Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
FASEB J ; 37(6): e22939, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37130013

RESUMEN

Traumatic spinal cord injury (SCI) most often leads to permanent paralysis due to the inability of axons to regenerate in the adult mammalian central nervous system (CNS). In the past, we have shown that mast cells (MCs) improve the functional outcome after SCI by suppressing scar tissue formation at the lesion site via mouse mast cell protease 6 (mMCP6). In this study, we investigated whether recombinant mMCP6 can be used therapeutically to improve the functional outcome after SCI. Therefore, we applied mMCP6 locally via an intrathecal catheter in the subacute phase after a spinal cord hemisection injury in mice. Our findings showed that hind limb motor function was significantly improved in mice that received recombinant mMCP6 compared with the vehicle-treated group. In contrast to our previous findings in mMCP6 knockout mice, the lesion size and expression levels of the scar components fibronectin, laminin, and axon-growth-inhibitory chondroitin sulfate proteoglycans were not affected by the treatment with recombinant mMCP6. Surprisingly, no difference in infiltration of CD4+ T cells and reactivity of Iba-1+ microglia/macrophages at the lesion site was observed between the mMCP6-treated mice and control mice. Additionally, local protein levels of the pro- and anti-inflammatory mediators IL-1ß, IL-2, IL-4, IL-6, IL-10, TNF-α, IFNγ, and MCP-1 were comparable between the two treatment groups, indicating that locally applied mMCP6 did not affect inflammatory processes after injury. However, the increase in locomotor performance in mMCP6-treated mice was accompanied by reduced demyelination and astrogliosis in the perilesional area after SCI. Consistently, we found that TNF-α/IL-1ß-astrocyte activation was decreased and that oligodendrocyte precursor cell (OPC) differentiation was increased after recombinant mMCP6 treatment in vitro. Mechanistically, this suggests effects of mMCP6 on reducing astrogliosis and improving (re)myelination in the spinal cord after injury. In conclusion, these data show for the first time that recombinant mMCP6 is therapeutically active in enhancing recovery after SCI.


Asunto(s)
Remielinización , Traumatismos de la Médula Espinal , Ratones , Animales , Gliosis/tratamiento farmacológico , Gliosis/metabolismo , Cicatriz/tratamiento farmacológico , Cicatriz/prevención & control , Mastocitos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/metabolismo , Médula Espinal/metabolismo , Ratones Noqueados , Recuperación de la Función , Modelos Animales de Enfermedad , Mamíferos
2.
J Neuroinflammation ; 19(1): 102, 2022 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-35488301

RESUMEN

BACKGROUND: Spinal cord injury (SCI) elicits a robust neuroinflammatory reaction which, in turn, exacerbates the initial mechanical damage. Pivotal players orchestrating this response are macrophages (Mφs) and microglia. After SCI, the inflammatory environment is dominated by pro-inflammatory Mφs/microglia, which contribute to secondary cell death and prevent regeneration. Therefore, reprogramming Mφ/microglia towards a more anti-inflammatory and potentially neuroprotective phenotype has gained substantial therapeutic interest in recent years. Interleukin-13 (IL-13) is a potent inducer of such an anti-inflammatory phenotype. In this study, we used genetically modified Mφs as carriers to continuously secrete IL-13 (IL-13 Mφs) at the lesion site. METHODS: Mφs were genetically modified to secrete IL-13 (IL-13 Mφs) and were phenotypically characterized using qPCR, western blot, and ELISA. To analyze the therapeutic potential, the IL-13 Mφs were intraspinally injected at the perilesional area after hemisection SCI in female mice. Functional recovery and histopathological improvements were evaluated using the Basso Mouse Scale score and immunohistochemistry. Neuroprotective effects of IL-13 were investigated using different cell viability assays in murine and human neuroblastoma cell lines, human neurospheroids, as well as murine organotypic brain slice cultures. RESULTS: In contrast to Mφs prestimulated with recombinant IL-13, perilesional transplantation of IL-13 Mφs promoted functional recovery following SCI in mice. This improvement was accompanied by reduced lesion size and demyelinated area. The local anti-inflammatory shift induced by IL-13 Mφs resulted in reduced neuronal death and fewer contacts between dystrophic axons and Mφs/microglia, suggesting suppression of axonal dieback. Using IL-4Rα-deficient mice, we show that IL-13 signaling is required for these beneficial effects. Whereas direct neuroprotective effects of IL-13 on murine and human neuroblastoma cell lines or human neurospheroid cultures were absent, IL-13 rescued murine organotypic brain slices from cell death, probably by indirectly modulating the Mφ/microglia responses. CONCLUSIONS: Collectively, our data suggest that the IL-13-induced anti-inflammatory Mφ/microglia phenotype can preserve neuronal tissue and ameliorate axonal dieback, thereby promoting recovery after SCI.


Asunto(s)
Neuroblastoma , Fármacos Neuroprotectores , Traumatismos de la Médula Espinal , Animales , Femenino , Humanos , Interleucina-13/uso terapéutico , Macrófagos/metabolismo , Ratones , Fármacos Neuroprotectores/uso terapéutico , Traumatismos de la Médula Espinal/patología
3.
Brain ; 144(10): 2933-2945, 2021 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-34244729

RESUMEN

After spinal cord injury, macrophages can exert either beneficial or detrimental effects depending on their phenotype. Aside from their critical role in inflammatory responses, macrophages are also specialized in the recognition, engulfment, and degradation of pathogens, apoptotic cells, and tissue debris. They promote remyelination and axonal regeneration by removing inhibitory myelin components and cellular debris. However, excessive intracellular presence of lipids and dysregulated intracellular lipid homeostasis result in the formation of foamy macrophages. These develop a pro-inflammatory phenotype that may contribute to further neurological decline. Additionally, myelin-activated macrophages play a crucial role in axonal dieback and retraction. Here, we review the opposing functional consequences of phagocytosis by macrophages in spinal cord injury, including remyelination and regeneration versus demyelination, degeneration, and axonal dieback. Furthermore, we discuss how targeting the phagocytic ability of macrophages may have therapeutic potential for the treatment of spinal cord injury.


Asunto(s)
Enfermedades Desmielinizantes/metabolismo , Macrófagos/fisiología , Fagocitosis/fisiología , Remielinización/fisiología , Traumatismos de la Médula Espinal/metabolismo , Animales , Enfermedades Desmielinizantes/inmunología , Humanos , Traumatismos de la Médula Espinal/inmunología
4.
Brain Behav Immun ; 80: 129-145, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30851378

RESUMEN

A disintegrin and metalloproteinase 17 (ADAM17) is the major sheddase involved in the cleavage of a plethora of cytokines, cytokine receptors and growth factors, thereby playing a substantial role in inflammatory and regenerative processes after central nervous system trauma. By making use of a hypomorphic ADAM17 knockin mouse model as well as pharmacological ADAM10/ADAM17 inhibitors, we showed that ADAM17-deficiency or inhibition significantly increases clearance of apoptotic cells, promotes axon growth and improves functional recovery after spinal cord injury (SCI) in mice. Microglia-specific ADAM17-knockout (ADAM17flox+/+-Cx3Cr1 Cre+/-) mice also showed improved functional recovery similar to hypomorphic ADAM17 mice. In contrast, endothelial-specific (ADAM17flox+/+-Cdh5Pacs Cre+/-) and macrophage-specific (ADAM17flox+/+-LysM Cre+/-) ADAM17-knockout mice or bone marrow chimera with transplanted ADAM17-deficient macrophages, displayed no functional improvement compared to wild type mice. These data indicate that ADAM17 expression on microglia cells (and not on macrophages or endothelial cells) plays a detrimental role in inflammation and functional recovery after SCI.


Asunto(s)
Proteína ADAM17/metabolismo , Microglía/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Inflamación/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fagocitosis/inmunología , Fagocitosis/fisiología , Recuperación de la Función/fisiología
5.
Kidney Int ; 94(5): 926-936, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30158055

RESUMEN

Current therapies for treating antineutrophil cytoplasm autoantibody (ANCA)-associated vasculitis include cyclophosphamide and corticosteroids. Unfortunately, these agents are associated with severe adverse effects, despite inducing remission in most patients. Histone deacetylase inhibitors are effective in rodent models of inflammation and act synergistically with many pharmacological agents, including alkylating agents like cyclophosphamide. EDO-S101 is an alkylating fusion histone deacetylase inhibitor molecule combining the DNA alkylating effect of Bendamustine with a pan-histone deacetylase inhibitor, Vorinostat. Here we studied the effects of EDO-S101 in two established rodent models of ANCA-associated vasculitis: a passive mouse model of anti-myeloperoxidase IgG-induced glomerulonephritis and an active rat model of myeloperoxidase-ANCA microscopic polyangiitis. Although pretreatment with EDO-S101 reduced circulating leukocytes, it did not prevent the development of passive IgG-induced glomerulonephritis in mice. On the other hand, treatment in rats significantly reduced glomerulonephritis and lung hemorrhage. EDO-S101 also significantly depleted rat B and T cells, and induced DNA damage and apoptosis in proliferating human B cells, suggesting a selective effect on the adaptive immune response. Thus, EDO-S101 may have a role in treatment of ANCA-associated vasculitis, operating primarily through its effects on the adaptive immune response to the autoantigen myeloperoxidase.


Asunto(s)
Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/tratamiento farmacológico , Bencimidazoles/uso terapéutico , Inhibidores de Histona Desacetilasas/uso terapéutico , Peroxidasa/inmunología , Inmunidad Adaptativa/efectos de los fármacos , Alquilación , Animales , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/patología , Apoptosis/efectos de los fármacos , Bencimidazoles/farmacología , Reparación del ADN/efectos de los fármacos , Femenino , Células HL-60 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Endogámicas WKY
6.
Stem Cells ; 34(7): 1971-84, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26992046

RESUMEN

Transplantation of mesenchymal stem cells (MSCs) into injured or diseased tissue-for the in situ delivery of a wide variety of MSC-secreted therapeutic proteins-is an emerging approach for the modulation of the clinical course of several diseases and traumata. From an emergency point-of-view, allogeneic MSCs have numerous advantages over patient-specific autologous MSCs since "off-the-shelf" cell preparations could be readily available for instant therapeutic intervention following acute injury. Although we confirmed the in vitro immunomodulatory capacity of allogeneic MSCs on antigen-presenting cells with standard coculture experiments, allogeneic MSC grafts were irrevocably rejected by the host's immune system upon either intramuscular or intracerebral transplantation. In an attempt to modulate MSC allograft rejection in vivo, we transduced MSCs with an interleukin-13 (IL13)-expressing lentiviral vector. Our data clearly indicate that prolonged survival of IL13-expressing allogeneic MSC grafts in muscle tissue coincided with the induction of an alternatively activated macrophage phenotype in vivo and a reduced number of alloantigen-reactive IFNγ- and/or IL2-producing CD8(+) T cells compared to nonmodified allografts. Similarly, intracerebral IL13-expressing MSC allografts also exhibited prolonged survival and induction of an alternatively activated macrophage phenotype, although a peripheral T cell component was absent. In summary, this study demonstrates that both innate and adaptive immune responses are effectively modulated in vivo by locally secreted IL13, ultimately resulting in prolonged MSC allograft survival in both muscle and brain tissue. Stem Cells 2016;34:1971-1984.


Asunto(s)
Supervivencia de Injerto/inmunología , Interleucina-13/farmacología , Isoantígenos/inmunología , Activación de Linfocitos/efectos de los fármacos , Macrófagos/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Linfocitos T/inmunología , Aloinjertos/efectos de los fármacos , Aloinjertos/inmunología , Animales , Formación de Anticuerpos/efectos de los fármacos , Células Presentadoras de Antígenos/efectos de los fármacos , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Ingeniería Genética , Inmunomodulación/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Ratones , Microglía/efectos de los fármacos , Microglía/patología , Linfocitos T/efectos de los fármacos
7.
FASEB J ; 30(5): 2040-57, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26917739

RESUMEN

An important barrier for axon regeneration and recovery after traumatic spinal cord injury (SCI) is attributed to the scar that is formed at the lesion site. Here, we investigated the effect of mouse mast cell protease (mMCP) 6, a mast cell (MC)-specific tryptase, on scarring and functional recovery after a spinal cord hemisection injury. Functional recovery was significantly impaired in both MC-deficient and mMCP6-knockout (mMCP6(-/-)) mice after SCI compared with wild-type control mice. This decrease in locomotor performance was associated with an increased lesion size and excessive scarring at the injury site. Axon growth-inhibitory chondroitin sulfate proteoglycans and the extracellular matrix components fibronectin, laminin, and collagen IV were significantly up-regulated in MC-deficient and mMCP6(-/-) mice, with an increase in scar volume between 23 and 32%. A degradation assay revealed that mMCP6 directly cleaves fibronectin and collagen IV in vitro In addition, gene expression levels of the scar components fibronectin, aggrecan, and collagen IV were increased up to 6.8-fold in mMCP6(-/-) mice in the subacute phase after injury. These data indicate that endogenous mMCP6 has scar-suppressing properties after SCI via indirect cleavage of axon growth-inhibitory scar components and alteration of the gene expression profile of these factors.-Vangansewinkel, T., Geurts, N., Quanten, K., Nelissen, S., Lemmens, S., Geboes, L., Dooley, D., Vidal, P. M., Pejler, G., Hendrix, S. Mast cells promote scar remodeling and functional recovery after spinal cord injury via mouse mast cell protease 6.


Asunto(s)
Cicatriz/metabolismo , Mastocitos/fisiología , Traumatismos de la Médula Espinal/metabolismo , Triptasas/metabolismo , Cicatrización de Heridas/fisiología , Animales , Citocinas/genética , Citocinas/metabolismo , Matriz Extracelular , Regulación Enzimológica de la Expresión Génica/fisiología , Ratones , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Triptasas/genética
8.
J Neuroinflammation ; 13(1): 101, 2016 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-27154002

RESUMEN

BACKGROUND: The cytokine, interleukin (IL)-25, is thought to be critically involved in inducing a type 2 immune response which may contribute to regeneration after central nervous system (CNS) trauma. We investigated whether applying recombinant IL-25, locally or systemically, in a mouse model of spinal cord injury (SCI) improves functional and histological recovery. FINDINGS: Repeated systemic administration of IL-25 did not influence functional recovery following SCI. In contrast, a single local administration of IL-25 significantly worsened locomotor outcome, which was evident from a decreased Basso mouse scale (BMS) score compared with phosphate-buffered saline (PBS)-treated controls. This was accompanied by a significant increase in lesion size, demyelination, and T helper cell infiltration. CONCLUSIONS: These data show for the first time that IL-25 is either ineffective when applied systemically or detrimental to spinal cord recovery when applied locally. Our findings question the potential neuroprotective role of IL-25 following CNS trauma.


Asunto(s)
Interleucinas/metabolismo , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/fisiopatología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Antígenos CD4/metabolismo , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Interleucinas/farmacología , Locomoción/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteínas de Microfilamentos/metabolismo , Proteína Básica de Mielina/metabolismo , Traumatismos de la Médula Espinal/patología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Factores de Tiempo
9.
J Neuroinflammation ; 13(1): 288, 2016 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-27829467

RESUMEN

BACKGROUND: Promoting the neuroprotective and repair-inducing effector functions of microglia and macrophages, by means of M2 polarisation or alternative activation, is expected to become a new therapeutic approach for central nervous system (CNS) disorders in which detrimental pro-inflammatory microglia and/or macrophages display a major contribution to the neuropathology. In this study, we present a novel in vivo approach using intracerebral grafting of mesenchymal stem cells (MSC) genetically engineered to secrete interleukin 13 (IL13-MSC). METHODS: In the first experimental setup, control MSC and IL13-MSC were grafted in the CNS of eGFP+ bone marrow chimaeric C57BL/6 mice to histologically evaluate IL13-mediated expression of several markers associated with alternative activation, including arginase1 and Ym1, on MSC graft-recognising microglia and MSC graft-infiltrating macrophages. In the second experimental setup, IL13-MSC were grafted on the right side (or on both the right and left sides) of the splenium of the corpus callosum in wild-type C57BL/6 mice and in C57BL/6 CX3CR1eGFP/+CCR2RFP/+ transgenic mice. Next, CNS inflammation and demyelination was induced by means of a cuprizone-supplemented diet. The influence of IL13-MSC grafting on neuropathological alterations was monitored by non-invasive T 2-weighted magnetic resonance imaging (MRI) and quantitative histological analyses, as compared to cuprizone-treated mice with control MSC grafts and/or cuprizone-treated mice without MSC injection. RESULTS: In the first part of this study, we demonstrate that MSC graft-associated microglia and MSC graft-infiltrating macrophages are forced into alternative activation upon grafting of IL13-MSC, but not upon grafting of control MSC. In the second part of this study, we demonstrate that grafting of IL13-MSC, in addition to the recruitment of M2 polarised macrophages, limits cuprizone-induced microgliosis, oligodendrocyte death and demyelination. Furthermore, we here demonstrate that injection of IL13-MSC at both sides of the splenium leads to a superior protective effect as compared to a single injection at one side of the splenium. CONCLUSIONS: Controlled and localised production of IL13 by means of intracerebral MSC grafting has the potential to modulate cell graft- and pathology-associated microglial/macrophage responses, and to interfere with oligodendrocyte death and demyelinating events in the cuprizone mouse model.


Asunto(s)
Cuprizona/toxicidad , Enfermedades Desmielinizantes , Gliosis/etiología , Interleucina-13/metabolismo , Trasplante de Células Madre Mesenquimatosas , Inhibidores de la Monoaminooxidasa/toxicidad , Oligodendroglía/patología , Animales , Línea Celular Transformada , Citocinas/genética , Citocinas/metabolismo , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/diagnóstico por imagen , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/cirugía , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Imagen por Resonancia Magnética , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Básica de Mielina/metabolismo , Oligodendroglía/efectos de los fármacos
10.
Med Res Rev ; 35(4): 653-77, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25546087

RESUMEN

Many researchers have attempted to pharmacologically modulate the adrenergic system to control locomotion, pain, and spasms after central nervous system (CNS) trauma, although such efforts have led to conflicting results. Despite this, multiple studies highlight that α-adrenoceptors (α-ARs) are promising therapeutic targets because in the CNS, they are involved in reactivity to stressors and regulation of locomotion, pain, and spasms. These functions can be activated by direct modulation of these receptors on neuronal networks in the brain and the spinal cord. In addition, these multifunctional receptors are also broadly expressed on immune cells. This suggests that they might play a key role in modulating immunological responses, which may be crucial in treating spinal cord injury and traumatic brain injury as both diseases are characterized by a strong inflammatory component. Reducing the proinflammatory response will create a more permissive environment for axon regeneration and may support neuromodulation in combination therapies. However, pharmacological interventions are hindered by adrenergic system complexity and the even more complicated anatomical and physiological changes in the CNS after trauma. This review is the first concise overview of the pros and cons of α-AR modulation in the context of CNS trauma.


Asunto(s)
Dolor/metabolismo , Parálisis/metabolismo , Receptores Adrenérgicos alfa/metabolismo , Espasmo/metabolismo , Traumatismos del Sistema Nervioso/metabolismo , Animales , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/metabolismo , Humanos , Dolor/complicaciones , Parálisis/complicaciones , Espasmo/complicaciones , Traumatismos del Sistema Nervioso/complicaciones
11.
Neurobiol Dis ; 62: 260-72, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24075853

RESUMEN

Mast cells (MCs) are found abundantly in the central nervous system and play a complex role in neuroinflammatory diseases such as multiple sclerosis and stroke. In the present study, we show that MC-deficient Kit(W-sh/W-sh) mice display significantly increased astrogliosis and T cell infiltration as well as significantly reduced functional recovery after spinal cord injury compared to wildtype mice. In addition, MC-deficient mice show significantly increased levels of MCP-1, TNF-α, IL-10 and IL-13 protein levels in the spinal cord. Mice deficient in mouse mast cell protease 4 (mMCP4), an MC-specific chymase, also showed increased MCP-1, IL-6 and IL-13 protein levels in spinal cord samples and a decreased functional outcome after spinal cord injury. A degradation assay using supernatant from MCs derived from either mMCP4(-/-) mice or controls revealed that mMCP4 cleaves MCP-1, IL-6, and IL-13 suggesting a protective role for MC proteases in neuroinflammation. These data show for the first time that MCs may be protective after spinal cord injury and that they may reduce CNS damage by degrading inflammation-associated cytokines via the MC-specific chymase mMCP4.


Asunto(s)
Citocinas/metabolismo , Mastocitos/metabolismo , Serina Endopeptidasas/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Animales , Astrocitos/patología , Femenino , Mediadores de Inflamación/metabolismo , Locomoción/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Traumatismos de la Médula Espinal/patología , Linfocitos T/metabolismo , Vértebras Torácicas/lesiones
12.
Cytokine Growth Factor Rev ; 69: 80-89, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36114092

RESUMEN

Traumatic spinal cord injury (SCI) is a devastating condition that significantly impacts motor, sensory and autonomic function in patients. Despite advances in therapeutic approaches, there is still no curative therapy currently available. Neuroinflammation is a persisting event of the secondary injury phase of SCI that affects functional recovery, and modulation of the inflammatory response towards a beneficial anti-inflammatory state can improve recovery in preclinical SCI models. In human SCI patients, rehabilitative exercise, or motor rehabilitation as we will refer to it from here on out, remains the cornerstone of treatment to increase functional capacity and prevent secondary health implications. Motor rehabilitation is known to have anti-inflammatory effects; however, current literature is lacking in the description of the effect of motor rehabilitation on inflammation in the context of SCI. Understanding the effect on different inflammatory markers after SCI should enable the optimization of motor rehabilitation as a therapeutic regime. This review extensively describes the effect of motor rehabilitation on selected inflammatory mediators in both preclinical and human SCI studies. Additionally, we summarize how the type, duration, and intensity of motor rehabilitation can affect the inflammatory response after SCI. In doing so, we introduce a new perspective on how motor rehabilitation can be optimized as an immunomodulatory therapy to improve patient outcome after SCI.


Asunto(s)
Traumatismos de la Médula Espinal , Humanos , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/rehabilitación , Recuperación de la Función/fisiología , Inflamación/complicaciones , Inmunomodulación
13.
Adv Healthc Mater ; 12(26): e2300951, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37114899

RESUMEN

Spinal cord injury (SCI) is a devastating condition with no curative therapy currently available. Immunomodulation can be applied as a therapeutic strategy to drive alternative immune cell activation and promote a proregenerative injury microenvironment. Locally injected hydrogels carrying immunotherapeutic cargo directly to injured tissue offer an encouraging treatment approach from an immunopharmacological perspective. Gelatin methacrylate (GelMA) hydrogels are promising in this regard, however, detailed analysis on the immunogenicity of GelMA in the specific context of the SCI microenvironment is lacking. Here, the immunogenicity of GelMA hydrogels formulated with a translationally relevant photoinitiator is analyzed in vitro and ex vivo. 3% (w/v) GelMA, synthesized from gelatin type-A, is first identified as the optimal hydrogel formulation based on mechanical properties and cytocompatibility. Additionally, 3% GelMA-A does not alter the expression profile of key polarization markers in BV2 microglia or RAW264.7 macrophages after 48 h. Finally, it is shown for the first time that 3% GelMA-A can support the ex vivo culture of primary murine organotypic spinal cord slices for 14 days with no direct effect on glial fibrillary acidic protein (GFAP+ ) astrocyte or ionized calcium-binding adaptor molecule 1 (Iba-1+ ) microglia reactivity. This provides evidence that GelMA hydrogels can act as an immunotherapeutic hydrogel-based platform for preclinical SCI.


Asunto(s)
Gelatina , Traumatismos de la Médula Espinal , Ratones , Animales , Gelatina/farmacología , Gelatina/química , Hidrogeles/farmacología , Hidrogeles/química , Metacrilatos/farmacología , Traumatismos de la Médula Espinal/tratamiento farmacológico
14.
J Mech Behav Biomed Mater ; 142: 105856, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37087955

RESUMEN

Traumatic spinal cord injuries result from high impact forces acting on the spine and are proceeded by an extensive secondary inflammatory response resulting in motor, sensory, and autonomic dysfunction. Experimental in vivo traumatic spinal cord injuries in rodents using a contusion model have been extremely useful in elucidating the underlying pathophysiology of these injuries. However, the relationship between the pathophysiology and the biomechanical factors is still not well understood. Therefore, the aim of this research is to provide a comprehensive analysis of the biomechanics of traumatic spinal cord injury in a rat contusion model. This is achieved through the development and validation of a finite element model of the thoracic rat spinal cord and subsequently simulating controlled cortical impact-induced traumatic spinal cord injury. The effects of impactor velocity, depth, and geometry on the resulting stresses and strains within the spinal cord are investigated. Our results show that increasing impactor depth results in larger stresses and strains within the spinal cord tissue as expected. Further, for the first time ever our results show that impactor geometry (spherical versus cylindrical) plays an important role in the distribution and magnitude of stresses and strains within the cord. Therefore, finite element modelling can be a powerful tool used to predict stresses and strains that occur in spinal cord tissue during trauma.


Asunto(s)
Contusiones , Traumatismos de la Médula Espinal , Ratas , Animales , Análisis de Elementos Finitos , Roedores , Médula Espinal , Modelos Animales de Enfermedad
15.
R Soc Open Sci ; 10(11): 231037, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38034122

RESUMEN

Live-cell imaging is a common technique in microscopy to investigate dynamic cellular behaviour and permits the accurate and relevant analysis of a wide range of cellular and tissue parameters, such as motility, cell division, wound healing responses and calcium (Ca2+) signalling in cell lines, primary cell cultures and ex vivo preparations. Furthermore, this can occur under many experimental conditions, making live-cell imaging indispensable for biological research. Systems which maintain cells at physiological conditions outside of a CO2 incubator are often bulky, expensive and use proprietary components. Here we present an inexpensive, open-source temperature control system for in vitro live-cell imaging. Our system 'ThermoCyte', which is constructed from standard electronic components, enables precise tuning, control and logging of a temperature 'set point' for imaging cells at physiological temperature. We achieved stable thermal dynamics, with reliable temperature cycling and a standard deviation of 0.42°C over 1 h. Furthermore, the device is modular in nature and is adaptable to the researcher's specific needs. This represents simple, inexpensive and reliable tool for laboratories to carry out custom live-cell imaging protocols, on a standard laboratory bench, at physiological temperature.

16.
Pharmacol Ther ; 234: 108043, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34813862

RESUMEN

Spinal cord injury (SCI) is a complex medical and psychological challenge for which there is no curative therapy currently available. Despite major progress in pharmacological and surgical approaches, clinical trials for SCI patients have been uniformly disappointing thus far as there are many practical and biological issues yet to be resolved. Neuroinflammation is a critical event of the secondary injury phase after SCI, and recent research strategies have focused on modulating the immune response after injury to provide a more favorable recovery environment. Biomaterials can serve this purpose by providing physical and trophic support to the injured spinal cord after SCI. Of all potential biomaterials, functional hydrogels are emerging as a key component in novel treatment strategies for SCI, including controlled and localized delivery of immunomodulatory therapies to drive polarization of immune cells towards a pro-regenerative phenotype. Here, we extensively review recent developments in the use of functional hydrogels as immunomodulatory therapies for SCI. We briefly describe physicochemical properties of hydrogels and demonstrate how advanced fabrication methods lead to the required heterogeneity and hierarchical arrangements that increasingly mimic complex spinal cord tissue. We then summarize potential SCI therapeutic modalities including: (i) hydrogels alone; (ii) hydrogels as cellular or (iii) bioactive molecule delivery vehicles, and; (iv) combinatorial approaches. By linking the structural properties of hydrogels to their functions in treatment with particular focus on immunopharmacological stimuli, this may accelerate further development of functional hydrogels for SCI, and indeed next-generation central nervous system regenerative therapies.


Asunto(s)
Hidrogeles , Traumatismos de la Médula Espinal , Materiales Biocompatibles/uso terapéutico , Humanos , Hidrogeles/uso terapéutico , Regeneración Nerviosa , Traumatismos de la Médula Espinal/tratamiento farmacológico
17.
J Neurotrauma ; 37(3): 564-571, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31210094

RESUMEN

A mounting body of evidence suggests that stress plays a major role in the injury progression after spinal cord injury (SCI). Injury activates the stress systems; this in turn may augment the generation of pro-inflammatory cytokines, stimulate pro-inflammatory immune cells, and alter the balance between the pro- and anti-inflammatory immune response. As a result, it is suggested that stress pathways may augment neuronal damage and loss after SCI. Considering these potential detrimental effects of stress after SCI, we hypothesized that inhibition of stress pathways immediately after SCI may offer protection from damage and improve recovery. To investigate the relevance of stress responses in SCI recovery, we investigated the effects of blocking three well-studied stress response axes in a mouse model of SCI. Propranolol, RU-486, and CP-99994 were administered to inhibit the sympathetic axis, the hypothalamus-pituitary-adrenal axis, and the neuropeptide axis, respectively. Surprisingly, assessing functional recovery by the Basso Mouse Scale revealed that RU-486 and CP-99994 did not affect functional outcome, indicating that these pathways are dispensable for neuroprotection or repair after SCI. Moreover, the beta-blocker propranolol worsened functional outcome in the mouse SCI model. In conclusion, immediate inhibition of three major stress axes has no beneficial effects on functional recovery after SCI in mice. These results suggest that injury-induced stress responses do not interfere with the healing process and hence, pharmacological targeting of stress responses is not a recommended treatment option for SCI. These findings are of great importance for other researchers to avoid unnecessary and potentially futile animal experiments.


Asunto(s)
Sistema Hipotálamo-Hipofisario/metabolismo , Neuropéptidos/metabolismo , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/metabolismo , Estrés Fisiológico/fisiología , Sistema Nervioso Simpático/metabolismo , Antagonistas Adrenérgicos beta/farmacología , Animales , Citocinas/antagonistas & inhibidores , Citocinas/metabolismo , Femenino , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Neuropéptidos/antagonistas & inhibidores , Recuperación de la Función/efectos de los fármacos , Traumatismos de la Médula Espinal/fisiopatología , Estrés Fisiológico/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos , Vértebras Torácicas/lesiones
18.
Sci Rep ; 9(1): 3715, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30842526

RESUMEN

Spinal cord injury (SCI) triggers the formation of a glial and fibrotic scar, which creates a major barrier for neuroregenerative processes. Previous findings indicate that mast cells (MCs) protect the spinal cord after mechanical damage by suppressing detrimental inflammatory processes via mouse mast cell protease 4 (mMCP4), a MC-specific chymase. In addition to these immunomodulatory properties, mMCP4 also plays an important role in tissue remodeling and extracellular matrix degradation. Therefore, we have investigated the effects of mMCP4 on the scarring response after SCI. We demonstrate that the decrease in locomotor performance in mMCP4-/- mice is correlated with excessive scar formation at the lesion. The expression of axon-growth inhibitory chondroitin sulfate proteoglycans was dramatically increased in the perilesional area in mMCP4-/- mice compared to wild type mice. Moreover, the fibronectin-, laminin-, and collagen IV-positive scar was significantly enlarged in mMCP4-/- mice at the lesion center. A degradation assay revealed that mMCP4 directly cleaves collagen IV in vitro. On the gene expression level, neurocan and GFAP were significantly higher in the mMCP4-/- group at day 2 and day 28 after injury respectively. In contrast, the expression of fibronectin and collagen IV was reduced in mMCP4-/- mice compared to WT mice at day 7 after SCI. In conclusion, our data show that mMCP4 modulates scar development after SCI by altering the gene and protein expression patterns of key scar factors in vivo. Therefore, we suggest a new mechanism via which endogenous mMCP4 can improve recovery after SCI.


Asunto(s)
Cicatriz/metabolismo , Serina Endopeptidasas/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Animales , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Quimasas/metabolismo , Cicatriz/patología , Fibronectinas/metabolismo , Fibrosis , Proteína Ácida Fibrilar de la Glía/metabolismo , Mastocitos/metabolismo , Ratones , Regeneración Nerviosa/fisiología , Neuroglía/patología , Serina Endopeptidasas/genética , Serina Endopeptidasas/fisiología , Médula Espinal/metabolismo
19.
Exp Neurobiol ; 27(5): 437-452, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30429652

RESUMEN

After spinal cord injury (SCI), monocyte derived macrophages play a detrimental role. Histone deacetylases (HDACs) are central epigenetic regulators of macrophage-polarization. We hypothesized that HDAC3 inhibition suppresses the pro-inflammatory macrophage phenotype (M1), promotes the anti-inflammatory phenotype (M2) and improves functional recovery after SCI. Therefore, two inhibitors of HDAC3 were selected, namely scriptaid and RGFP966. The impact on macrophage polarization was studied by investigating the effect on gene and protein expression of selected M1 and M2 markers. We show that scriptaid differentially influences M1 and M2 markers. It increases CD86 and iNOS gene expression and decreases GPR18, CD38, FPR2 and Arg-1 gene expression as well as the production of IL-6 and NO. RGFP966 primarily increased the expression of the M2 markers Arg-1 and Ym1 and reduced the production of IL-6 (M1). RGFP966 and scriptaid reduced the formation of foamy macrophages. Finally, to investigate the impact of HDAC3 inhibition on functional recovery after SCI, we studied the effects of RGFP966 and scriptaid in an in vivo T-cut hemisection SCI model. Histological analyses were performed on spinal cord sections to determine lesion size and astrogliosis, demyelinated area and selected infiltrating immune cells. RGFP966 and scriptaid did not affect functional recovery or histopathological outcome after SCI. In conclusion, these results indicate that specific HDAC3 inhibition with RGFP966 promotes alternative activation of macrophages and reduces the formation of foamy macrophages, but does not lead to a better functional recovery after SCI.

20.
Neurosci Biobehav Rev ; 83: 298-312, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29107829

RESUMEN

Damage to the motor cortex induced by stroke or traumatic brain injury (TBI) can result in chronic motor deficits. For the development and improvement of therapies, animal models which possess symptoms comparable to the clinical population are used. However, the use of experimental animals raises valid ethical and methodological concerns. To decrease discomfort by experimental procedures and to increase the quality of results, non-invasive and sensitive rodent motor tests are needed. A broad variety of rodent motor tests are available to determine deficits after stroke or TBI. The current review describes and evaluates motor tests that fall into three categories: Tests to evaluate fine motor skills and grip strength, tests for gait and inter-limb coordination and neurological deficit scores. In this review, we share our thoughts on standardized data presentation to increase data comparability between studies. We also critically evaluate current methods and provide recommendations for choosing the best behavioral test for a new research line.


Asunto(s)
Extremidades/fisiología , Marcha/fisiología , Corteza Motora/fisiología , Destreza Motora/fisiología , Animales , Modelos Animales , Roedores/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA