiento farmacológico , Inflamación/etiología , Inflamación/patología , Antígeno Ki-67/metabolismo , Leucocitos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/patología , Esclerosis Múltiple/complicaciones , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Mutación/genética , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , Nitrilos , Receptor de Muerte Celular Programada 1/metabolismo , Retina/patología
9.
Mol Ther ; 25(8): 1889-1899, 2017 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-28506594

RESUMEN

CLN diseases are rare lysosomal storage diseases characterized by progressive axonal degeneration and neuron loss in the CNS, manifesting in disability, blindness, and premature death. We have previously demonstrated that, in animal models of infantile and juvenile forms of CLN disease (CLN1 and CLN3, respectively), secondary neuroinflammation in the CNS substantially amplifies neural damage, opening the possibility that immunomodulatory treatment might improve disease outcome. First, we recapitulated the inflammatory phenotype, originally seen in mice in autopsies of CLN patients. We then treated mouse models of CLN1 and CLN3 disease with the clinically approved immunomodulatory compounds fingolimod (0.5 mg/kg/day) and teriflunomide (10 mg/kg/day) by consistent supply in the drinking water for 5 months. The treatment was well tolerated and reduced T cell numbers and microgliosis in the CNS of both models. Moreover, axonal damage, neuron loss, retinal thinning, and brain atrophy were substantially attenuated in both models, along with reduced frequency of myoclonic jerks in Ppt1-/- mice. Based on these findings, and because side effects were not detected, we suggest that clinically approved immune modulators such as fingolimod and teriflunomide may be suitable to attenuate progression of CLN1 and CLN3 disease and, possibly, other orphan diseases with pathogenically relevant neuroinflammation.


Asunto(s)
Crotonatos/farmacología , Clorhidrato de Fingolimod/farmacología , Lipofuscinosis Ceroideas Neuronales/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Toluidinas/farmacología , Adolescente , Adulto , Aminopeptidasas/genética , Aminopeptidasas/metabolismo , Animales , Axones/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Niño , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Hidroxibutiratos , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Lipofuscinosis Ceroideas Neuronales/tratamiento farmacológico , Lipofuscinosis Ceroideas Neuronales/patología , Neuronas/patología , Nitrilos , Serina Proteasas/genética , Serina Proteasas/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Tioléster Hidrolasas/deficiencia , Tripeptidil Peptidasa 1 , Adulto Joven
10.
J Neurosci ; 36(6): 1890-901, 2016 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-26865613

RESUMEN

Previous studies in myelin-mutant mouse models of the inherited and incurable nerve disorder, Charcot-Marie-Tooth (CMT) neuropathy, have demonstrated that low-grade secondary inflammation implicating phagocytosing macrophages amplifies demyelination, Schwann cell dedifferentiation and perturbation of axons. The cytokine colony stimulating factor-1 (CSF-1) acts as an important regulator of these macrophage-related disease mechanisms, as genetic and pharmacologic approaches to block the CSF-1/CSF-1R signaling result in a significant alleviation of pathological alterations in mutant peripheral nerves. In mouse models of CMT1A and CMT1X, as well as in human biopsies, CSF-1 is predominantly expressed by endoneurial fibroblasts, which are closely associated with macrophages, suggesting local stimulatory mechanisms. Here we investigated the impact of cell-surface and secreted isoforms of CSF-1 on macrophage-related disease in connexin32-deficient (Cx32def) mice, a mouse model of CMT1X. Our present observations suggest that the secreted proteoglycan isoform (spCSF-1) is predominantly expressed by fibroblasts, whereas the membrane-spanning cell-surface isoform (csCSF-1) is expressed by macrophages. Using crossbreeding approaches to selectively restore or overexpress distinct isoforms in CSF-1-deficient (osteopetrotic) Cx32def mice, we demonstrate that both isoforms equally regulate macrophage numbers dose-dependently. However, spCSF-1 mediates macrophage activation and macrophage-related neural damage, whereas csCSF-1 inhibits macrophage activation and attenuates neuropathy. These results further corroborate the important role of secondary inflammation in mouse models of CMT1 and might identify specific targets for therapeutic approaches to modulate innate immune reactions. SIGNIFICANCE STATEMENT: Mouse models of Charcot-Marie-Tooth neuropathy have indicated that low-grade secondary inflammation involving phagocytosing macrophages amplifies demyelination, Schwann cell dedifferentiation, and perturbation of axons. The recruitment and pathogenic activation of detrimental macrophages is regulated by CSF-1, a cytokine that is mostly expressed by fibroblasts in the diseased nerve and exists in three isoforms. We show that the cell-surface and secreted isoforms of CSF-1 have opposing effects on macrophage activation and disease progression in a mouse model of CMT1X. These insights into opposing functions of disease-modulating cytokine isoforms might enable the development of specific therapeutic approaches.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Conexinas/genética , Conexinas/metabolismo , Factor Estimulante de Colonias de Macrófagos/genética , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/patología , Proteínas de la Membrana/metabolismo , Neuronas/patología , Animales , Enfermedad de Charcot-Marie-Tooth/metabolismo , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/patología , Dosificación de Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades del Sistema Nervioso Periférico/genética , Enfermedades del Sistema Nervioso Periférico/patología , Transducción de Señal/efectos de los fármacos , Proteína beta1 de Unión Comunicante
11.
Glia ; 65(9): 1407-1422, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28568966

RESUMEN

Genetically caused neurological disorders of the central nervous system (CNS) are usually orphan diseases with poor or even fatal clinical outcome and few or no treatments that will improve longevity or at least quality of life. Neuroinflammation is common to many of these disorders, despite the fact that a plethora of distinct mutations and molecular changes underlie the disorders. In this article, data from corresponding animal models are analyzed to define the roles of innate and adaptive inflammation as modifiers and amplifiers of disease. We describe both common and distinct patterns of neuroinflammation in genetically mediated CNS disorders and discuss the contrasting mechanisms that lead to adverse versus neuroprotective effects. Moreover, we identify the juxtaparanode as a neuroanatomical compartment commonly associated with inflammatory cells and ongoing axonopathic changes, in models of diverse diseases. The identification of key immunological effector pathways that amplify neuropathic features should lead to realistic possibilities for translatable therapeutic interventions using existing immunomodulators. Moreover, evidence emerges that neuroinflammation is not only able to modify primary neural damage-related symptoms but also may lead to unexpected clinical outcomes such as neuropsychiatric syndromes.


Asunto(s)
Enfermedades del Sistema Nervioso Central/inmunología , Enfermedades del Sistema Nervioso Central/terapia , Inflamación/fisiopatología , Inflamación/terapia , Animales , Enfermedades del Sistema Nervioso Central/genética , Humanos , Inflamación/genética , Neuroinmunomodulación/fisiología
12.
Glia ; 64(5): 792-809, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26775238

RESUMEN

CLN diseases are mostly fatal lysosomal storage diseases that lead to neurodegeneration in the CNS. We have previously shown that CD8+ T-lymphocytes contribute to axonal perturbation and neuron loss in the CNS of Ppt1(-/-) mice, a model of CLN1 disease. We now investigated the role of the inflammation-related cell adhesion molecule sialoadhesin (Sn) in Ppt1(-/-) and Cln3(-/-) mice, a model of the most frequent form, CLN3 disease. Microglia/macrophages in the CNS of both models showed an upregulation of Sn and markers for proinflammatory M1 polarization and antigen presentation. Sn+ microglia/macrophages associated with SMI32+ axonal spheroids and CD8+ T-lymphocytes. To analyze their pathogenic impact, we crossbred both models with Sn-deficient mice and scored axonal degeneration and neuronal integrity using immunohistochemistry, electron microscopy and optical coherence tomography. Degenerative alterations in the retinotectal pathway of Ppt1(-/-)Sn(-/-) and Cln3(-/-)Sn(-/-) mice were significantly reduced. Ppt1(-/-)Sn(-/-) mice also showed a substantially improved clinical phenotype and extended lifespan, attenuated numbers of M1-polarized microglia/macrophages and reduced expression levels of proinflammatory cytokines. This was accompanied by an increased frequency of CD8+CD122+ T-lymphocytes in the CNS of Ppt1(-/-)Sn(-/-) mice, the regulatory phenotype of which was demonstrated by impaired survival of CD8+CD122- effector T-lymphocytes in co-culture experiments. We show for the first time that increased Sn expression on microglia/macrophages contributes to neural perturbation in two distinct models of CLN disease. Our data also indicate that a rarely described CD8+CD122+ T-cell population can regulate the corresponding diseases. These studies provide insights into CLN pathogenesis and may guide in designing immuno-regulatory treatment strategies.


Asunto(s)
Astrocitos/patología , Encefalitis/etiología , Lipofuscinosis Ceroideas Neuronales/complicaciones , Lipofuscinosis Ceroideas Neuronales/patología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/metabolismo , Animales , Antígenos CD/metabolismo , Astrocitos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Proteínas de Unión al Calcio/metabolismo , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Encefalitis/patología , Macrófagos/metabolismo , Macrófagos/patología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Microglía/metabolismo , Microglía/patología , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Retina/patología , Retina/ultraestructura , Lectina 1 Similar a Ig de Unión al Ácido Siálico/genética , Tioléster Hidrolasas/genética , Tioléster Hidrolasas/metabolismo , Tomografía de Coherencia Óptica
13.
Brain ; 138(Pt 11): 3193-205, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26297559

RESUMEN

See Scherer (doi:10.1093/awv279) for a scientific commentary on this article.Charcot-Marie-Tooth type 1 neuropathies are inherited disorders of the peripheral nervous system caused by mutations in Schwann cell-related genes. Typically, no causative cure is presently available. Previous preclinical data of our group highlight the low grade, secondary inflammation common to distinct Charcot-Marie-Tooth type 1 neuropathies as a disease amplifier. In the current study, we have tested one of several available clinical agents targeting macrophages through its inhibition of the colony stimulating factor 1 receptor (CSF1R). We here show that in two distinct mouse models of Charcot-Marie-Tooth type 1 neuropathies, the systemic short- and long-term inhibition of CSF1R by oral administration leads to a robust decline in nerve macrophage numbers by ∼70% and substantial reduction of the typical histopathological and functional alterations. Interestingly, in a model for the dominant X-linked form of Charcot-Marie-Tooth type 1 neuropathy, the second most common form of the inherited neuropathies, macrophage ablation favours maintenance of axonal integrity and axonal resprouting, leading to preserved muscle innervation, increased muscle action potential amplitudes and muscle strengths in the range of wild-type mice. In another model mimicking a mild, demyelination-related Charcot-Marie-Tooth type 1 neuropathy caused by reduced P0 (MPZ) gene dosage, macrophage blockade causes an improved preservation of myelin, increased muscle action potential amplitudes, improved nerve conduction velocities and ameliorated muscle strength. These observations suggest that disease-amplifying macrophages can produce multiple adverse effects in the affected nerves which likely funnel down to common clinical features. Surprisingly, treatment of mouse models mimicking Charcot-Marie-Tooth type 1A neuropathy also caused macrophage blockade, but did not result in neuropathic or clinical improvements, most likely due to the late start of treatment of this early onset disease model. In summary, our study shows that targeting peripheral nerve macrophages by an orally administered inhibitor of CSF1R may offer a highly efficacious and safe treatment option for at least two distinct forms of the presently non-treatable Charcot-Marie-Tooth type 1 neuropathies.


Asunto(s)
Axones/efectos de los fármacos , Enfermedad de Charcot-Marie-Tooth/inmunología , Enfermedades Desmielinizantes/inmunología , Fuerza de la Mano , Macrófagos/efectos de los fármacos , Conducción Nerviosa/efectos de los fármacos , Nervios Periféricos/efectos de los fármacos , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Animales , Axones/patología , Enfermedad de Charcot-Marie-Tooth/genética , Enfermedad de Charcot-Marie-Tooth/patología , Conexinas/deficiencia , Conexinas/genética , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Transgénicos , Proteína P0 de la Mielina/genética , Proteínas de la Mielina/genética , Nervios Periféricos/inmunología , Nervios Periféricos/patología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Proteína beta1 de Unión Comunicante
14.
Glia ; 63(6): 977-86, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25628221

RESUMEN

We investigated connexin 32 (Cx32)-deficient mice, a model for the X-linked form of Charcot-Marie-Tooth neuropathy (CMT1X), regarding the impact of low-grade inflammation on Schwann cell phenotype. Whereas we previously identified macrophages as amplifiers of the neuropathy, we now explicitly focus on the impact of the phagocytes on Schwann cell dedifferentiation, a so far not-yet addressed disease-related mechanism for CMT1X. Using mice heterozygously deficient for Cx32 and displaying both Cx32-positive and -negative Schwann cells in one and the same nerve, we could demonstrate that macrophage clusters rather than single macrophages precisely associate with mutant but not with Cx32-positive Schwann cells. Similarly, in an advanced stage of Schwann cell perturbation, macrophage clusters were strongly associated with NCAM- and L1-positive, dedifferentiated Schwann cells. To clarify the role of macrophages regarding Schwann cell dedifferentiation, we generated Cx32-deficient mice additionally deficient for the macrophage-directed cytokine colony-stimulating factor (CSF)-1. In the absence of CSF-1, Cx32-deficient Schwann cells not only showed the expected amelioration in myelin preservation but also failed to upregulate the Schwann cell dedifferentiation markers NCAM and L1. Another novel and unexpected finding in the double mutants was the retained activation of ERK signaling, a pathway which is detrimental for Schwann cell homeostasis in myelin mutant models. Our findings demonstrate that increased ERK signaling can be compatible with the maintenance of Schwann cell differentiation and homeostasis in vivo and identifies CSF-1-activated macrophages as crucial mediators of detrimental Schwann cell dedifferentiation in Cx32-deficient mice.


Asunto(s)
Conexinas/deficiencia , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/fisiología , Células de Schwann/fisiología , Animales , Desdiferenciación Celular/fisiología , Enfermedad de Charcot-Marie-Tooth , Quimiocina CCL2/metabolismo , Conexinas/genética , Modelos Animales de Enfermedad , Femenino , Homeostasis/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Factor Estimulante de Colonias de Macrófagos/genética , Macrófagos/ultraestructura , Ratones Endogámicos C57BL , Ratones Transgénicos , Vaina de Mielina/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Células de Schwann/ultraestructura , Nervio Ciático/fisiopatología , Nervio Ciático/ultraestructura , Proteína beta1 de Unión Comunicante
15.
J Neuroinflammation ; 12: 49, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25879857

RESUMEN

BACKGROUND: We could previously identify components of both the innate and the adaptive immune system as disease modifiers in the pathogenesis of models for Charcot-Marie-Tooth (CMT) neuropathies type 1B and 1X. As part of the adaptive immune system, here we investigated the role of antibodies in a model for CMT1B. METHODS: Antibodies were localized and characterized in peripheral nerves of the CMT1B model by immunohistochemistry and Western blot analysis. Experimental ablation of antibodies was performed by cross breeding the CMT1B models with mutants deficient in B-lymphocytes (JHD-/- mutants). Ameliorated demyelination by antibody deficiency was reverted by intravenous injection of mouse IgG fractions. Histopathological analysis was performed by immunocytochemistry and light and quantitative electron microscopy. RESULTS: We demonstrate that in peripheral nerves of a mouse model for CMT1B, endogenous antibodies strongly decorate endoneurial tubes of peripheral nerves. These antibodies comprise IgG and IgM subtypes and are preferentially, but not exclusively, associated with nerve fiber aspects nearby the nodes of Ranvier. In the absence of antibodies, the early demyelinating phenotype is substantially ameliorated. Reverting the neuropathy by reconstitution with murine IgG fractions identified accumulating antibodies as potentially pathogenic at this early stage of disease. CONCLUSIONS: Our study demonstrates that in a mouse model for CMT1B, endogenous antibodies contribute to early macrophage-mediated demyelination and disease progression. Thus, both the innate and adaptive immune system are mutually interconnected in a genetic model for demyelination. Since in Wallerian degeneration antibodies have also been shown to be involved in myelin phagocytosis, our study supports our view that inherited demyelination and Wallerian degeneration share common mechanisms, which are detrimental when activated under nonlesion conditions.


Asunto(s)
Anticuerpos/metabolismo , Enfermedad de Charcot-Marie-Tooth/complicaciones , Enfermedades Desmielinizantes/etiología , Análisis de Varianza , Animales , Antígenos de Diferenciación/metabolismo , Enfermedad de Charcot-Marie-Tooth/genética , Enfermedades Desmielinizantes/inmunología , Modelos Animales de Enfermedad , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína P0 de la Mielina/deficiencia , Proteína P0 de la Mielina/genética , Proteína P0 de la Mielina/inmunología , Neuropatía Ciática/metabolismo , Neuropatía Ciática/patología , Transducción de Señal/inmunología
16.
Glia ; 62(5): 736-50, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24526449

RESUMEN

We investigated three models for Charcot-Marie-Tooth type 1 (CMT1) neuropathy, comprising mice lacking connexin 32 (Cx32def), mice with reduced myelin protein zero (P0) expression (P0het) and transgenic mouse mutants overexpressing peripheral myelin protein 22 (PMP22tg), with regard of the expression of the developmentally regulated molecules NCAM, L1, the low-affinity NGF-receptor p75 (p75(NTR) ) and the transcription factor component c-Jun. We found that all molecules were uniformly expressed by myelin deficient and supernumerary Schwann cells. The mutant myelinating Schwann cells of PMP22tg mice showed a robust NCAM-immunoreactivity in Schmidt-Lanterman incisures (SLI) that accompanies other early onset abnormalities, such as the presence of supernumerary Schwann cells and impaired myelin formation in some fibers. In line with this, Cx32def and P0het mice, which represent demyelinating models, only rarely express NCAM in SLI. Surprisingly, c-Jun immunoreactivity displayed a mosaic-like pattern with mostly negative and some weakly or moderately positive nuclei both in myelinating Schwann cells and Remak cells of wildtype (wt), P0het and PMP22tg mice. However, c-Jun expression was substantially upregulated in myelinating Schwann cells of Cx32def mice and spatially associated with axon perturbation, a typical predemyelinating feature of Cx32 deficiency. Additionally, c-Jun upregulation was correlated with an elevated level of GDNF, possibly causally linked to the typical compensatory sprouting of axons in Cx32def mice and CMT1X patients. Our findings suggest that in myelinating Schwann cells of distinct models of CMT1, c-Jun upregulation is a marker for predemyelinating axonal perturbation while myelin-related NCAM expression is indicative for early Schwann cell abnormalities.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/metabolismo , Genes jun/fisiología , Fibras Nerviosas Mielínicas/metabolismo , Moléculas de Adhesión de Célula Nerviosa/biosíntesis , Células de Schwann/metabolismo , Regulación hacia Arriba/fisiología , Animales , Axones/metabolismo , Axones/patología , Enfermedad de Charcot-Marie-Tooth/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fibras Nerviosas Mielínicas/patología , Células de Schwann/patología
17.
Am J Pathol ; 183(3): 655-60, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23831295

RESUMEN

Wallerian degeneration (WD) and inherited demyelinating neuropathies of the Charcot-Marie-Tooth type 1 (CMT1) appear to represent completely distinct events. CMT1-like diseases are chronic disorders of peripheral nerves that are genetically caused and lead to secondary neurodegenerative events, resulting in usually non-treatable disabilities, whereas WD is an acute, usually transient, reaction on injuries, aiming to allow peripheral nerve regeneration. Despite these differences, there are some striking similarities regarding molecular characteristics of neural cells in the affected peripheral nerves. The most conspicuous similarities might comprise the inflammatory component in both situations, as identified in appropriate mouse models. However, although inflammation is a beneficial component in WD, leading to removal of regrowth-repellent myelin debris, inflammation in CMT1 mouse models causes damage of initially intact nerve fibers. We hypothesize that, in CMT1 models, molecular pathways are activated that are shared with an important repair program after peripheral nerve injury, but lead to neural perturbation when activated under nonlesion conditions, as is the case in CMT1. These novel insights into the pathogenesis of CMT1 might be instrumental for the development of new therapeutic options in humans.


Asunto(s)
Axones/patología , Enfermedades Desmielinizantes/patología , Enfermedades Genéticas Congénitas/patología , Vaina de Mielina/patología , Regeneración Nerviosa , Degeneración Walleriana/complicaciones , Degeneración Walleriana/patología , Animales , Enfermedades Desmielinizantes/complicaciones , Enfermedades Genéticas Congénitas/complicaciones , Humanos , Inflamación/complicaciones , Inflamación/patología
18.
Brain ; 136(Pt 4): 1083-101, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23485853

RESUMEN

The neuronal ceroid lipofuscinoses are fatal neurodegenerative disorders in which the visual system is affected early in disease progression. A typical accompanying feature is neuroinflammation, the pathogenic impact of which is presently obscure. Here we investigated the role of inflammatory cells in palmitoyl protein thioesterase 1-deficient (Ppt1(-/-)) mice, a model of infantile neuronal ceroid lipofuscinosis (CLN1 disease, infantile), predominantly focusing on the visual system. We detected an early infiltration of CD8+ T-lymphocytes and observed activation of microglia/macrophage-like cells. To analyse the pathogenic impact of lymphocytes, we crossbred Ppt1(-/-) mice with mutants lacking lymphocytes (Rag1(-/-)), and scored axonal transport, axonal perturbation and neuronal survival. This lack of lymphocytes led to a significant amelioration of disease phenotypes, not only in the retino-tectal system, but also in other regions of the central nervous system. Finally, reconstitution experiments revealed a crucial role of CD8+ T-lymphocytes in pathogenesis. Our study provides novel pathomechanistic insights that may be crucial for developing treatment strategies.


Asunto(s)
Axones/patología , Lipofuscinosis Ceroideas Neuronales/inmunología , Lipofuscinosis Ceroideas Neuronales/patología , Neuronas/inmunología , Neuronas/patología , Animales , Axones/inmunología , Modelos Animales de Enfermedad , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Lipofuscinosis Ceroideas Neuronales/genética , Fenotipo , Linfocitos T , Tioléster Hidrolasas/deficiencia , Tioléster Hidrolasas/genética
19.
Front Neurosci ; 18: 1299554, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38435059

RESUMEN

We have previously demonstrated that neuroinflammation by the adaptive immune system acts as a robust and targetable disease amplifier in a mouse model of Spastic Paraplegia, type 11 (SPG11), a complicated form of Hereditary Spastic Paraplegia (HSP). While we identified an impact of neuroinflammation on distinct neuropathological changes and gait performance, neuropsychological features, typical and clinically highly relevant symptoms of complicated HSPs, were not addressed. Here we show that the corresponding SPG11 mouse model shows distinct behavioral abnormalities, particularly related to social behavior thus partially reflecting the neuropsychological changes in patients. We provide evidence that some behavioral abnormalities can be mitigated by genetic inactivation of the adaptive immune system. Translating this into a clinically applicable approach, we show that treatment with the established immunomodulators fingolimod or teriflunomide significantly attenuates distinct behavioral abnormalities, with the most striking effect on social behavior. This study links neuroinflammation to behavioral abnormalities in a mouse model of SPG11 and may thus pave the way for using immunomodulators as a treatment approach for SPG11 and possibly other complicated forms of HSP with neuropsychological involvement.

20.
Brain ; 135(Pt 1): 88-104, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22094537

RESUMEN

Previous studies in our laboratory have shown that in models for three distinct forms of the inherited and incurable nerve disorder, Charcot-Marie-Tooth neuropathy, low-grade inflammation implicating phagocytosing macrophages mediates demyelination and perturbation of axons. In the present study, we focus on colony-stimulating factor-1, a cytokine implicated in macrophage differentiation, activation and proliferation and fostering neural damage in a model for Charcot-Marie-Tooth neuropathy 1B. By crossbreeding a model for the X-linked form of Charcot-Marie-Tooth neuropathy with osteopetrotic mice, a spontaneous null mutant for colony-stimulating factor-1, we demonstrate a robust and persistent amelioration of demyelination and axon perturbation. Furthermore, functionally important domains of the peripheral nervous system, such as juxtaparanodes and presynaptic terminals, were preserved in the absence of colony-stimulating factor-1-dependent macrophage activation. As opposed to other Schwann cell-derived cytokines, colony-stimulating factor-1 is expressed by endoneurial fibroblasts, as revealed by in situ hybridization, immunocytochemistry and detection of ß-galactosidase expression driven by the colony-stimulating factor-1 promoter. By both light and electron microscopic studies, we detected extended cell-cell contacts between the colony-stimulating factor-1-expressing fibroblasts and endoneurial macrophages as a putative prerequisite for the effective and constant activation of macrophages by fibroblasts in the chronically diseased nerve. Interestingly, in human biopsies from patients with Charcot-Marie-Tooth type 1, we also found frequent cell-cell contacts between macrophages and endoneurial fibroblasts and identified the latter as main source for colony-stimulating factor-1. Therefore, our study provides strong evidence for a similarly pathogenic role of colony-stimulating factor-1 in genetically mediated demyelination in mice and Charcot-Marie-Tooth type 1 disease in humans. Thus, colony-stimulating factor-1 or its cognate receptor are promising target molecules for treating the detrimental, low-grade inflammation of several inherited neuropathies in humans.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/metabolismo , Neuronas/metabolismo , Animales , Enfermedad de Charcot-Marie-Tooth/genética , Enfermedad de Charcot-Marie-Tooth/patología , Conexinas/genética , Conexinas/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Activación de Macrófagos , Factor Estimulante de Colonias de Macrófagos/genética , Macrófagos/patología , Ratones , Neuronas/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Nervio Sural/metabolismo , Nervio Sural/patología , Regulación hacia Arriba , beta-Galactosidasa/metabolismo , Proteína beta1 de Unión Comunicante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA