Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 696
Filter
1.
Nat Rev Neurosci ; 23(1): 23-34, 2022 01.
Article in English | MEDLINE | ID: mdl-34671105

ABSTRACT

Recent transcriptomic, histological and functional studies have begun to shine light on the fibroblasts present in the meninges, choroid plexus and perivascular spaces of the brain and spinal cord. Although the origins and functions of CNS fibroblasts are still being described, it is clear that they represent a distinct cell population, or populations, that have likely been confused with other cell types on the basis of the expression of overlapping cellular markers. Recent work has revealed that fibroblasts play crucial roles in fibrotic scar formation in the CNS after injury and inflammation, which have also been attributed to other perivascular cell types such as pericytes and vascular smooth muscle cells. In this Review, we describe the current knowledge of the location and identity of CNS perivascular cell types, with a particular focus on CNS fibroblasts, including their origin, subtypes, roles in health and disease, and future areas for study.


Subject(s)
Central Nervous System Diseases/physiopathology , Central Nervous System/injuries , Central Nervous System/physiology , Fibroblasts/physiology , Animals , Central Nervous System/cytology , Humans
2.
Brain Behav Immun ; 122: 583-595, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39222725

ABSTRACT

Damage-associated molecular patterns (DAMPs) are endogenous molecules released in tissues upon cellular damage and necrosis, acting to initiate sterile inflammation. Constitutive DAMPs (cDAMPs) have the particularity to be present within the intracellular compartments of healthy cells, where they exert diverse functions such as regulation of gene expression and cellular homeostasis. However, after injury to the central nervous system (CNS), cDAMPs are rapidly released by stressed, damaged or dying neuronal, glial and endothelial cells, and can trigger inflammation without undergoing structural modifications. Several cDAMPs have been described in the injured CNS, such as interleukin (IL)-1α, IL-33, nucleotides (e.g. ATP), and high-mobility group box protein 1. Once in the extracellular milieu, these molecules are recognized by the remaining surviving cells through specific DAMP-sensing receptors, thereby inducing a cascade of molecular events leading to the production and release of proinflammatory cytokines and chemokines, as well as cell adhesion molecules. The ensuing immune response is necessary to eliminate cellular debris caused by the injury, allowing for damage containment. However, seeing as some molecules associated with the inflammatory response are toxic to surviving resident CNS cells, secondary damage occurs, aggravating injury and exacerbating neurological and behavioral deficits. Thus, a better understanding of these cDAMPs, as well as their receptors and downstream signaling pathways, could lead to identification of novel therapeutic targets for treating CNS injuries such as SCI, TBI, and stroke. In this review, we summarize the recent literature on cDAMPs, their specific functions, and the therapeutic potential of interfering with cDAMPs or their signaling pathways.


Subject(s)
Alarmins , Central Nervous System , Humans , Alarmins/metabolism , Animals , Central Nervous System/metabolism , Central Nervous System/injuries , Inflammation/metabolism , Cytokines/metabolism , HMGB1 Protein/metabolism , Interleukin-33/metabolism , Interleukin-1alpha/metabolism , Signal Transduction/physiology
3.
Cell Biochem Funct ; 42(4): e4056, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38812104

ABSTRACT

Previous studies suggested that central nervous system injury is often accompanied by the activation of Toll-like receptor 4/NF-κB pathway, which leads to the upregulation of proapoptotic gene expression, causes mitochondrial oxidative stress, and further aggravates the inflammatory response to induce cell apoptosis. Subsequent studies have shown that NF-κB and IκBα can directly act on mitochondria. Therefore, elucidation of the specific mechanisms of NF-κB and IκBα in mitochondria may help to discover new therapeutic targets for central nervous system injury. Recent studies have suggested that NF-κB (especially RelA) in mitochondria can inhibit mitochondrial respiration or DNA expression, leading to mitochondrial dysfunction. IκBα silencing will cause reactive oxygen species storm and initiate the mitochondrial apoptosis pathway. Other research results suggest that RelA can regulate mitochondrial respiration and energy metabolism balance by interacting with p53 and STAT3, thus initiating the mitochondrial protection mechanism. IκBα can also inhibit apoptosis in mitochondria by interacting with VDAC1 and other molecules. Regulating the biological role of NF-κB signaling pathway in mitochondria by targeting key proteins such as p53, STAT3, and VDAC1 may help maintain the balance of mitochondrial respiration and energy metabolism, thereby protecting nerve cells and reducing inflammatory storms and death caused by ischemia and hypoxia.


Subject(s)
Mitochondria , NF-kappa B , Signal Transduction , Toll-Like Receptor 4 , Humans , Mitochondria/metabolism , Toll-Like Receptor 4/metabolism , NF-kappa B/metabolism , Animals , Apoptosis , Central Nervous System/metabolism , Central Nervous System/pathology , Central Nervous System/injuries
4.
Semin Cell Dev Biol ; 89: 118-124, 2019 05.
Article in English | MEDLINE | ID: mdl-30287388

ABSTRACT

The tenascin family of glycoproteins comprises four members in vertebrates, of which tenascin-C (Tnc) and tenascin-R (Tnr) are particularly important in the context of lesions in the central nervous system (CNS). Tnc is expressed in the developing CNS, before it is down-regulated and mainly restricted to the adult neural stem cell niches. It regulates numerous processes including differentiation, adhesion, migration and neurite outgrowth. These aspects are critical in the developing organism, but also after damage. Interestingly, Tnc is indeed re-expressed in the injured CNS. Additionally, Tnc is an activator of the immune response, another important aspect after lesion. Tnr is part of perineuronal nets, a specialized form of extracellular matrix that enwraps subtypes of neurons and limits synaptic plasticity. We summarize the role of tenascins in the context of stem cell niches, barrier formation, synaptic plasticity and immune response in the damaged mammalian CNS.


Subject(s)
Central Nervous System/metabolism , Nervous System Diseases/genetics , Tenascin/genetics , Animals , Cell Adhesion/genetics , Cell Differentiation/genetics , Cell Movement/genetics , Central Nervous System/injuries , Central Nervous System/pathology , Extracellular Matrix/genetics , Humans , Nervous System Diseases/pathology , Neural Stem Cells/metabolism , Neuronal Plasticity/genetics , Neurons/metabolism , Neurons/pathology , Stem Cell Niche/genetics , Vertebrates/genetics
5.
PLoS Biol ; 16(10): e2005264, 2018 10.
Article in English | MEDLINE | ID: mdl-30332405

ABSTRACT

Infiltrating monocyte-derived macrophages (MDMs) and resident microglia dominate central nervous system (CNS) injury sites. Differential roles for these cell populations after injury are beginning to be uncovered. Here, we show evidence that MDMs and microglia directly communicate with one another and differentially modulate each other's functions. Importantly, microglia-mediated phagocytosis and inflammation are suppressed by infiltrating macrophages. In the context of spinal cord injury (SCI), preventing such communication increases microglial activation and worsens functional recovery. We suggest that macrophages entering the CNS provide a regulatory mechanism that controls acute and long-term microglia-mediated inflammation, which may drive damage in a variety of CNS conditions.


Subject(s)
Macrophages/physiology , Microglia/physiology , Spinal Cord Injuries/immunology , Adult , Animals , Central Nervous System/immunology , Central Nervous System/injuries , Female , Healthy Volunteers , Humans , Inflammation/immunology , Leukocyte Count , Male , Mice , Mice, Inbred C57BL , Microglia/immunology , Monocytes , Phagocytosis , Recovery of Function
6.
Acta Biochim Biophys Sin (Shanghai) ; 53(1): 1-9, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33258872

ABSTRACT

Thousands of nerve injuries occur in the world each year. Axon regeneration is a very critical process for the restoration of the injured nervous system's function. However, the precise molecular mechanism or signaling cascades that control axon regeneration are not clearly understood, especially in mammals. Therefore, there is almost no ideal treatment method to repair the nervous system's injury until now. Mammalian axonal regeneration requires multiple signaling pathways to coordinately regulate gene expression in soma and assembly of the cytoskeleton protein in the growth cone. A better understanding of their molecular mechanisms, such as axon regeneration regulatory signaling cascades, will be helpful in developing new treatment strategies for promoting axon regeneration. In this review, we mainly focus on describing these regeneration-associated signaling cascades, which regulate axon regeneration.


Subject(s)
Axons/metabolism , Nerve Regeneration/genetics , Animals , Cell Body/metabolism , Central Nervous System/injuries , Growth Cones/metabolism , Invertebrates , Mammals , Signal Transduction
7.
Vet Clin North Am Equine Pract ; 37(2): 245-258, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34119403

ABSTRACT

Acute central nervous system (CNS) trauma in the field is best approached by a systematic and thorough physical and neurologic examination that allows the practitioner to localize the brain or spinal cord injury. The skull and vertebral canal are complex 3-dimensional structures, and orthogonal radiographic views are necessary for an accurate diagnosis. Therapeutics aimed at decreasing pain, inflammation, and edema or increased intracranial pressure in the case of traumatic brain injury should be administered. Survival and return to athleticism can be achieved even in moderate-to-severe traumatic CNS injury with appropriate medical management.


Subject(s)
Horse Diseases/diagnosis , Spinal Cord Injuries/veterinary , Animals , Central Nervous System/injuries , Horse Diseases/therapy , Horses , Humans , Inflammation/veterinary , Pain/veterinary , Spinal Cord Injuries/diagnosis , Spinal Cord Injuries/therapy , Wounds and Injuries/veterinary
8.
J Cell Physiol ; 235(4): 3864-3873, 2020 04.
Article in English | MEDLINE | ID: mdl-31603252

ABSTRACT

Neuropathic pain correlates with a lesion or other dysfunction in the nervous system. Sphingosine-1-phosphate receptor 2 (S1P2) is expressed in the central nervous system and modulates synaptic plasticity. The present study aimed to investigate the role of S1P2 in neuropathic pain caused by chronic constriction injury (CCI). Sprague-Dawley rats were allocated into eight groups (n = 15 for each group): sham, CCI, CCI + green fluorescent protein, CCI + S1P2, CCI + Ctrl-short hairpin RNA (shRNA), CCI + S1P2 shRNA, CCI + S1P2 + CYM-5442, and CCI + S1P2 shRNA + CYM-5442. The CCI model was established via sciatic nerve ligation. S1P2 was overexpressed or knocked down by intrathecal injection of adeno-associated virus-S1P2 (AAV-S1P2) or AAV-S1P2 shRNA. The S1P1 agonist, CYM-5442 (1 mg/kg), was injected intraperitoneally after surgery. S1P2 expression, pain thresholds, apoptosis signaling, inflammation, and oxidative stress in rats were then examined. We found that sciatic nerve injury downregulated S1P2 expression in the spinal cords of rats. S1P2 overexpression enhanced pain thresholds. In contrast, S1P2 knockdown decreased pain thresholds in rats exposed to CCI. CCI and S1P2 silencing increased secretion of interleukin-1ß (IL-1ß), IL-6, and CCL2, whereas S1P2 overexpression decreased. S1P2 impeded CCI-induced reactive oxygen species (ROS) production and runt-related transcription factors 3 (RUNX3) downregulation, and S1P2 knockdown had the opposite effect. S1P2 overexpression suppressed Bax and active caspase 3 expression and promoted Bcl-2 expression, whereas loss of S1P2 reversed their expression. Additionally, S1P1 activation counteracted the effect of S1P2 on pain sensitivity. In conclusion, S1P2 is downregulated in CCI rats and may help modulate neuropathic pain via the ROS/RUNX3 pathway.


Subject(s)
Central Nervous System/injuries , Core Binding Factor Alpha 3 Subunit/genetics , Neuralgia/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Sphingosine-1-Phosphate Receptors/genetics , Animals , Central Nervous System/metabolism , Central Nervous System/pathology , Constriction, Pathologic/genetics , Disease Models, Animal , Gene Expression Regulation/genetics , Gene Silencing , Humans , Interleukin-1beta/genetics , Interleukin-6/genetics , Ligation , Neuralgia/pathology , RNA, Small Interfering/genetics , Rats , Reactive Oxygen Species/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Sciatic Neuropathy/genetics , Sciatic Neuropathy/pathology , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors
9.
Neurochem Res ; 45(3): 630-642, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31997102

ABSTRACT

Oligodendrocytes (OLs) generate myelin membranes for the rapid propagation of electrical signals along axons in the central nervous system (CNS) and provide metabolites to support axonal integrity and function. Differentiation of OLs from oligodendrocyte progenitor cells (OPCs) is orchestrated by a multitude of intrinsic and extrinsic factors in the CNS. Disruption of this process, or OL loss in the developing or adult brain, as observed in various neurological conditions including hypoxia/ischemia, stroke, and demyelination, results in axonal dystrophy, neuronal dysfunction, and severe neurological impairments. While much is known regarding the intrinsic regulatory signals required for OL lineage cell progression in development, studies from pathological conditions highlight the importance of the CNS environment and external signals in regulating OL genesis and maturation. Here, we review the recent findings in OL biology in the context of the CNS physiological and pathological conditions, focusing on extrinsic factors that facilitate OL development and regeneration.


Subject(s)
Central Nervous System Diseases/physiopathology , Central Nervous System/physiology , Oligodendroglia/cytology , Stem Cells/cytology , Animals , Cell Differentiation/physiology , Cell Lineage , Central Nervous System/injuries , Disease Progression , Humans , Myelin Sheath
10.
Exp Cell Res ; 376(1): 49-57, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30658092

ABSTRACT

Astrocyte activation is crucial for wound contraction and glial scar formation following central nervous system injury, but the mechanism by which activation leads to astrocyte contractility and matrix reorganization in the central nervous system (CNS) is unknown. Current means to measure cell traction forces within three-dimensional scaffolds are limited to analyzing individual or small groups of cells, within extracellular matrices, whereas gap junctions and other cell-cell adhesions connect astrocytes to form a functional syncytium within the glial scar. Here, we measure the viscoelastic properties of cell-seeded hydrogels to yield insight into the collective contractility of astrocytes as they exert tension on the surrounding matrix and change its bulk mechanical properties. Our results indicate that incorporation of the CNS matrix component hyaluronan into a collagen hydrogel increases expression of the intermediate filament protein GFAP and results in a higher shear storage modulus of the cell/matrix composite, establishing the correlation between astrocyte activation and increased cell contractility. The effects of thrombin and blebbistatin, known mediators of actomyosin-mediated contraction, verify that cell-matrix tension dictates the hydrogel mechanical properties. Viability assays indicate that increased cell traction exacerbates cell death at the center of the scaffold, and message level analysis reveals that cells in the hyaluronan-containing matrix have a ~ 3-fold increase in HIF-1α gene expression. Overall, these findings suggest that astrocyte activation not only increases cell traction, but may also contribute to hypoxia near sites of central nervous system injury.


Subject(s)
Central Nervous System/drug effects , Extracellular Matrix/drug effects , Hydrogels/pharmacology , Neuroglia/drug effects , Astrocytes/drug effects , Cell Culture Techniques , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Central Nervous System/injuries , Cicatrix/drug therapy , Cicatrix/pathology , Collagen/chemistry , Collagen/pharmacology , Cytosol/chemistry , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Hydrogels/chemistry , Neuroglia/pathology , Oxygen/metabolism , Rheology/methods , Viscoelastic Substances/chemistry , Viscoelastic Substances/pharmacology , Wound Healing/drug effects
11.
Curr Osteoporos Rep ; 18(6): 666-676, 2020 12.
Article in English | MEDLINE | ID: mdl-33085000

ABSTRACT

PURPOSE OF REVIEW: Neurogenic heterotopic ossification (NHO) is the abnormal formation of extra-skeletal bones in periarticular muscles after damage to the central nervous system (CNS) such as spinal cord injury (SCI), traumatic brain injury (TBI), stroke, or cerebral anoxia. The purpose of this review is to summarize recent developments in the understanding of NHO pathophysiology and pathogenesis. Recent animal models of NHO and recent findings investigating the communication between CNS injury, tissue inflammation, and upcoming NHO therapeutics are discussed. RECENT FINDINGS: Animal models of NHO following TBI or SCI have shown that NHO requires the combined effects of a severe CNS injury and soft tissue damage, in particular muscular inflammation and the infiltration of macrophages into damaged muscles plays a key role. In the context of a CNS injury, the inflammatory response to soft tissue damage is exaggerated and persistent with excessive signaling via substance P-, oncostatin M-, and TGF-ß1-mediated pathways. This review provides an overview of the known animal models and mechanisms of NHO and current therapeutic interventions for NHO patients. While some of the inflammatory mechanisms leading to NHO are common with other forms of traumatic and genetic heterotopic ossifications (HO), NHOs uniquely involve systemic changes in response to CNS injury. Future research into these CNS-mediated mechanisms is likely to reveal new targetable pathways to prevent NHO development in patients.


Subject(s)
Central Nervous System/injuries , Ossification, Heterotopic/etiology , Ossification, Heterotopic/physiopathology , Animals , Disease Models, Animal , Humans , Ossification, Heterotopic/therapy
12.
Int J Mol Sci ; 21(15)2020 Jul 30.
Article in English | MEDLINE | ID: mdl-32751444

ABSTRACT

Nogo-A, a glycoprotein expressed in oligodendrocytes and central nervous system myelin, inhibits regeneration after injury. Antibodies against Nogo-A neutralize this inhibitory activity, improve locomotor recovery in spinal cord-injured adult mammals, and promote regrowth/sprouting/saving of damaged axons beyond the lesion site. Nogo-A is also expressed by neurons. Complete ablation of Nogo-A in all cell types expressing it has been found to lead to recovery in some studies but not in others. Neuronal ablation of Nogo-A reduces axonal regrowth after injury. In view of these findings, we hypothesized that, in addition to neutralizing Nogo-A in oligodendrocytes and myelin, Nogo-A antibodies may act directly on neuronal Nogo-A to trigger neurite outgrowth and neuronal survival. Here, we show that polyclonal and monoclonal antibodies against Nogo-A enhance neurite growth and survival of cultured cerebellar granule neurons and increase expression of the neurite outgrowth-promoting L1 cell adhesion molecule and polysialic acid. Application of inhibitors of signal transducing molecules, such as c-src, c-fyn, protein kinase A, and casein kinase II reduce antibody-triggered neurite outgrowth. These observations indicate that the recovery-promoting functions of antibodies against Nogo-A may not only be due to neutralizing Nogo-A in oligodendrocytes and myelin, but also to their interactions with Nogo-A on neurons.


Subject(s)
Antibodies, Monoclonal/metabolism , Cell Survival , Myelin Proteins/metabolism , Neurites/metabolism , Neuronal Outgrowth , Oligodendroglia/metabolism , Peptide Fragments/metabolism , Animals , Cells, Cultured , Central Nervous System/injuries , Female , Male , Mice , Myelin Proteins/antagonists & inhibitors , Nerve Regeneration , Neurites/pathology , Oligodendroglia/pathology , Peptide Fragments/antagonists & inhibitors
13.
Int J Mol Sci ; 21(24)2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33333849

ABSTRACT

Neuropathic pain is characterized by spontaneous pain, pain sensations, and tactile allodynia. The pain sensory system normally functions under a fine balance between excitation and inhibition. Neuropathic pain arises when this balance is lost for some reason. In past reports, various mechanisms of neuropathic pain development have been reported, one of which is the downregulation of K+-Cl--cotransporter-2 (KCC2) expression. In fact, various neuropathic pain models indicate a decrease in KCC2 expression. This decrease in KCC2 expression is often due to a brain-derived neurotrophic factor that is released from microglia. However, a similar reaction has been reported in astrocytes, and it is unclear whether astrocytes or microglia are more important. This review discusses the hypothesis that astrocytes have a crucial influence on the alteration of KCC2 expression.


Subject(s)
Astrocytes/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Central Nervous System/metabolism , Neuralgia/metabolism , Signal Transduction/physiology , Symporters/metabolism , Animals , Astrocytes/enzymology , Central Nervous System/injuries , Cytokines/metabolism , Humans , Inflammation/metabolism , Matrix Metalloproteinases/metabolism , Neuralgia/enzymology , Receptor, trkB/metabolism , Wounds and Injuries/enzymology , Wounds and Injuries/metabolism , K Cl- Cotransporters
14.
Rev Med Suisse ; 16(692): 904-906, 2020 May 06.
Article in French | MEDLINE | ID: mdl-32374534

ABSTRACT

Spasticity is a common sign of central nervous system lesions and its management is difficult because it is usually associated with other symptoms of upper motoneuron syndrome (paresis, spastic dystonia, contractures, …). We propose an interprofessional evaluation, which demonstrates that a standardized evaluation, a common approach and a gait analysis improve the therapeutic decision.


La spasticité est très fréquente après une lésion du système nerveux central, et sa prise en charge demeure difficile, car elle se combine avec d'autres symptômes caractéristiques du syndrome du motoneurone supérieur (parésie, dystonie spastique, contractures…). Afin de faciliter le choix thérapeutique, nous présentons le modèle d'une évaluation interprofessionnelle, qui démontre qu'une évaluation standardisée, une approche commune et une analyse de la marche permettent une meilleure prise en charge thérapeutique.


Subject(s)
Central Nervous System/injuries , Central Nervous System/physiopathology , Muscle Spasticity/diagnosis , Muscle Spasticity/therapy , Gait Analysis , Humans , Muscle Spasticity/physiopathology
15.
Biomacromolecules ; 20(9): 3445-3452, 2019 09 09.
Article in English | MEDLINE | ID: mdl-31460746

ABSTRACT

Insufficient endogenous neural stem cell (NSC) migration to injury sites and incomplete replenishment of neurons complicates recovery following central nervous system (CNS) injury. Such insufficient migration can be addressed by delivering soluble chemotactic factors, such as stromal cell-derived factor 1-α (SDF-1α), to sites of injury. However, simply enhancing NSC migration is likely to result in insufficient regeneration, as the cells need to be given additional signals. Immobilized proteins, such as interferon-γ (IFN-γ) can encourage neurogenic differentiation of NSCs. Here, we combined both protein delivery paradigms: soluble SDF-1α delivery to enhance NSC migration alongside covalently tethered IFN-γ to differentiate the recruited NSCs into neurons. To slow the release of soluble SDF-1α, we copolymerized methacrylated heparin with methacrylamide chitosan (MAC), to which we tethered IFN-γ. We found that this hydrogel system could result in soft hydrogels with a ratio of up to 70:30 MAC/heparin by mass, which enabled the continuous release of SDF-1α over a period of 2 weeks. The hydrogels recruited NSCs in vitro over 2 weeks, proportional to their release rate: the 70:30 heparin gels recruited a consistent number of NSCs at each time point, while the formulations with less heparin recruited NSCs at only early time points. After remaining in contact with the hydrogels for 8 days, NSCs successfully differentiated into neurons. CNS regeneration is a complex challenge, and this system provides a foundation to address multiple aspects of that challenge.


Subject(s)
Central Nervous System/drug effects , Chemokine CXCL12/genetics , Interferon-gamma/genetics , Neural Stem Cells/drug effects , Acrylamides/chemistry , Acrylamides/pharmacology , Animals , Cell Differentiation/drug effects , Cell Movement/drug effects , Cells, Cultured , Central Nervous System/injuries , Central Nervous System/pathology , Chitosan/chemistry , Chitosan/pharmacology , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Immobilized Proteins/chemistry , Immobilized Proteins/pharmacology , Nerve Regeneration/drug effects , Neurogenesis/drug effects , Neurons/drug effects , Rats , Regeneration/drug effects , Solubility/drug effects
16.
Nano Lett ; 18(1): 1-8, 2018 01 10.
Article in English | MEDLINE | ID: mdl-29178811

ABSTRACT

Extracellular biophysical cues have a profound influence on a wide range of cell behaviors, including growth, motility, differentiation, apoptosis, gene expression, adhesion, and signal transduction. Cells not only respond to definitively mechanical cues from the extracellular matrix (ECM) but can also sometimes alter the mechanical properties of the matrix and hence influence subsequent matrix-based cues in both physiological and pathological processes. Interactions between cells and materials in vitro can modify cell phenotype and ECM structure, whether intentionally or inadvertently. Interactions between cell and matrix mechanics in vivo are of particular importance in a wide variety of disorders, including cancer, central nervous system injury, fibrotic diseases, and myocardial infarction. Both the in vitro and in vivo effects of this coupling between mechanics and biology hold important implications for clinical applications.


Subject(s)
Extracellular Matrix/metabolism , Mechanotransduction, Cellular , Animals , Biophysics , Cell Adhesion , Cell Differentiation , Cell Movement , Cell Proliferation , Central Nervous System/injuries , Central Nervous System/metabolism , Central Nervous System/pathology , Cytoskeleton/metabolism , Cytoskeleton/pathology , Extracellular Matrix/pathology , Focal Adhesions/metabolism , Focal Adhesions/pathology , Humans , Integrins/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Neoplasms/metabolism , Neoplasms/pathology , Translational Research, Biomedical
17.
Int J Mol Sci ; 20(2)2019 Jan 11.
Article in English | MEDLINE | ID: mdl-30642007

ABSTRACT

Astrocytes comprise the major non-neuronal cell population in the mammalian neurovascular unit. Traditionally, astrocytes are known to play broad roles in central nervous system (CNS) homeostasis, including the management of extracellular ion balance and pH, regulation of neurotransmission, and control of cerebral blood flow and metabolism. After CNS injury, cell⁻cell signaling between neuronal, glial, and vascular cells contribute to repair and recovery in the neurovascular unit. In this mini-review, we propose the idea that astrocytes play a central role in organizing these signals. During CNS recovery, reactive astrocytes communicate with almost all CNS cells and peripheral progenitors, resulting in the promotion of neurogenesis and angiogenesis, regulation of inflammatory response, and modulation of stem/progenitor response. Reciprocally, changes in neurons and vascular components of the remodeling brain should also influence astrocyte signaling. Therefore, understanding the complex and interdependent signaling pathways of reactive astrocytes after CNS injury may reveal fundamental mechanisms and targets for re-integrating the neurovascular unit and augmenting brain recovery.


Subject(s)
Astrocytes/cytology , Cell Communication , Central Nervous System/injuries , Astrocytes/metabolism , Central Nervous System/blood supply , Central Nervous System/metabolism , Cerebrovascular Circulation , Humans , Hydrogen-Ion Concentration , Signal Transduction , Synaptic Transmission
18.
Dev Dyn ; 247(1): 18-23, 2018 01.
Article in English | MEDLINE | ID: mdl-28643358

ABSTRACT

The failure of damaged axons to regrow underlies disability in central nervous system injury and disease. Therapies that stimulate axon repair will be critical to restore function. Extensive axon regeneration can be induced by manipulation of oncogenes and tumor suppressors; however, it has been difficult to translate this into functional recovery in models of spinal cord injury. The current challenge is to maximize the functional integration of regenerating axons to recover motor and sensory behaviors. Insights into axonal growth and wiring during nervous system development are helping guide new approaches to boost regeneration and functional connectivity after injury in the mature nervous system. Here we discuss our current understanding of axonal behavior after injury and prospects for the development of drugs to optimize axon regeneration and functional recovery after CNS injury. Developmental Dynamics 247:18-23, 2018. © 2017 Wiley Periodicals, Inc.


Subject(s)
Axons/physiology , Central Nervous System/injuries , Nerve Regeneration/physiology , Neurogenesis/physiology , Spinal Cord Injuries/physiopathology , Animals , Central Nervous System/physiopathology , Humans
19.
J Neurosci ; 37(45): 10808-10816, 2017 11 08.
Article in English | MEDLINE | ID: mdl-29118209

ABSTRACT

In vivo optical imaging has emerged as a powerful tool with which to study cellular responses to injury and disease in the mammalian CNS. Important new insights have emerged regarding axonal degeneration and regeneration, glial responses and neuroinflammation, changes in the neurovascular unit, and, more recently, neural transplantations. Accompanying a 2017 SfN Mini-Symposium, here, we discuss selected recent advances in understanding the neuronal, glial, and other cellular responses to CNS injury and disease with in vivo imaging of the rodent brain or spinal cord. We anticipate that in vivo optical imaging will continue to be at the forefront of breakthrough discoveries of fundamental mechanisms and therapies for CNS injury and disease.


Subject(s)
Central Nervous System Diseases/diagnostic imaging , Central Nervous System/diagnostic imaging , Central Nervous System/injuries , Neuroimaging/methods , Animals , Humans , Mice , Neuroimaging/instrumentation , Rats
20.
Crit Care Med ; 46(5): 791-798, 2018 05.
Article in English | MEDLINE | ID: mdl-29443814

ABSTRACT

OBJECTIVES: Sterile and infectious critical illnesses often result in vasoplegic shock and a robust systemic inflammatory response that are similar in presentation. The innate immune system is at the center of the response to both infectious and traumatic insults. Damage-associated molecular patterns are small molecules that are released from stressed or dying cells. Damage-associated molecular patterns activate pattern recognition receptors and coordinate the leading edge of the innate immune response. This review introduces the concept of damage-associated molecular patterns and how they activate a systemic inflammatory response, specifically in trauma, neurologic injury, and infection. It also explores how, when carried to extremes, damage-associated molecular patterns may even perpetuate multisystem organ failure. DATA SOURCES: Basic and clinical studies were obtained from a PubMed search through August 2017. STUDY SELECTION: Articles considered include original articles, review articles, and conference proceedings. DATA EXTRACTION: An analysis of scientific, peer-reviewed data was performed. High quality preclinical and clinical studies adjudicated by the authors were included and summarized. DATA SYNTHESIS: Pattern recognition receptors respond to damage-associated molecular patterns and then activate inflammatory pathways. Damage-associated molecular patterns have been linked to the recruitment of sentinel leukocytes and the initiation of the inflammatory cascade. Damage-associated molecular patterns have been linked to many conditions in critical care illnesses. Preclinical models have added insight into how they may mediate distant organ dysfunction. CONCLUSIONS: Damage-associated molecular pattern activation and release is an important research for intensive care practitioners. It will add to our understanding of the phase and state of the innate immune response to an insult. Early work is encouraging. However, only with improved understanding of damage-associated molecular pattern activation and function, we can perhaps hope to target damage-associated molecular patterns as diagnostic and/or therapeutic modalities in the future.


Subject(s)
Alarmins/physiology , Intensive Care Units , Central Nervous System/injuries , Critical Illness , Homeostasis , Humans , Immunity, Innate/physiology , Wounds and Injuries/immunology , Wounds and Injuries/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL