ABSTRACT
OBJECTIVES: Oestrogen deficiency is a rare disease and leads inter alia to arthralgia and osteoporosis in men. The clinical relevance of aromatase to a functioning male metabolism has become evident since 1991, when cases of patients with oestrogen deficiency caused by aromatase mutation were first described. Only few cases are known so far, which will now be presented in a case report and review of the literature. METHODS: All available publications since the first description in 1991 dealing with loss-of-function aromatase mutation in men were summarised and our case report was added. RESULTS: The mutations that cause the aromatase protein to lose function leads to a rather heterogeneous clinical picture. It is, however, clear that oestrogens play a central role in male patients, especially in bone metabolism. Most frequently, tall stature, unclosed epiphyseal joints, and osteoporosis are detected in affected individuals as a consequence of the change in hormonal status. CONCLUSIONS: As low oestrogen is associated with arthralgia, patients with aromatase mutation may be referred to a rheumatologist. Despite aromatase deficiency being a rare disease, the study of the effects of oestrogen on male bone development provides important insights for endocrine bone regulation. It has been demonstrated that androgens alone are not sufficient for adequate skeletal development in males. The described effects of loss of oestrogens are known from the aromatase inhibitor therapy in breast cancer treatment. This work highlights the important role of oestrogens in individual health and disease in men. Molecular effects of oestrogens on bone metabolism are summarised.
Subject(s)
Aromatase , Osteoporosis , Humans , Male , Aromatase/genetics , Aromatase/metabolism , Rare Diseases , Estrogens , Osteoporosis/drug therapy , Osteoporosis/genetics , MutationABSTRACT
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.
Subject(s)
Gray Matter/metabolism , Neural Stem Cells/cytology , Oligodendroglia/metabolism , White Matter/metabolism , Adult Stem Cells/metabolism , Animals , Astrocytes/metabolism , Cell Differentiation/physiology , Cell Lineage/physiology , Mice, Inbred C57BL , Neuroglia/metabolism , RatsABSTRACT
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.
Subject(s)
Mesenchymal Stem Cells/cytology , Neural Stem Cells/cytology , Oligodendroglia/cytology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Adult Stem Cells/cytology , Animals , Cell Differentiation , Cells, Cultured , Culture Media, Conditioned/chemistry , Female , Mesenchymal Stem Cells/metabolism , Primary Cell Culture , Proteomics , Rats , Stem Cell TransplantationABSTRACT
Stem cell transplantation is a promising therapeutic strategy to enhance axonal regeneration after spinal cord injury. Unrestricted somatic stem cells (USSC) isolated from human umbilical cord blood is an attractive stem cell population available at GMP grade without any ethical concerns. It has been shown that USSC transplantation into acute injured rat spinal cords leads to axonal regrowth and significant locomotor recovery, yet lacking cell replacement. Instead, USSC secrete trophic factors enhancing neurite growth of primary cortical neurons in vitro. Here, we applied a functional secretome approach characterizing proteins secreted by USSC for the first time and validated candidate neurite growth promoting factors using primary cortical neurons in vitro. By mass spectrometric analysis and exhaustive bioinformatic interrogation we identified 1156 proteins representing the secretome of USSC. Using Gene Ontology we revealed that USSC secretome contains proteins involved in a number of relevant biological processes of nerve regeneration such as cell adhesion, cell motion, blood vessel formation, cytoskeleton organization and extracellular matrix organization. We found for instance that 31 well-known neurite growth promoting factors like, e.g. neuronal growth regulator 1, NDNF, SPARC, and PEDF span the whole abundance range of USSC secretome. By the means of primary cortical neurons in vitro assays we verified SPARC and PEDF as significantly involved in USSC mediated neurite growth and therewith underline their role in improved locomotor recovery after transplantation. From our data we are convinced that USSC are a valuable tool in regenerative medicine as USSC's secretome contains a comprehensive network of trophic factors supporting nerve regeneration not only by a single process but also maintained its regenerative phenotype by a multitude of relevant biological processes.
Subject(s)
Fetal Blood/cytology , Nerve Growth Factors/metabolism , Stem Cells/metabolism , Axons/physiology , Cells, Cultured , Humans , Neurons/metabolism , Phenotype , Regeneration , Stem Cell TransplantationABSTRACT
BACKGROUND: Pruritus is a cardinal symptom of atopic dermatitis, and an increased cutaneous sensory network is thought to contribute to pruritus. Although the immune cell-IL-31-neuron axis has been implicated in severe pruritus during atopic skin inflammation, IL-31's neuropoietic potential remains elusive. OBJECTIVE: We sought to analyze the IL-31-related transcriptome in sensory neurons and to investigate whether IL-31 promotes sensory nerve fiber outgrowth. METHODS: In vitro primary sensory neuron culture systems were subjected to whole-transcriptome sequencing, ingenuity pathway analysis, immunofluorescence, and nerve elongation, as well as branching assays after IL-31 stimulation. In vivo we investigated the cutaneous sensory neuronal network in wild-type, Il31-transgenic, and IL-31 pump-equipped mice. RESULTS: Transgenic Il31 overexpression and subcutaneously delivered IL-31 induced an increase in the cutaneous nerve fiber density in lesional skin in vivo. Transcriptional profiling of IL-31-activated dorsal root ganglia neurons revealed enrichment for genes promoting nervous system development and neuronal outgrowth and negatively regulating cell death. Moreover, the growth cones of primary small-diameter dorsal root ganglia neurons showed abundant IL-31 receptor α expression. Indeed, IL-31 selectively promoted nerve fiber extension only in small-diameter neurons. Signal transducer and activator of transcription 3 phosphorylation mediated IL-31-induced neuronal outgrowth, and pharmacologic inhibition of signal transducer and activator of transcription 3 completely abolished this effect. In contrast, transient receptor potential cation channel vanilloid subtype 1 channels were dispensable for IL-31-induced neuronal sprouting. CONCLUSIONS: The pruritus- and TH2-associated novel cytokine IL-31 induces a distinct transcriptional program in sensory neurons, leading to nerve elongation and branching both in vitro and in vivo. This finding might help us understand the clinical observation that patients with atopic dermatitis experience increased sensitivity to minimal stimuli inducing sustained itch.
Subject(s)
Interleukins/metabolism , Pruritus/immunology , Pruritus/metabolism , Sensory Receptor Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , Cluster Analysis , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Gene Expression , Gene Expression Profiling , Humans , Interleukins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Nerve Fibers/metabolism , Phosphorylation , Pruritus/genetics , STAT3 Transcription Factor/metabolism , Skin/immunology , Skin/innervation , Skin/metabolismABSTRACT
A lesion to the rat rubrospinal tract is a model for traumatic spinal cord lesions and results in atrophy of the red nucleus neurons, axonal dieback, and locomotor deficits. In this study, we used adeno-associated virus (AAV)-mediated over-expression of BAG1 and ROCK2-shRNA in the red nucleus to trace [by co-expression of enhanced green fluorescent protein (EGFP)] and treat the rubrospinal tract after unilateral dorsal hemisection. We investigated the effects of targeted gene therapy on neuronal survival, axonal sprouting of the rubrospinal tract, and motor recovery 12 weeks after unilateral dorsal hemisection at Th8 in rats. In addition to the evaluation of BAG1 and ROCK2 as therapeutic targets in spinal cord injury, we aimed to demonstrate the feasibility and the limits of an AAV-mediated protein over-expression versus AAV.shRNA-mediated down-regulation in this traumatic CNS lesion model. Our results demonstrate that BAG1 and ROCK2-shRNA both promote neuronal survival of red nucleus neurons and enhance axonal sprouting proximal to the lesion.
Subject(s)
DNA-Binding Proteins/biosynthesis , Nerve Regeneration/physiology , Neurons/pathology , Spinal Cord Injuries/pathology , Transcription Factors/biosynthesis , rho-Associated Kinases/biosynthesis , Animals , Axons , Base Sequence , Blotting, Western , Cell Survival , DNA-Binding Proteins/genetics , Dependovirus , Disease Models, Animal , Female , Genetic Therapy/methods , Genetic Vectors , Immunohistochemistry , Molecular Sequence Data , RNA, Small Interfering , Rats , Rats, Wistar , Recovery of Function , Red Nucleus/pathology , Transcription Factors/genetics , rho-Associated Kinases/geneticsABSTRACT
Growth/differentiation factor-15 (GDF-15) is a distant member of the transforming growth factor beta (TGF-ß) superfamily. It is widely distributed in the nervous system, where it has been shown to play an important role in neuronal maintenance. The present study investigates the role of endogenous GDF-15 in sciatic nerve (SN) lesions using wild-type (WT) and GDF-15 knock-out (KO) mice. SN of 5-6-month-old mice were crushed or transected. Dorsal root ganglia (DRG) and nerve tissue were analyzed at different time points from 6 h to 9 weeks post-lesion. Both crush and transection induced GDF-15 mRNA and protein in the distal portion of the nerve, with a peak at day 7. DRG neuron death did not significantly differ between the genotypes; similarly, remyelination of regenerating axons was not affected by the genotype. Alternative macrophage activation and macrophage recruitment were more pronounced in the KO nerve. Protrusion speed of axons was similar in the two genotypes but WT axons showed better maturation, as indicated by larger caliber at 9 weeks. Furthermore, the regenerated WT nerve showed better performance in the electromyography test, indicating better functional recovery. We conclude that endogenous GDF-15 is beneficial for axon regeneration following SN crush.
Subject(s)
Axons/metabolism , Ganglia, Spinal/metabolism , Growth Differentiation Factor 15/metabolism , Nerve Regeneration/physiology , Sciatic Nerve/metabolism , Animals , Mice, Inbred C57BL , Mice, Transgenic , Nerve Crush/methods , Nerve Regeneration/genetics , Transforming Growth Factor beta/metabolismABSTRACT
We identified a suitable biomatrix that improved axon regeneration and functional outcome after partial (moderate) and complete (severe) chronic spinal cord injury (SCI) in rat. Five weeks after dorsal thoracic hemisection injury the lesion scar was resected via aspiration and the resulting cavity was filled with different biopolymers such as Matrigel™, alginate-hydrogel and polyethylene glycol 600 (PEG) all of which have not previously been used as sole graft-materials in chronic SCI. Immunohistological staining revealed marked differences between these compounds regarding axon regeneration, invasion/elongation of astrocytes, fibroblasts, endothelial and Schwann cells, revascularization, and collagen deposition. According to axon regeneration-supporting effects, the biopolymers could be ranked in the order PEG>>alginate-hydrogel>Matrigel™. Even after complete chronic transection, the PEG-bridge allowed long-distance axon regeneration through the grafted area and for, at least, 1cm beyond the lesion/graft border. As revealed by electron microscopy, bundles of regenerating axons within the matrix area received myelin ensheathment from Schwann cells. The beneficial effects of PEG-implantation into the resection-cavity were accompanied by long-lasting significant locomotor improvement over a period of 8months. Following complete spinal re-transection at the rostral border of the PEG-graft the locomotor recovery was aborted, suggesting a functional role of regenerated axons in the initial locomotor improvement. In conclusion, scar resection and subsequent implantation of PEG into the generated cavity leads to tissue recovery, axon regeneration, myelination and functional improvement that have not been achieved before in severe chronic SCI.
Subject(s)
Axons/ultrastructure , Cicatrix/surgery , Nerve Regeneration , Polyethylene Glycols/therapeutic use , Spinal Cord Injuries/surgery , Animals , Female , Myelin Sheath/pathology , Rats , Rats, Wistar , Recovery of Function , Schwann Cells/pathologyABSTRACT
Stem cell therapy is a potential treatment for spinal cord injury and different stem cell types have been grafted into animal models and humans suffering from spinal trauma. Due to inconsistent results, it is still an important and clinically relevant question which stem cell type will prove to be therapeutically effective. Thus far, stem cells of human sources grafted into spinal cord mostly included barely defined heterogeneous mesenchymal stem cell populations derived from bone marrow or umbilical cord blood. Here, we have transplanted a well-defined unrestricted somatic stem cell isolated from human umbilical cord blood into an acute traumatic spinal cord injury of adult immune suppressed rat. Grafting of unrestricted somatic stem cells into the vicinity of a dorsal hemisection injury at thoracic level eight resulted in hepatocyte growth factor-directed migration and accumulation within the lesion area, reduction in lesion size and augmented tissue sparing, enhanced axon regrowth and significant functional locomotor improvement as revealed by three behavioural tasks (open field Basso-Beattie-Bresnahan locomotor score, horizontal ladder walking test and CatWalk gait analysis). To accomplish the beneficial effects, neither neural differentiation nor long-lasting persistence of the grafted human stem cells appears to be required. The secretion of neurite outgrowth-promoting factors in vitro further suggests a paracrine function of unrestricted somatic stem cells in spinal cord injury. Given the highly supportive functional characteristics in spinal cord injury, production in virtually unlimited quantities at GMP grade and lack of ethical concerns, unrestricted somatic stem cells appear to be a highly suitable human stem cell source for clinical application in central nervous system injuries.
Subject(s)
Cord Blood Stem Cell Transplantation , Recovery of Function/physiology , Spinal Cord Injuries/therapy , Animals , Axons/physiology , Cells, Cultured , Female , Humans , Motor Activity/physiology , Rats , Rats, Wistar , Spinal Cord Injuries/physiopathologyABSTRACT
The paradigm of evidence-based medicine requires that medical decisions are made on the basis of the best available knowledge published in the literature. Existing evidence is often summarized in the form of systematic reviews and/or meta-reviews and is rarely available in a structured form. Manual compilation and aggregation is costly, and conducting a systematic review represents a high effort. The need to aggregate evidence arises not only in the context of clinical trials, but is also important in the context of pre-clinical animal studies. In this context, evidence extraction is important to support translation of the most promising pre-clinical therapies into clinical trials or to optimize clinical trial design. Aiming at developing methods that facilitate the task of aggregating evidence published in pre-clinical studies, in this paper a new system is presented that automatically extracts structured knowledge from such publications and stores it in a so-called domain knowledge graph. The approach follows the paradigm of model-complete text comprehension by relying on guidance from a domain ontology creating a deep relational data-structure that reflects the main concepts, protocol, and key findings of studies. Focusing on the domain of spinal cord injuries, a single outcome of a pre-clinical study is described by up to 103 outcome parameters. Since the problem of extracting all these variables together is intractable, we propose a hierarchical architecture that incrementally predicts semantic sub-structures according to a given data model in a bottom-up fashion. At the heart of our approach is a statistical inference method that relies on conditional random fields to infer the most likely instance of the domain model given the text of a scientific publication as input. This approach allows modeling dependencies between the different variables describing a study in a semi-joint fashion. We present a comprehensive evaluation of our system to understand the extent to which our system can capture a study in the depth required to enable the generation of new knowledge. We conclude the article with a brief description of some applications of the populated knowledge graph and show the potential implications of our work for supporting evidence-based medicine.
Subject(s)
Comprehension , Spinal Cord Injuries , Animals , Pattern Recognition, Automated , Systematic Reviews as Topic , Evidence-Based MedicineABSTRACT
Recruitment of inflammatory cells is known to drive the secondary damage cascades that are common to injuries of the central nervous system (CNS). Cell activation and infiltration to the injury site is orchestrated by changes in the expression of chemokines, the chemoattractive cytokines. Reducing the numbers of recruited inflammatory cells by the blocking of the action of chemokines has turned out be a promising approach to diminish neuroinflammation and to improve tissue preservation and neovascularization. In addition, several chemokines have been shown to be essential for stem/progenitor cell attraction, their survival, differentiation and cytokine production. Thus, chemokines might indirectly participate in remyelination, neovascularization and neuroprotection, which are important prerequisites for CNS repair after trauma. Moreover, CXCL12 promotes neurite outgrowth in the presence of growth inhibitory CNS myelin and enhances axonal sprouting after spinal cord injury (SCI). Here, we review current knowledge about the exciting functions of chemokines in CNS trauma, including SCI, traumatic brain injury and stroke. We identify common principles of chemokine action and discuss the potentials and challenges of therapeutic interventions with chemokines.
Subject(s)
Central Nervous System/immunology , Central Nervous System/injuries , Chemokines/metabolism , Wound Healing , Central Nervous System/pathology , Humans , Receptors, Chemokine/metabolism , Signal Transduction/immunologyABSTRACT
Spinal cord injury (SCI) is a rare condition, which even after decades of research, to date still presents an incurable condition with a complex symptomatology. An SCI can result in paralysis, pain, loss of sensation, bladder and sexual dysfunction, and muscle degeneration, to name but a few. The large number of publications makes it difficult to keep track of current progress in the field and of the many treatment options that have been suggested and are being proposed with increasing frequency. Scientific databases with user-oriented search options will offer possible solutions, but they are still mostly in the development phase. In this meta-analysis, we summarize and narrow down SCI therapeutic approaches applied in pre-clinical and clinical research. Statistical analyses of treatment clusters-assorted after counting annual publication numbers in PubMed and ClinicalTrials.gov databases-were performed to allow the comparison of research foci and of their translation efficacy into clinical therapy. Using the example of SCI research, our findings demonstrate the challenges that come with the accelerating research progress-an issue that many research fields are faced with today. The analyses point out similarities and differences in the prioritization of SCI research in pre-clinical versus clinical therapy strategies. Moreover, the results demonstrate the rapidly growing importance of modern (bio-)engineering technologies.
Subject(s)
Spinal Cord Injuries , Databases, Factual , Humans , Spinal Cord , Spinal Cord Injuries/therapy , Urinary BladderABSTRACT
Hydrophobically modified associating polymers could be effective drag-reducing agents containing weak "links" which after degradation can reform, protecting the polymer backbone from fast scission. Previous studies using hydrophobically modified polymers in drag reduction applications used polymers with M w ≥ 1000 kg/mol. Homopolymers of this high M w already show significant drag reduction (DR), and the contribution of macromolecular associations on DR remained unclear. We synthesized associating poly(acrylamide-co-styrene) copolymers with M w ≤ 1000 kg/mol and various hydrophobic moiety content. Their DR effectiveness in turbulent flow was studied using a pilot-scale pipe flow facility and a rotating "disc" apparatus. We show that hydrophobically modified copolymers with M w ≈ 1000 kg/mol increase DR in pipe flow by a factor of â¼2 compared to the unmodified polyacrylamide of similar M w albeit at low DR level. Moreover, we discuss challenges encountered when using hydrophobically modified polymers synthesized via micellar polymerization.
ABSTRACT
Axonal damage is an early step in traumatic and neurodegenerative disorders of the central nervous system (CNS). Damaged axons are not able to regenerate sufficiently in the adult mammalian CNS, leading to permanent neurological deficits. Recently, we showed that inhibition of the autophagic protein ULK1 promotes neuroprotection in different models of neurodegeneration. Moreover, we demonstrated previously that axonal protection improves regeneration of lesioned axons. However, whether axonal protection mediated by ULK1 inhibition could also improve axonal regeneration is unknown. Here, we used an adeno-associated viral (AAV) vector to express a dominant-negative form of ULK1 (AAV.ULK1.DN) and investigated its effects on axonal regeneration in the CNS. We show that AAV.ULK1.DN fosters axonal regeneration and enhances neurite outgrowth in vitro. In addition, AAV.ULK1.DN increases neuronal survival and enhances axonal regeneration after optic nerve lesion, and promotes long-term axonal protection after spinal cord injury (SCI) in vivo. Interestingly, AAV.ULK1.DN also increases serotonergic and dopaminergic axon sprouting after SCI. Mechanistically, AAV.ULK1.DN leads to increased ERK1 activation and reduced expression of RhoA and ROCK2. Our findings outline ULK1 as a key regulator of axonal degeneration and regeneration, and define ULK1 as a promising target to promote neuroprotection and regeneration in the CNS.
Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Axons/metabolism , Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors , Nerve Regeneration , Optic Nerve Injuries/therapy , Optic Nerve/metabolism , Spinal Cord Injuries/therapy , Spinal Cord/metabolism , Animals , Autophagy-Related Protein-1 Homolog/genetics , Axons/pathology , Cells, Cultured , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Down-Regulation , Female , Mitogen-Activated Protein Kinase 3/metabolism , Neuronal Outgrowth , Optic Nerve/pathology , Optic Nerve Injuries/genetics , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Rats, Wistar , Serotonergic Neurons/metabolism , Serotonergic Neurons/pathology , Spinal Cord/pathology , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Time Factors , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolismABSTRACT
Impaired axonal regeneration is a common observation after central nervous system (CNS) injury. The stromal cell-derived factor-1, SDF-1/CXCL12, has previously been shown to promote axonal growth in the presence of potent chemorepellent molecules known to be important in nervous system development. Here, we report that treatment with SDF-1alpha is sufficient to overcome neurite outgrowth inhibition mediated by CNS myelin towards cultured postnatal dorsal root ganglion neurons. While we found both cognate SDF-1 receptors, CXCR4 and CXCR7/RDC1, to be coexpressed on myelin-sensitive dorsal root ganglion neurons, the distinct expression pattern of CXCR4 on growth cones and branching points of neurites suggests a function of this receptor in chemokine-mediated growth promotion and/or arborization. These in vitro findings were further corroborated as local intrathecal infusion of SDF-1 into spinal cord injury following thoracic dorsal hemisection resulted in enhanced sprouting of corticospinal tract axons into white and grey matter. Our findings indicate that SDF-1 receptor activation might constitute a novel therapeutic approach to promote axonal growth in the injured CNS.
Subject(s)
Cell Culture Techniques , Central Nervous System/metabolism , Chemokine CXCL12/metabolism , Myelin Sheath/metabolism , Neurites/physiology , Animals , Cells, Cultured , Chemokine CXCL12/genetics , Chemokine CXCL12/pharmacology , Female , Ganglia, Spinal/cytology , Nerve Regeneration/physiology , Neurites/drug effects , Neurites/ultrastructure , Pyramidal Tracts/cytology , Pyramidal Tracts/drug effects , Pyramidal Tracts/metabolism , Pyramidal Tracts/pathology , Rats , Rats, Wistar , Receptors, CXCR/genetics , Receptors, CXCR/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Spinal Cord/anatomy & histology , Spinal Cord/drug effects , Spinal Cord/surgeryABSTRACT
At present the pathogenesis of CMT1A neuropathy, caused by the overexpression of PMP22, has not yet been entirely understood. The PMP22-overexpressing C61 mutant mouse is a suitable animal model, which mimics the human CMT1A disorder. We observed that myelin gene expression in the sciatic nerve of the C61 mouse was up-regulated at postnatal day 4 to 7 (P4-P7). When investigating the morphology of peripheral nerves in C61 and wildtype mice at early stages of postnatal development, hypermyelination could be detected in the femoral quadriceps and sciatic nerve of transgenic animals at postnatal day 7 (P7). In order to identify genes, other than Pmp22, that are modulated in sciatic nerve of P7 transgenic mice, we applied microarray technology. Amongst the regulated genes, the gene encoding the alpha-chemokine CXCL14 was most prominently up-regulated. We report that Cxcl14 was expressed exclusively by Schwann cells of the sciatic nerve, as well as by cultured Schwann cells triggered to differentiate. Furthermore, in cultured Schwann cells CXCL14 modulated the expression of myelin genes and altered cell proliferation. Our findings demonstrate that early overexpression of PMP22, in a mouse model of CMT1A, results in a strong up-regulation of CXCL14, which seems to play a novel regulatory role in Schwann cell differentiation.
Subject(s)
Charcot-Marie-Tooth Disease/genetics , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , Myelin Basic Protein/genetics , Myelin P0 Protein/genetics , Schwann Cells/metabolism , Sciatic Nerve/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Charcot-Marie-Tooth Disease/metabolism , Disease Models, Animal , Gene Expression , Mice , Mice, Transgenic , Myelin Basic Protein/metabolism , Myelin P0 Protein/metabolism , Myelin Proteins/genetics , Nerve Fibers, Myelinated/metabolism , Oligonucleotide Array Sequence Analysis , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Schwann Cells/cytology , Schwann Cells/ultrastructure , Up-RegulationABSTRACT
We analysed the effect of scar-suppressing treatment (anti-scarring treatment; AST) on augmenting axonal regeneration of various neuronal populations following spinal cord injury (SCI) in adult rat. AST included local iron chelator (2,2'-dipyridine-5,5'-dicarboxylic acid) injection and 8-bromo-cyclic adenosine monophosphate application to the lesion core. In previous studies, this treatment promoted long-distance regeneration of cut corticospinal tract axons, neuroprotection of projecting cortical neurons and functional improvement of treated rats [N. Klapka et al. (2005)Eur. J. Neurosci., 22, 3047-3058]. Treatment yielded significantly enhanced regrowth of descending serotonergic (5-HT), catecholaminergic (tyrosine hydroxylase; TH), corticospinal and rubrospinal axons into the lesion zone, as assessed by anterograde tracing and immunohistochemistry followed by quantification of axon profiles at 5 and 12 weeks post-injury. In addition, the determination of axons crossing the proximal borderline from uninjured tissue into fibrous scar area revealed a significant AST-promoted increase of intersecting fibres for 5-HT, TH and calcitonin gene-related peptide containing ascending sensory fibres. For a prolonged time period after lesion, the delayed (secondary) scar developing in treated rats is significantly more permeable for all analysed axon tracts than the initial (primary) scar forming in injured control animals lacking treatment. Furthermore, enhanced outgrowth of descending axons from fibrous scar into distal healthy spinal tissue was achieved in treated animals, and is in line with previous functional studies [S. Hermanns et al. (2001) Restor. Neurol. Neurosci., 19,139-148; N. Klapka et al. (2005)Eur. J. Neurosci., 22, 3047-3058]. Our findings indicate that AST exerts a prolonged beneficial effect on fibrous scarring allowing enhanced axonal regrowth of different fibre tracts in SCI regardless of their distinct regenerative demands.
Subject(s)
Axons/drug effects , Cicatrix/etiology , Cicatrix/prevention & control , Iron Chelating Agents/pharmacology , Nerve Regeneration/drug effects , Spinal Cord Injuries/complications , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Biotin/analogs & derivatives , Biotin/metabolism , Calcitonin Gene-Related Peptide/metabolism , Cicatrix/pathology , Cicatrix/physiopathology , Dextrans/metabolism , Dicarboxylic Acids/pharmacology , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glial Fibrillary Acidic Protein/metabolism , Nerve Regeneration/physiology , Pyramidal Tracts/pathology , Rats , Rats, Wistar , Serotonin/metabolism , Spinal Cord Injuries/drug therapy , Time Factors , Tyrosine 3-Monooxygenase/metabolismABSTRACT
Glucose-dependent insulinotropic polypeptide (GIP) was initially described to be rapidly regulated by endocrine cells in response to nutrient ingestion, with stimulatory effects on insulin synthesis and release. Previously, we demonstrated a significant up-regulation of GIP mRNA in the rat subiculum after fornix injury. To gain more insight into the lesion-induced expression of GIP and its receptor (GIPR), expression profiles of the mRNAs were studied after rat sciatic nerve crush injury in 1) affected lumbar dorsal root ganglia (DRG), 2) spinal cord segments, and 3) proximal and distal nerve fragments by means of quantitative RT-PCR. Our results clearly identified lesion-induced as well as tissue type-specific mRNA regulation of GIP and its receptor. Furthermore, comprehensive immunohistochemical stainings not only confirmed and exceeded the previous observation of neuronal GIP expression but also revealed corresponding GIPR expression, implying putative modulatory functions of GIP/GIPR signaling in adult neurons. In complement, we also observed expression of GIP and its receptor in myelinating Schwann cells and oligodendrocytes. Polarized localization of GIPR in the abaxonal Schwann cell membranes, plasma membrane-associated GIPR expression of satellite cells, and ependymal GIPR expression strongly suggests complex cell type-specific functions of GIP and GIPR in the adult nervous system that are presumably mediated by autocrine and paracrine interactions, respectively. Notably, in vivo analyses with GIPR-deficient mice suggest a critical role of GIP/GIPR signal transduction in promoting spontaneous recovery after nerve crush, insofar as traumatic injury of GIPR-deficient mouse sciatic nerve revealed impaired axonal regeneration compared with wild-type mice.
Subject(s)
Ganglia, Spinal/metabolism , Gastric Inhibitory Polypeptide/genetics , Growth Cones/metabolism , Nerve Regeneration/physiology , Receptors, Gastrointestinal Hormone/genetics , Sciatic Neuropathy/metabolism , Animals , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Ganglia, Spinal/physiopathology , Gastric Inhibitory Polypeptide/metabolism , Gene Expression Regulation/genetics , Growth Cones/ultrastructure , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Gastrointestinal Hormone/metabolism , Schwann Cells/cytology , Schwann Cells/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Sciatic Nerve/physiopathology , Sciatic Neuropathy/physiopathology , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism , Spinal Cord/metabolism , Spinal Cord/physiopathologyABSTRACT
Recently, it has been shown that human unrestricted somatic stem cells (USSCs) from umbilical cord blood represent pluripotent, neonatal, nonhematopoietic stem cells with the potential to differentiate into the neural lineage. However, molecular and functional characterization of the neural phenotype and evaluation of the degree of maturity of the resulting cells are still lacking. In this study, we addressed the question of neuronal differentiation and maturation induced by a defined composition of growth and differentiation factors (XXL medium). We demonstrated the expression of different neuronal markers and their enrichment in USSC cultures during XXL medium incubation. Furthermore, we showed enrichment of USSCs expressing tyrosine hydroxylase (TH), an enzyme specific for dopaminergic neurons and other catecholamine-producing neurons, accompanied by induction of Nurr1, a factor regulating dopaminergic neurogenesis. The functionality of USSCs has been analyzed by patch-clamp recordings and high-performance liquid chromatography (HPLC). Voltage-gated sodium-channels could be identified in laminin-predifferentiated USSCs. In addition, HPLC analysis revealed synthesis and release of the neurotransmitter dopamine by USSC-derived cells, thus correlating well with the detection of TH transcripts and protein. This study provides novel insight into the potential of unrestricted somatic stem cells from human umbilical cord blood to acquire a neuronal phenotype and function.