Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 4096, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35835751

ABSTRACT

Traumatic spinal cord injury (SCI) triggers a neuro-inflammatory response dominated by tissue-resident microglia and monocyte derived macrophages (MDMs). Since activated microglia and MDMs are morphologically identical and express similar phenotypic markers in vivo, identifying injury responses specifically coordinated by microglia has historically been challenging. Here, we pharmacologically depleted microglia and use anatomical, histopathological, tract tracing, bulk and single cell RNA sequencing to reveal the cellular and molecular responses to SCI controlled by microglia. We show that microglia are vital for SCI recovery and coordinate injury responses in CNS-resident glia and infiltrating leukocytes. Depleting microglia exacerbates tissue damage and worsens functional recovery. Conversely, restoring select microglia-dependent signaling axes, identified through sequencing data, in microglia depleted mice prevents secondary damage and promotes recovery. Additional bioinformatics analyses reveal that optimal repair after SCI might be achieved by co-opting key ligand-receptor interactions between microglia, astrocytes and MDMs.


Subject(s)
Spinal Cord Injuries , Spinal Cord Regeneration , Animals , Macrophages/pathology , Mice , Mice, Inbred C57BL , Microglia/pathology , Spinal Cord/pathology
2.
J Immunol ; 209(1): 157-170, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35697382

ABSTRACT

Pulmonary infection is a leading cause of morbidity and mortality after spinal cord injury (SCI). Although SCI causes atrophy and dysfunction in primary and secondary lymphoid tissues with a corresponding decrease in the number and function of circulating leukocytes, it is unknown whether this SCI-dependent systemic immune suppression also affects the unique tissue-specific antimicrobial defense mechanisms that protect the lung. In this study, we tested the hypothesis that SCI directly impairs pulmonary immunity and subsequently increases the risk for developing pneumonia. Using mouse models of severe high-level SCI, we find that recruitment of circulating leukocytes and transcriptional control of immune signaling in the lung is impaired after SCI, creating an environment that is permissive for infection. Specifically, we saw a sustained loss of pulmonary leukocytes, a loss of alveolar macrophages at chronic time points postinjury, and a decrease in immune modulatory genes, especially cytokines, needed to eliminate pulmonary infections. Importantly, this injury-dependent impairment of pulmonary antimicrobial defense is only partially overcome by boosting the recruitment of immune cells to the lung with the drug AMD3100, a Food and Drug Administration-approved drug that mobilizes leukocytes and hematopoietic stem cells from bone marrow. Collectively, these data indicate that the immune-suppressive effects of SCI extend to the lung, a unique site of mucosal immunity. Furthermore, preventing lung infection after SCI will likely require novel strategies, beyond the use of orthodox antibiotics, to reverse or block tissue-specific cellular and molecular determinants of pulmonary immune surveillance.


Subject(s)
Spinal Cord Injuries , Animals , Cytokines , Disease Models, Animal , Immunity , Lung , Mice , Spinal Cord
3.
Exp Neurol ; 355: 114114, 2022 09.
Article in English | MEDLINE | ID: mdl-35568187

ABSTRACT

Glucocorticoid receptors (GRs), part of the nuclear receptor superfamily of transcription factors (TFs), are ubiquitously expressed in all cell types and regulate cellular responses to glucocorticoids (e.g., cortisol in humans; corticosterone in rodents). In myeloid cells, glucocorticoids binding to GRs can enhance or repress gene transcription, thereby imparting distinct and context-dependent functions in macrophages at sites of inflammation. In experimental models and in humans, glucocorticoids are widely used as anti-inflammatory treatments to promote recovery of function after SCI. Thus, we predicted that deleting GR in mouse myeloid lineage cells (i.e., microglia and monocyte-derived macrophages) would enhance inflammation at the site of injury and worsen functional recovery after traumatic spinal cord injury (SCI). Contrary to our prediction, the intraspinal macrophage response to a moderate (75 kdyne) spinal contusion SCI was reduced in Cx3cr1-Cre;GRf/f conditional knockout mice (with GR specifically deleted in myeloid cells). This phenotype was associated with improvements in hindlimb motor recovery, myelin sparing, axon sparing/regeneration, and microvascular protection/plasticity relative to SCI mice with normal myeloid cell GR expression. Further analysis revealed that macrophage GR deletion impaired lipid and myelin phagocytosis and foamy macrophage formation. Together, these data reveal endogenous GR signaling as a key pathway that normally inhibits mechanisms of macrophage-mediated repair after SCI.


Subject(s)
Receptors, Glucocorticoid , Spinal Cord Injuries , Animals , Inflammation/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Microglia/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Recovery of Function/physiology , Spinal Cord/metabolism
4.
Glia ; 70(7): 1359-1379, 2022 07.
Article in English | MEDLINE | ID: mdl-35394085

ABSTRACT

Microglial control of activity-dependent plasticity and synaptic remodeling in neuronal networks has been the subject of intense research in the past several years. Although microglia-neuron interactions have been extensively studied, less is known about how microglia influence astrocyte-dependent control over neuronal structure and function. Here, we explored a role for microglia in regulating the structure and function of the astrocyte syncytium in mouse hippocampus. After depleting microglia using a CSF1R antagonist (PLX5622, Plexxikon), we observed severe disruption of astrocyte syncytial isopotentiality and dye coupling. A decrease in astrocyte-specific gap junction connexin (Cx) 30 and 43 expression, at least partially accounts for these microglia-dependent changes in astrocytes. Because neuronal function requires intact astrocyte coupling, we also evaluated the effects of microglia depletion on synaptic transmission in the hippocampus. Without microglia, the strength of synaptic transmission was reduced at baseline and after long-term potentiation (LTP). Conversely, priming microglia with systemic injections of lipopolysaccharide enhanced CA3-CA1 synaptic transmission. This microglia-induced scaling of synaptic transmission was associated with increased expression of post-synaptic scaffold proteins (Homer1) in CA1. However, astrocyte network function was not affected by microglia priming, indicating that microglia-dependent effects on astrocytes and neurons vary across functional states. Through manipulation of microglia in the brain, our results reveal the importance of microglia in homeostatic regulation of the astrocyte syncytium and scaling of synaptic transmission. These novel mechanisms uncover a new direction for future studies interrogating microglia function in various physiological and pathological contexts.


Subject(s)
Astrocytes , Microglia , Animals , Astrocytes/metabolism , Connexin 30/metabolism , Hippocampus/metabolism , Long-Term Potentiation/physiology , Mice , Microglia/metabolism , Neuronal Plasticity/physiology , Synaptic Transmission/physiology
5.
J Neurosci ; 42(17): 3659-3675, 2022 04 27.
Article in English | MEDLINE | ID: mdl-35304427

ABSTRACT

Traumatic spinal cord injury (SCI) above the major spinal sympathetic outflow (T6 level) disinhibits sympathetic neurons from supraspinal control, causing systems-wide "dysautonomia." We recently showed that remarkable structural remodeling and plasticity occurs within spinal sympathetic circuitry, creating abnormal sympathetic reflexes that exacerbate dysautonomia over time. As an example, thoracic VGluT2+ spinal interneurons (SpINs) become structurally and functionally integrated with neurons that comprise the spinal-splenic sympathetic network and immunological dysfunction becomes progressively worse after SCI. To test whether the onset and progression of SCI-induced sympathetic plasticity is neuron activity dependent, we selectively inhibited (or excited) thoracic VGluT2+ interneurons using chemogenetics. New data show that silencing VGluT2+ interneurons in female and male mice with a T3 SCI, using hM4Di designer receptors exclusively activated by designer drugs (Gi DREADDs), blocks structural plasticity and the development of dysautonomia. Specifically, silencing VGluT2+ interneurons prevents the structural remodeling of spinal sympathetic networks that project to lymphoid and endocrine organs, reduces the frequency of spontaneous autonomic dysreflexia (AD), and reduces the severity of experimentally induced AD. Features of SCI-induced structural plasticity can be recapitulated in the intact spinal cord by activating excitatory hM3Dq-DREADDs in VGluT2+ interneurons. Collectively, these data implicate VGluT2+ excitatory SpINs in the onset and propagation of SCI-induced structural plasticity and dysautonomia, and reveal the potential for neuromodulation to block or reduce dysautonomia after severe high-level SCI.SIGNIFICANCE STATEMENT In response to stress or dangerous stimuli, autonomic spinal neurons coordinate a "fight or flight" response marked by increased cardiac output and release of stress hormones. After a spinal cord injury (SCI), normally harmless stimuli like bladder filling can result in a "false" fight or flight response, causing pathological changes throughout the body. We show that progressive hypertension and immune suppression develop after SCI because thoracic excitatory VGluT2+ spinal interneurons (SpINs) provoke structural remodeling in autonomic networks within below-lesion spinal levels. These pathological changes can be prevented in SCI mice or phenocopied in uninjured mice using chemogenetics to selectively manipulate activity in VGluT2+ SpINs. Targeted neuromodulation of SpINs could prevent structural plasticity and subsequent autonomic dysfunction in people with SCI.


Subject(s)
Autonomic Dysreflexia , Primary Dysautonomias , Spinal Cord Injuries , Animals , Autonomic Dysreflexia/etiology , Female , Humans , Interneurons/pathology , Male , Mice , Primary Dysautonomias/complications , Spinal Cord/pathology
7.
Cell Rep ; 34(4): 108667, 2021 01 26.
Article in English | MEDLINE | ID: mdl-33503436

ABSTRACT

After spinal cord injury (SCI), normally innocuous visceral or somatic stimuli can trigger uncontrolled reflex activation of sympathetic circuitry, causing pathological dysautonomia. We show that remarkable structural remodeling and plasticity occur within spinal autonomic circuitry, creating abnormal sympathetic reflexes that promote dysautonomia. However, when mice are treated early after SCI with human-equivalent doses of the US Food and Drug Administration (FDA)-approved drug gabapentin (GBP), it is possible to block multi-segmental excitatory synaptogenesis and abolish sprouting of autonomic neurons that innervate immune organs and sensory afferents that trigger pain and autonomic dysreflexia (AD). This "prophylactic GBP" regimen decreases the frequency and severity of AD and protects against SCI-induced immune suppression. These benefits persist even 1 month after stopping treatment. GBP could be repurposed to prevent dysautonomia in at-risk individuals with high-level SCI.


Subject(s)
Autonomic Dysreflexia/therapy , Calcium Channel Blockers/therapeutic use , Excitatory Amino Acid Antagonists/therapeutic use , Gabapentin/therapeutic use , Spinal Cord Injuries/therapy , Animals , Autonomic Dysreflexia/pathology , Calcium Channel Blockers/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Gabapentin/pharmacology , Humans , Male , Mice , Spinal Cord Injuries/pathology
8.
J Neurosci ; 40(47): 9103-9120, 2020 11 18.
Article in English | MEDLINE | ID: mdl-33051350

ABSTRACT

Microglia are dynamic immunosurveillance cells in the CNS. Whether microglia are protective or pathologic is context dependent; the outcome varies as a function of time relative to the stimulus, activation state of neighboring cells in the microenvironment or within progression of a particular disease. Although brain microglia can be "primed" using bacterial lipopolysaccharide (LPS)/endotoxin, it is unknown whether LPS delivered systemically can also induce neuroprotective microglia in the spinal cord. Here, we show that serial systemic injections of LPS (1 mg/kg, i.p., daily) for 4 consecutive days (LPSx4) consistently elicit a reactive spinal cord microglia response marked by dramatic morphologic changes, increased production of IL-1, and enhanced proliferation without triggering leukocyte recruitment or overt neuropathology. Following LPSx4, reactive microglia frequently contact spinal cord endothelial cells. Targeted ablation or selective expression of IL-1 and IL-1 receptor (IL-1R) in either microglia or endothelia reveal that IL-1-dependent signaling between these cells mediates microglia activation. Using a mouse model of ischemic spinal cord injury in male and female mice, we show that preoperative LPSx4 provides complete protection from ischemia-induced neuron loss and hindlimb paralysis. Neuroprotection is partly reversed by either pharmacological elimination of microglia or selective removal of IL-1R in microglia or endothelia. These data indicate that spinal cord microglia are amenable to therapeutic reprogramming via systemic manipulation and that this potential can be harnessed to protect the spinal cord from injury.SIGNIFICANCE STATEMENT Data in this report indicate that a neuroprotective spinal cord microglia response can be triggered by daily systemic injections of LPS over a period of 4 d (LPSx4). The LPSx4 regimen induces morphologic transformation and enhances proliferation of spinal cord microglia without causing neuropathology. Using advanced transgenic mouse technology, we show that IL-1-dependent microglia-endothelia cross talk is necessary for eliciting this spinal cord microglia phenotype and also for conferring optimal protection to spinal motor neurons from ischemic spinal cord injury (ISCI). Collectively, these novel data show that it is possible to consistently elicit spinal cord microglia via systemic delivery of inflammogens to achieve a therapeutically effective neuroprotective response against ISCI.


Subject(s)
Cell Communication/drug effects , Endothelial Cells/drug effects , Interleukin-1/physiology , Lipopolysaccharides/pharmacology , Microglia/drug effects , Neuroprotective Agents/pharmacology , Spinal Cord/drug effects , Animals , Bromodeoxyuridine/pharmacology , Endothelial Cells/metabolism , Female , Interleukin-1/biosynthesis , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Neurons/drug effects , Neurons/pathology , Paralysis/chemically induced , Receptors, Interleukin-1 Type I/drug effects , Receptors, Interleukin-1 Type I/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Spinal Cord/metabolism
9.
Nat Commun ; 11(1): 3702, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32710081

ABSTRACT

Spinal cord injury (SCI) causes immune dysfunction, increasing the risk of infectious morbidity and mortality. Since bone marrow hematopoiesis is essential for proper immune function, we hypothesize that SCI disrupts bone marrow hematopoiesis. Indeed, SCI causes excessive proliferation of bone marrow hematopoietic stem and progenitor cells (HSPC), but these cells cannot leave the bone marrow, even after challenging the host with a potent inflammatory stimulus. Sequestration of HSPCs in bone marrow after SCI is linked to aberrant chemotactic signaling that can be reversed by post-injury injections of Plerixafor (AMD3100), a small molecule inhibitor of CXCR4. Even though Plerixafor liberates HSPCs and mature immune cells from bone marrow, competitive repopulation assays show that the intrinsic long-term functional capacity of HSPCs is still impaired in SCI mice. Together, our data suggest that SCI causes an acquired bone marrow failure syndrome that may contribute to chronic immune dysfunction.


Subject(s)
Bone Marrow Failure Disorders/etiology , Bone Marrow/metabolism , Spinal Cord Injuries/complications , Animals , Benzylamines , Bone Marrow/pathology , Bone Marrow Cells , Bone Marrow Failure Disorders/pathology , Cell Proliferation , Chemokine CXCL12 , Cyclams , Disease Models, Animal , Female , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Heterocyclic Compounds/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Receptors, CXCR4/antagonists & inhibitors , Signal Transduction , Spinal Cord Injuries/immunology
10.
Nucleic Acids Res ; 48(W1): W275-W286, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32421805

ABSTRACT

A group of genes controlled as a unit, usually by the same repressor or activator gene, is known as a regulon. The ability to identify active regulons within a specific cell type, i.e., cell-type-specific regulons (CTSR), provides an extraordinary opportunity to pinpoint crucial regulators and target genes responsible for complex diseases. However, the identification of CTSRs from single-cell RNA-Seq (scRNA-Seq) data is computationally challenging. We introduce IRIS3, the first-of-its-kind web server for CTSR inference from scRNA-Seq data for human and mouse. IRIS3 is an easy-to-use server empowered by over 20 functionalities to support comprehensive interpretations and graphical visualizations of identified CTSRs. CTSR data can be used to reliably characterize and distinguish the corresponding cell type from others and can be combined with other computational or experimental analyses for biomedical studies. CTSRs can, therefore, aid in the discovery of major regulatory mechanisms and allow reliable constructions of global transcriptional regulation networks encoded in a specific cell type. The broader impact of IRIS3 includes, but is not limited to, investigation of complex diseases hierarchies and heterogeneity, causal gene regulatory network construction, and drug development. IRIS3 is freely accessible from https://bmbl.bmi.osumc.edu/iris3/ with no login requirement.


Subject(s)
RNA-Seq , Regulon , Single-Cell Analysis , Software , Animals , Brain/metabolism , Cluster Analysis , Mice
11.
Sci Rep ; 9(1): 19105, 2019 12 13.
Article in English | MEDLINE | ID: mdl-31836828

ABSTRACT

Humanized mice can be used to better understand how the human immune system responds to central nervous system (CNS) injury and inflammation. The optimal parameters for using humanized mice in preclinical CNS injury models need to be established for appropriate use and interpretation. Here, we show that the developmental age of the human immune system significantly affects anatomical and functional outcome measures in a preclinical model of traumatic spinal cord injury (SCI). Specifically, it takes approximately 3-4 months for a stable and functionally competent human immune system to develop in neonatal immune compromised mice after they are engrafted with human umbilical cord blood stem cells. Humanized mice receiving a SCI before or after stable engraftment exhibit significantly different neuroinflammatory profiles. Importantly, the development of a mature human immune system was associated with worse lesion pathology and neurological recovery after SCI. In these mice, human T cells infiltrate the spinal cord lesion and directly contact human macrophages. Together, data in this report establish an optimal experimental framework for using humanized mice to help translate promising preclinical therapies for CNS injury.


Subject(s)
Cord Blood Stem Cell Transplantation , Spinal Cord Injuries/immunology , Spinal Cord Injuries/therapy , Animals , Disease Models, Animal , Female , Fetal Blood/cytology , Humans , Immune System , Inflammation , Lipopolysaccharides , Lymphocytes/cytology , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Spinal Cord/pathology , Spleen/cytology , T-Lymphocytes, Cytotoxic/cytology
12.
JCI Insight ; 4(9)2019 05 02.
Article in English | MEDLINE | ID: mdl-31045582

ABSTRACT

Traumatic spinal cord injury (SCI) triggers an acute-phase response that leads to systemic inflammation and rapid mobilization of bone marrow (BM) neutrophils into the blood. These mobilized neutrophils then accumulate in visceral organs and the injured spinal cord where they cause inflammatory tissue damage. The receptor for complement activation product 3a, C3aR1, has been implicated in negatively regulating the BM neutrophil response to tissue injury. However, the mechanism via which C3aR1 controls BM neutrophil mobilization, and also its influence over SCI outcomes, are unknown. Here, we show that the C3a/C3aR1 axis exerts neuroprotection in SCI by acting as a physiological antagonist against neutrophil chemotactic signals. We show that C3aR1 engages phosphatase and tensin homolog (PTEN), a negative regulator of the phosphatidylinositol 3-kinase (PI3K)/AKT pathway, to restrain C-X-C chemokine receptor type 2-driven BM neutrophil mobilization following trauma. These findings are of direct clinical significance as lower circulating neutrophil numbers at presentation were identified as a marker for improved recovery in human SCI. Our work thus identifies C3aR1 and its downstream intermediary, PTEN, as therapeutic targets to broadly inhibit neutrophil mobilization/recruitment following tissue injury and reduce inflammatory pathology.


Subject(s)
Neutrophils/metabolism , Receptors, Complement/genetics , Receptors, Complement/metabolism , Receptors, Interleukin-8B/metabolism , Spinal Cord Injuries/metabolism , Adult , Animals , Bone Marrow/pathology , Cell Adhesion , Cell Movement , Disease Models, Animal , Female , Humans , Inflammation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases , Receptor, Anaphylatoxin C5a/genetics , Spinal Cord Injuries/pathology , Transcriptome , Wounds and Injuries/pathology , Young Adult
13.
J Neuroimmunol ; 321: 1-11, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29957379

ABSTRACT

Traumatic spinal cord injury (SCI) causes widespread damage to neurons, glia and endothelia located throughout the spinal parenchyma. In response to the injury, resident and blood-derived leukocytes orchestrate an intraspinal inflammatory response that propagates secondary neuropathology and also promotes tissue repair. SCI also negatively affects autonomic control over peripheral immune organs, notably the spleen. The spleen is the largest secondary lymphoid organ in mammals, with major roles in blood filtration and host defense. Splenic function is carefully regulated by neuroendocrine mechanisms that ensure that the immune responses to infection or injury are proportionate to the initiating stimulus, and can be terminated when the stimulus is cleared. After SCI, control over the viscera, including endocrine and lymphoid tissues is lost due to damage to spinal autonomic (sympathetic) circuitry. This review begins by examining the normal structure and function of the spleen including patterns of innervation and the role played by the nervous system in regulating spleen function. We then describe how after SCI, loss of proper neural control over splenic function leads to systems-wide neuropathology, immune suppression and autoimmunity. We conclude by discussing opportunities for targeting the spleen to restore immune homeostasis, reduce morbidity and mortality, and improve functional recovery after SCI.


Subject(s)
Autoimmunity/physiology , Neuroimmunomodulation/physiology , Spinal Cord Injuries/immunology , Spleen/immunology , Animals , Humans , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/therapy , Spleen/innervation , Spleen/metabolism
14.
Curr Opin Neurol ; 31(3): 334-344, 2018 06.
Article in English | MEDLINE | ID: mdl-29465433

ABSTRACT

PURPOSE OF REVIEW: In adult mammals, a traumatic spinal cord injury (SCI) elicits a chronic unregulated neuroinflammatory response accompanied by seemingly paradoxical suppression of systemic immunity. These SCI-induced changes in immune function contribute to poor neurological outcomes and enhanced morbidity or mortality. Nonspecific anti-inflammatory or proinflammatory therapies are ineffective and can even worsen outcomes. Therefore, recent experimental SCI research has advanced the understanding of how neuroimmune cross-talk contributes to spinal cord and systemic pathology. RECENT FINDINGS: It is now appreciated that the immune response caused by injury to the brain or spinal cord encompasses heterogeneous elements that can drive events on the spectrum between exacerbating pathology and promoting tissue repair, within the spinal cord and throughout the body. Recent novel discoveries regarding the role and regulation of soluble factors, monocytes/macrophages, microRNAs, lymphocytes and systemic immune function are highlighted in this review. SUMMARY: A more nuanced understanding of how the immune system responds and reacts to nervous system injury will present an array of novel therapeutic opportunities for clinical SCI and other forms of neurotrauma.


Subject(s)
Brain/immunology , Recovery of Function/immunology , Spinal Cord Injuries/immunology , Spinal Cord/immunology , Animals , Humans , MicroRNAs
15.
J Neurotrauma ; 34(12): 2075-2085, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28173736

ABSTRACT

This study investigated the role of the alternative receptor for complement activation fragment C5a, C5aR2, in secondary inflammatory pathology after contusive spinal cord injury (SCI) in mice. C5ar2-/- mice exhibited decreased intraparenchymal tumor necrosis factor alpha and interleukin-6 acutely post-injury, but these reductions did not translate into improved outcomes. We show that loss of C5aR2 leads to increased lesion volumes, reduced myelin sparing, and significantly worsened recovery from SCI in C5ar2-/- animals compared to wild-type (WT) controls. Loss of C5aR2 did not alter leukocyte mobilization from the bone marrow in response to SCI, and neutrophil recruitment/presence at the lesion site was also not different between genotypes. Acute treatment of SCI mice with the selective C5aR1 antagonist, PMX205, improved SCI outcomes, compared to vehicle controls, and, importantly, fully alleviated the worsened recovery of C5ar2-/- mice compared to their WT counterparts. Collectively, these findings indicate that C5aR2 is neuroprotective and a novel target to restrain injurious C5a signaling after a major neurotraumatic event.


Subject(s)
Neuroprotection , Receptor, Anaphylatoxin C5a/physiology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Animals , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptides, Cyclic/pharmacology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/deficiency , Spinal Cord Injuries/immunology
16.
Ann Clin Transl Neurol ; 3(7): 495-511, 2016 07.
Article in English | MEDLINE | ID: mdl-27386499

ABSTRACT

OBJECTIVE: Traumatic spinal cord injury (SCI) elicits immediate neural cell death, axonal damage, and disruption of the blood-spinal cord barrier, allowing circulating immune cells and blood proteins into the spinal parenchyma. The inflammatory response to SCI involves robust complement system activation, which contributes to secondary injury and impairs neurological recovery. This study aimed to determine whether intravenous immunoglobulin (IVIg), an FDA-approved treatment for inflammatory conditions, can scavenge complement activation products and improve recovery from contusive SCI. METHODS: We used functional testing, noninvasive imaging, and detailed postmortem analysis to assess whether IVIg therapy is effective in a mouse model of severe contusive SCI. RESULTS: IVIg therapy at doses of 0.5-2 g/kg improved the functional and histopathological outcomes from SCI, conferring protection against lesion enlargement, demyelination, central canal dilation, and axonal degeneration. The benefits of IVIg were detectable through noninvasive diffusion tensor imaging (DTI), with IVIg treatment counteracting the progressive SCI-induced increase in radial diffusivity (RD) in white matter. Diffusion indices significantly correlated with the functional performance of individual mice and accurately predicted the degree of myelin preservation. Further experiments revealed that IVIg therapy reduced the presence of complement activation products and phagocytically active macrophages at the lesion site, providing insight as to its mechanisms of action. INTERPRETATION: Our findings highlight the potential of using IVIg as an immunomodulatory treatment for SCI, and the value of DTI to assess tissue damage and screen for the efficacy of candidate intervention strategies in preclinical models of SCI, both quantitatively and noninvasively.

17.
Semin Immunol ; 28(3): 292-308, 2016 06.
Article in English | MEDLINE | ID: mdl-27049459

ABSTRACT

The recognition that complement proteins are abundantly present and can have pathological roles in neurological conditions offers broad scope for therapeutic intervention. Accordingly, an increasing number of experimental investigations have explored the potential of harnessing the unique activation pathways, proteases, receptors, complexes, and natural inhibitors of complement, to mitigate pathology in acute neurotrauma and chronic neurodegenerative diseases. Here, we review mechanisms of complement activation in the central nervous system (CNS), and explore the effects of complement inhibition in cerebral ischemic-reperfusion injury, traumatic brain injury, spinal cord injury, Alzheimer's disease, amyotrophic lateral sclerosis, Parkinson's disease and Huntington's disease. We consider the challenges and opportunities arising from these studies. As complement therapies approach clinical translation, we provide perspectives on how promising complement-targeted therapeutics could become part of novel and effective future treatment options to improve outcomes in the initiation and progression stages of these debilitating CNS disorders.


Subject(s)
Brain/pathology , Complement Inactivating Agents/therapeutic use , Complement System Proteins/metabolism , Neurodegenerative Diseases/therapy , Wounds and Injuries/therapy , Clinical Trials as Topic , Complement Activation , Disease Progression , Humans , Molecular Targeted Therapy , Neurodegenerative Diseases/immunology , Neuroprotection , Treatment Outcome , Wounds and Injuries/immunology
18.
Neural Regen Res ; 10(10): 1596-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26692854
19.
J Neurosci ; 35(16): 6517-31, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25904802

ABSTRACT

This study investigated the role of the complement activation fragment C5a in secondary pathology following contusive spinal cord injury (SCI). C5ar(-/-) mice, which lack the signaling receptor for C5a, displayed signs of improved locomotor recovery and reduced inflammation during the first week of SCI compared with wild-type mice. Intriguingly, the early signs of improved recovery in C5ar(-/-) mice deteriorated from day 14 onward, with absence of C5aR ultimately leading to poorer functional outcomes, larger lesion volumes, reduced myelin content, and more widespread inflammation at 35 d SCI. Pharmacological blockade of C5aR with a selective antagonist (C5aR-A) during the first 7 d after SCI improved recovery compared with vehicle-treated mice, and this phenotype was sustained up to 35 d after injury. Consistent with observations made in C5ar(-/-) mice, these improvements were, however, lost if C5aR-A administration was continued into the more chronic phase of SCI. Signaling through the C5a-C5aR axis thus appears injurious in the acute period but serves a protective and/or reparative role in the post-acute phase of SCI. Further experiments in bone marrow chimeric mice suggested that the dual and opposing roles of C5aR on SCI outcomes primarily relate to its expression on CNS-resident cells and not infiltrating leukocytes. Additional in vivo and in vitro studies provided direct evidence that C5aR signaling is required during the postacute phase for astrocyte hyperplasia, hypertrophy, and glial scar formation. Collectively, these findings highlight the complexity of the inflammatory response to SCI and emphasize the importance of optimizing the timing of therapeutic interventions.


Subject(s)
Gliosis/physiopathology , Inflammation/physiopathology , Receptor, Anaphylatoxin C5a/physiology , Spinal Cord Injuries/physiopathology , Animals , Astrocytes/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Complement Activation/drug effects , Complement Activation/physiology , Complement C5a/biosynthesis , Female , Gliosis/complications , Gliosis/drug therapy , Gliosis/pathology , Inflammation/complications , Inflammation/drug therapy , Inflammation/pathology , Mice , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/biosynthesis , Receptor, Anaphylatoxin C5a/genetics , Recovery of Function/physiology , Spinal Cord Injuries/complications , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism
20.
Nat Commun ; 5: 3450, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24625684

ABSTRACT

Stroke is a major cause of death worldwide and the leading cause of permanent disability. Although reperfusion is currently used as treatment, the restoration of blood flow following ischaemia elicits a profound inflammatory response mediated by proinflammatory cytokines such as tumour necrosis factor (TNF), exacerbating tissue damage and worsening the outcomes for stroke patients. Phosphoinositide 3-kinase delta (PI3Kδ) controls intracellular TNF trafficking in macrophages and therefore represents a prospective target to limit neuroinflammation. Here we show that PI3Kδ inhibition confers protection in ischaemia/reperfusion models of stroke. In vitro, restoration of glucose supply following an episode of glucose deprivation potentiates TNF secretion from primary microglia-an effect that is sensitive to PI3Kδ inhibition. In vivo, transient middle cerebral artery occlusion and reperfusion in kinase-dead PI3Kδ (p110δ(D910A/D910A)) or wild-type mice pre- or post-treated with the PI3Kδ inhibitor CAL-101, leads to reduced TNF levels, decreased leukocyte infiltration, reduced infarct size and improved functional outcome. These data identify PI3Kδ as a potential therapeutic target in ischaemic stroke.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Stroke/enzymology , Stroke/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Class I Phosphatidylinositol 3-Kinases , Disease Models, Animal , Inflammation/metabolism , Male , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...