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










Publication year range
1.
JCI Insight ; 8(22)2023 Nov 22.
Article in English | MEDLINE | ID: mdl-37991018

ABSTRACT

We previously established that vascular smooth muscle-derived adventitial progenitor cells (AdvSca1-SM) preferentially differentiate into myofibroblasts and contribute to fibrosis in response to acute vascular injury. However, the role of these progenitor cells in chronic atherosclerosis has not been defined. Using an AdvSca1-SM cell lineage tracing model, scRNA-Seq, flow cytometry, and histological approaches, we confirmed that AdvSca1-SM-derived cells localized throughout the vessel wall and atherosclerotic plaques, where they primarily differentiated into fibroblasts, smooth muscle cells (SMC), or remained in a stem-like state. Krüppel-like factor 4 (Klf4) knockout specifically in AdvSca1-SM cells induced transition to a more collagen-enriched fibroblast phenotype compared with WT mice. Additionally, Klf4 deletion drastically modified the phenotypes of non-AdvSca1-SM-derived cells, resulting in more contractile SMC and atheroprotective macrophages. Functionally, overall plaque burden was not altered with Klf4 deletion, but multiple indices of plaque composition complexity, including necrotic core area, macrophage accumulation, and fibrous cap thickness, were reduced. Collectively, these data support that modulation of AdvSca1-SM cells through KLF4 depletion confers increased protection from the development of potentially unstable atherosclerotic plaques.


Subject(s)
Plaque, Atherosclerotic , Mice , Animals , Plaque, Atherosclerotic/pathology , Kruppel-Like Factor 4 , Myocytes, Smooth Muscle/pathology , Stem Cells/pathology , Muscle, Smooth/pathology
2.
bioRxiv ; 2023 Jul 19.
Article in English | MEDLINE | ID: mdl-37503181

ABSTRACT

We previously established that vascular smooth muscle-derived adventitial progenitor cells (AdvSca1-SM) preferentially differentiate into myofibroblasts and contribute to fibrosis in response to acute vascular injury. However, the role of these progenitor cells in chronic atherosclerosis has not been defined. Using an AdvSca1-SM lineage tracing model, scRNA-Seq, flow cytometry, and histological approaches, we confirmed that AdvSca1-SM cells localize throughout the vessel wall and atherosclerotic plaques, where they primarily differentiate into fibroblasts, SMCs, or remain in a stem-like state. Klf4 knockout specifically in AdvSca1-SM cells induced transition to a more collagen-enriched myofibroblast phenotype compared to WT mice. Additionally, Klf4 depletion drastically modified the phenotypes of non-AdvSca1-SM-derived cells, resulting in more contractile SMCs and atheroprotective macrophages. Functionally, overall plaque burden was not altered with Klf4 depletion, but multiple indices of plaque vulnerability were reduced. Collectively, these data support that modulating the AdvSca1-SM population confers increased protection from the development of unstable atherosclerotic plaques.

3.
JCI Insight ; 8(9)2023 05 08.
Article in English | MEDLINE | ID: mdl-36976650

ABSTRACT

Vascular smooth muscle-derived Sca1+ adventitial progenitor (AdvSca1-SM) cells are tissue-resident, multipotent stem cells that contribute to progression of vascular remodeling and fibrosis. Upon acute vascular injury, AdvSca1-SM cells differentiate into myofibroblasts and are embedded in perivascular collagen and the extracellular matrix. While the phenotypic properties of AdvSca1-SM-derived myofibroblasts have been defined, the underlying epigenetic regulators driving the AdvSca1-SM-to-myofibroblast transition are unclear. We show that the chromatin remodeler Smarca4/Brg1 facilitates AdvSca1-SM myofibroblast differentiation. Brg1 mRNA and protein were upregulated in AdvSca1-SM cells after acute vascular injury, and pharmacological inhibition of Brg1 by the small molecule PFI-3 attenuated perivascular fibrosis and adventitial expansion. TGF-ß1 stimulation of AdvSca1-SM cells in vitro reduced expression of stemness genes while inducing expression of myofibroblast genes that was associated with enhanced contractility; PFI blocked TGF-ß1-induced phenotypic transition. Similarly, genetic knockdown of Brg1 in vivo reduced adventitial remodeling and fibrosis and reversed AdvSca1-SM-to-myofibroblast transition in vitro. Mechanistically, TGF-ß1 promoted redistribution of Brg1 from distal intergenic sites of stemness genes and recruitment to promoter regions of myofibroblast-related genes, which was blocked by PFI-3. These data provide insight into epigenetic regulation of resident vascular progenitor cell differentiation and support that manipulating the AdvSca1-SM phenotype will provide antifibrotic clinical benefits.


Subject(s)
Myofibroblasts , Vascular System Injuries , Humans , Myofibroblasts/metabolism , Transforming Growth Factor beta1/metabolism , Chromatin/metabolism , Vascular System Injuries/metabolism , Vascular System Injuries/pathology , Epigenesis, Genetic , Cell Differentiation , Muscle, Smooth, Vascular , Fibrosis , DNA Helicases/genetics , DNA Helicases/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Cardiovasc Res ; 118(6): 1452-1465, 2022 05 06.
Article in English | MEDLINE | ID: mdl-33989378

ABSTRACT

Cardiovascular diseases are characterized by chronic vascular dysfunction and provoke pathological remodelling events, such as neointima formation, atherosclerotic lesion development, and adventitial fibrosis. While lineage-tracing studies have shown that phenotypically modulated smooth muscle cells (SMCs) are the major cellular component of neointimal lesions, the cellular origins and microenvironmental signalling mechanisms that underlie remodelling along the adventitial vascular layer are not fully understood. However, a growing body of evidence supports a unique population of adventitial lineage-restricted progenitor cells expressing the stem cell marker, stem cell antigen-1 (Sca1; AdvSca1 cells) as important effectors of adventitial remodelling and suggests that they are at least partially responsible for subsequent pathological changes that occur in the media and intima. AdvSca1 cells are being studied in murine models of atherosclerosis, perivascular fibrosis, and neointima formation in response to acute vascular injury. Depending on the experimental conditions, AdvSca1 cells exhibit the capacity to differentiate into SMCs, endothelial cells, chondrocytes, adipocytes, and pro-remodelling cells, such as myofibroblasts and macrophages. These data indicate that AdvSca1 cells may be a targetable cell population to influence the outcomes of pathologic vascular remodelling. Important questions remain regarding the origins of AdvSca1 cells and the essential signalling mechanisms and microenvironmental factors that regulate both maintenance of their stem-like, progenitor phenotype and their differentiation into lineage-specified cell types. Adding complexity to the story, recent data indicate that the collective population of adventitial progenitor cells is likely composed of several smaller, lineage-restricted subpopulations, which are not fully defined by their transcriptomic profile and differentiation capabilities. The aim of this review is to outline the heterogeneity of Sca1+ adventitial progenitor cells, summarize their role in vascular homeostasis and remodelling, and comment on their translational relevance in humans.


Subject(s)
Atherosclerosis , Spinocerebellar Ataxias , Animals , Atherosclerosis/metabolism , Cell Differentiation/genetics , Endothelial Cells/pathology , Fibrosis , Homeostasis , Mice , Myocytes, Smooth Muscle/metabolism , Neointima/metabolism , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/pathology , Stem Cells/metabolism , Vascular Remodeling
5.
JCI Insight ; 5(23)2020 12 03.
Article in English | MEDLINE | ID: mdl-33119549

ABSTRACT

Resident vascular adventitial SCA1+ progenitor (AdvSca1) cells are essential in vascular development and injury. However, the heterogeneity of AdvSca1 cells presents a unique challenge in understanding signaling pathways orchestrating their behavior in homeostasis and injury responses. Using smooth muscle cell (SMC) lineage-tracing models, we identified a subpopulation of AdvSca1 cells (AdvSca1-SM) originating from mature SMCs that undergo reprogramming in situ and exhibit a multipotent phenotype. Here we employed lineage tracing and RNA-sequencing to define the signaling pathways regulating SMC-to-AdvSca1-SM cell reprogramming and AdvSca1-SM progenitor cell phenotype. Unbiased hierarchical clustering revealed that genes related to hedgehog/WNT/beta-catenin signaling were significantly enriched in AdvSca1-SM cells, emphasizing the importance of this signaling axis in the reprogramming event. Leveraging AdvSca1-SM-specific expression of GLI-Kruppel family member GLI1 (Gli1), we generated Gli1-CreERT2-ROSA26-YFP reporter mice to selectively track AdvSca1-SM cells. We demonstrated that physiologically relevant vascular injury or AdvSca1-SM cell-specific Kruppel-like factor 4 (Klf4) depletion facilitated the proliferation and differentiation of AdvSca1-SM cells to a profibrotic myofibroblast phenotype rather than macrophages. Surprisingly, AdvSca1-SM cells selectively contributed to adventitial remodeling and fibrosis but little to neointima formation. Together, these findings strongly support therapeutics aimed at preserving the AdvSca1-SM cell phenotype as a viable antifibrotic approach.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Vascular Remodeling/genetics , Animals , Arteries/metabolism , Cell Differentiation/genetics , Female , Fibrosis/genetics , Fibrosis/metabolism , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Myofibroblasts/metabolism , Stem Cells/metabolism , Vascular Remodeling/physiology , Wnt Signaling Pathway
6.
Arterioscler Thromb Vasc Biol ; 40(8): 1854-1869, 2020 08.
Article in English | MEDLINE | ID: mdl-32580634

ABSTRACT

OBJECTIVE: Our recent work demonstrates that PTEN (phosphatase and tensin homolog) is an important regulator of smooth muscle cell (SMC) phenotype. SMC-specific PTEN deletion promotes spontaneous vascular remodeling and PTEN loss correlates with increased atherosclerotic lesion severity in human coronary arteries. In mice, PTEN overexpression reduces plaque area and preserves SMC contractile protein expression in atherosclerosis and blunts Ang II (angiotensin II)-induced pathological vascular remodeling, suggesting that pharmacological PTEN upregulation could be a novel therapeutic approach to treat vascular disease. Approach and Results: To identify novel PTEN activators, we conducted a high-throughput screen using a fluorescence based PTEN promoter-reporter assay. After screening ≈3400 compounds, 11 hit compounds were chosen based on level of activity and mechanism of action. Following in vitro confirmation, we focused on 5-azacytidine, a DNMT1 (DNA methyltransferase-1) inhibitor, for further analysis. In addition to PTEN upregulation, 5-azacytidine treatment increased expression of genes associated with a differentiated SMC phenotype. 5-Azacytidine treatment also maintained contractile gene expression and reduced inflammatory cytokine expression after PDGF (platelet-derived growth factor) stimulation, suggesting 5-azacytidine blocks PDGF-induced SMC de-differentiation. However, these protective effects were lost in PTEN-deficient SMCs. These findings were confirmed in vivo using carotid ligation in SMC-specific PTEN knockout mice treated with 5-azacytidine. In wild type controls, 5-azacytidine reduced neointimal formation and inflammation while maintaining contractile protein expression. In contrast, 5-azacytidine was ineffective in PTEN knockout mice, indicating that the protective effects of 5-azacytidine are mediated through SMC PTEN upregulation. CONCLUSIONS: Our data indicates 5-azacytidine upregulates PTEN expression in SMCs, promoting maintenance of SMC differentiation and reducing pathological vascular remodeling in a PTEN-dependent manner.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , High-Throughput Screening Assays/methods , PTEN Phosphohydrolase/physiology , Vascular Remodeling/drug effects , Animals , Azacitidine/pharmacology , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/drug effects , PTEN Phosphohydrolase/genetics , Platelet-Derived Growth Factor/pharmacology , Promoter Regions, Genetic
7.
Arterioscler Thromb Vasc Biol ; 40(2): 394-403, 2020 02.
Article in English | MEDLINE | ID: mdl-31852223

ABSTRACT

OBJECTIVE: Pathological vascular remodeling and excessive perivascular fibrosis are major contributors to reduced vessel compliance that exacerbates cardiovascular diseases, for instance, promoting clinically relevant myocardial remodeling. Inflammation plays a significant role in both pathological vascular remodeling and fibrosis. We previously demonstrated that smooth muscle cell-specific PTEN depletion promotes significant vascular fibrosis and accumulation of inflammatory cells. In the current study, we aimed to determine the beneficial role of systemic PTEN elevation on Ang II (angiotensin II)-induced vascular fibrosis and remodeling. Approach and Results: Transgenic mice carrying additional copies of the wild-type Pten gene (super PTEN [sPTEN]) and WT littermates were subjected to Ang II or saline infusion for 14 or 28 days. Compared with WT, Ang II-induced vascular fibrosis was significantly blunted in sPTEN mice, as shown by histochemical stainings and label-free second harmonic generation imaging. The protection against Ang II was recapitulated in sPTEN mice bearing WT bone marrow but not in WT mice reconstituted with sPTEN bone marrow. Ang II-induced elevation of profibrotic and proinflammatory gene expression observed in WT mice was blocked in aortic tissue of sPTEN mice. Immunofluorescent staining and flow cytometry both indicated that perivascular infiltration of T cells and macrophages was significantly inhibited in sPTEN mice. In vitro induction of PTEN expression suppressed Ang II-induced Ccl2 expression in vascular smooth muscle cells. CONCLUSIONS: Systemic PTEN elevation mediates protection against Ang II-induced vascular inflammation and fibrosis predominantly through effects in resident vascular cells. Our data highly support that pharmacological upregulation of PTEN could be a novel and viable approach for the treatment of pathological vascular fibrosis.


Subject(s)
Gene Expression Regulation , Muscle, Smooth, Vascular/metabolism , PTEN Phosphohydrolase/genetics , Vascular Diseases/genetics , Vascular Remodeling/genetics , Angiotensin II/toxicity , Animals , Blotting, Western , Cells, Cultured , Disease Models, Animal , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Flow Cytometry , Male , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/pathology , PTEN Phosphohydrolase/biosynthesis , RNA/genetics , Rats , Vascular Diseases/metabolism , Vascular Diseases/pathology
8.
Anesth Analg ; 128(1): 161-167, 2019 01.
Article in English | MEDLINE | ID: mdl-29596097

ABSTRACT

BACKGROUND: Opioids are effective postoperative analgesics. Disturbingly, we have previously reported that opioids such as morphine can worsen inflammatory pain and peripheral and central neuropathic pain. These deleterious effects are mediated by immune mediators that promote neuronal hyperexcitability in the spinal dorsal horn. Herein, we tested whether perioperative morphine could similarly prolong postoperative pain in male rats. METHODS: Rats were treated with morphine for 7 days, beginning immediately after laparotomy, while the morphine was tapered in a second group. Expression of genes for inflammatory mediators was quantified in the spinal dorsal horn. In the final experiment, morphine was administered before laparotomy for 7 days. RESULTS: We found that morphine treatment after laparotomy extended postoperative pain by more than 3 weeks (time × treatment: P < .001; time: P < .001; treatment: P < .05). Extension of postoperative pain was not related to morphine withdrawal, as it was not prevented by dose tapering (time × treatment: P = .8; time: P < .001; treatment: P = .9). Prolonged postsurgical pain was associated with increased expression of inflammatory genes, including those encoding Toll-like receptor 4, NOD like receptor protein 3 (NLRP3), nuclear factor kappa B (NFκB), caspase-1, interleukin-1ß, and tumor necrosis factor (P < .05). Finally, we showed that of preoperative morphine, concluding immediately before laparotomy, similarly prolonged postoperative pain (time × treatment: P < .001; time: P < .001; treatment: P < .001). There is a critical window for morphine potentiation of pain, as a 7-day course of morphine that concluded 1 week before laparotomy did not prolong postsurgical pain. CONCLUSIONS: These studies indicate the morphine can have a deleterious effect on postoperative pain. These studies further suggest that longitudinal studies could be performed to test whether opioids similarly prolong postoperative pain in the clinic.


Subject(s)
Analgesics, Opioid/toxicity , Hyperalgesia/chemically induced , Morphine/toxicity , Pain Threshold/drug effects , Pain, Postoperative/chemically induced , Posterior Horn Cells/drug effects , Analgesics, Opioid/administration & dosage , Animals , Disease Models, Animal , Drug Administration Schedule , Hyperalgesia/diagnostic imaging , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Inflammation Mediators/metabolism , Laparotomy , Male , Morphine/administration & dosage , Pain Measurement , Pain, Postoperative/drug therapy , Pain, Postoperative/metabolism , Pain, Postoperative/physiopathology , Posterior Horn Cells/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Time Factors
9.
Brain Res ; 1692: 9-11, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29723521

ABSTRACT

We have recently reported that a short course of morphine, starting 10 days after sciatic chronic constriction injury (CCI), prolonged the duration of mechanical allodynia for months after morphine ceased. Maintenance of this morphine-induced persistent sensitization was dependent on microglial reactivity and Toll-like receptor 4 signaling. Given that microRNAs (miRNAs) such as miR-124 and miR-146a possess the ability to modulate such signaling, we directly compared their function in this model. We found that both miRNAs reversed established allodynia in our model of morphine-induced persistent sensitization. The efficacy of miR-124 and miR-146a were comparable, and in both cases allodynia returned within hours to days of miRNA dosing conclusion. Our findings demonstrate that miRNAs targeting Toll-like receptor signaling are effective in reversing neuropathic pain, which underscores the clinical potential of these non-coding RNAs.


Subject(s)
Analgesics, Opioid/adverse effects , Anti-Inflammatory Agents/therapeutic use , MicroRNAs/therapeutic use , Morphine/adverse effects , Neuralgia/physiopathology , Analgesics, Opioid/therapeutic use , Animals , Disease Models, Animal , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Morphine/therapeutic use , Neuralgia/drug therapy , Pain Threshold/drug effects , Rats , Rats, Inbred F344
10.
Exp Neurol ; 304: 125-131, 2018 06.
Article in English | MEDLINE | ID: mdl-29530713

ABSTRACT

The absence of selective pharmacological tools is a major barrier to the in vivo study of microglia. To address this issue, we developed a Gq- and Gi-coupled Designer Receptor Exclusively Activated by a Designer Drug (DREADD) to enable selective stimulation or inhibition of microglia, respectively. DREADDs under a CD68 (microglia/macrophage) promoter were intrathecally transfected via an AAV9 vector. Naïve male rats intrathecally transfected with Gq (stimulatory) DREADDs exhibited significant allodynia following intrathecal administration of the DREADD-selective ligand clozapine-N-oxide (CNO), which was abolished by intrathecal interleukin-1 receptor antagonist. Chronic constriction injury-induced allodynia was attenuated by intrathecal CNO in male rats intrathecally transfected with Gi (inhibitory) DREADDs. To explore mechanisms, BV2 cells were stably transfected with Gq or Gi DREADDs in vitro. CNO treatment induced pro-inflammatory mediator production per se from cells expressing Gq-DREADDs, and inhibited lipopolysaccharide- and CCL2-induced inflammatory signaling from cells expressing Gi-DREADDs. These studies are the first to manipulate microglia function using DREADDs, which allow the role of glia in pain to be conclusively demonstrated, unconfounded by neuronal off-target effects that exist for all other drugs that also inhibit glia. Hence, these studies are the first to conclusively demonstrate that in vivo stimulation of resident spinal microglia in intact spinal cord is a) sufficient for allodynia, and b) necessary for allodynia induced by peripheral nerve injury. DREADDs are a unique tool to selectively explore the physiological and pathological role of microglia in vivo.


Subject(s)
Microglia/metabolism , Neuralgia/physiopathology , Receptors, G-Protein-Coupled/metabolism , Animals , Clozapine/analogs & derivatives , Clozapine/pharmacology , Designer Drugs/pharmacology , Disease Models, Animal , Inflammation/metabolism , Inflammation/physiopathology , Male , Microglia/drug effects , Neuralgia/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/physiopathology , Transfection
11.
Brain Behav Immun ; 72: 45-50, 2018 08.
Article in English | MEDLINE | ID: mdl-28860068

ABSTRACT

We have recently reported that a short course of morphine, starting 10days after sciatic chronic constriction injury (CCI), prolonged the duration of mechanical allodynia for months after morphine ceased. Maintenance of this morphine-induced persistent sensitization was dependent on spinal NOD-like receptor protein 3 (NLRP3) inflammasomes-protein complexes that proteolytically activate interleukin-1ß (IL-1ß) via caspase-1. However, it is still unclear how NLRP3 inflammasome signaling is maintained long after morphine is cleared. Here, we demonstrate that spinal levels of the damage associated molecular patterns (DAMPs) high mobility group box 1 (HMGB1) and biglycan are elevated during morphine-induced persistent sensitization in male rats; that is, 5weeks after cessation of morphine dosing. We also show that HMGB1 and biglycan levels are at least partly dependent on the initial activation of caspase-1, as well as Toll like receptor 4 (TLR4) and the purinergic receptor P2X7R-receptors responsible for priming and activation of NLRP3 inflammasomes. Finally, pharmacological attenuation of the DAMPs HMGB1, biglycan, heat shock protein 90 and fibronectin persistently reversed morphine-prolonged allodynia. We conclude that after peripheral nerve injury, morphine treatment results in persistent DAMP release via TLR4, P2X7R and caspase-1, which are involved in formation/activation of NLRP3 inflammasomes. These DAMPs are responsible for maintaining persistent allodynia, which may be due to engagement of a positive feedback loop, in which NLRP3 inflammasomes are persistently activated by DAMPs signaling at TLR4 and P2X7R.


Subject(s)
Alarmins/physiology , Peripheral Nerve Injuries/drug therapy , Spinal Injuries/immunology , Alarmins/drug effects , Animals , Caspase 1/metabolism , HMGB1 Protein/metabolism , Hyperalgesia/metabolism , Inflammasomes/metabolism , Injections, Spinal , Interleukin-1beta/metabolism , Male , Morphine/metabolism , Morphine/therapeutic use , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuralgia/metabolism , Rats , Rats, Inbred F344 , Receptors, Purinergic P2X7/metabolism , Spinal Injuries/drug therapy , Toll-Like Receptor 4/metabolism
12.
Brain Behav Immun ; 59: 49-54, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27189037

ABSTRACT

Relapsing-remitting multiple sclerosis is commonly associated with motor impairments, neuropathic pain, fatigue, mood disorders, and decreased life expectancy. However, preclinical pharmacological studies predominantly rely on clinical scoring of motor deficit as the sole behavioral endpoint. Thus, the translational potential of these studies is limited. Here, we have assessed the therapeutic potential of a novel anti-inflammatory interleukin-10 (IL-10) non-viral gene therapy formulation (XT-101-R) in a rat relapsing remitting experimental autoimmune encephalomyelitis (EAE) model. EAE induced motor deficits and neuropathic pain as reflected by induction of low-threshold mechanical allodynia, suppressed voluntary wheel running, decreased social exploration, and was associated with markedly enhanced mortality. We also noted that voluntary wheel running was depressed prior to the onset of motor deficit, and may therefore serve as a predictor of clinical symptoms onset. XT-101-R was intrathecally dosed only once at the onset of motor deficits, and attenuated each of the EAE-induced symptoms and improved survival, relative to vehicle control. This is the first pharmacological assessment of such a broad range of EAE symptoms, and provides support for IL-10 gene therapy as a clinical strategy for the treatment of multiple sclerosis.


Subject(s)
Anxiety/psychology , Anxiety/therapy , Behavior, Animal/drug effects , Encephalomyelitis, Autoimmune, Experimental/psychology , Encephalomyelitis, Autoimmune, Experimental/therapy , Fatigue/psychology , Fatigue/therapy , Interleukin-10/genetics , Neuralgia/psychology , Neuralgia/therapy , Animals , Exploratory Behavior , Genetic Therapy , Hyperalgesia/psychology , Hyperalgesia/therapy , Injections, Spinal , Interpersonal Relations , Life Expectancy , Male , Motor Activity , Rats
13.
Pain ; 157(9): 2012-23, 2016 09.
Article in English | MEDLINE | ID: mdl-27355182

ABSTRACT

Exercise is known to exert a systemic anti-inflammatory influence, but whether its effects are sufficient to protect against subsequent neuropathic pain is underinvestigated. We report that 6 weeks of voluntary wheel running terminating before chronic constriction injury (CCI) prevented the full development of allodynia for the ∼3-month duration of the injury. Neuroimmune signaling was assessed at 3 and 14 days after CCI. Prior exercise normalized ipsilateral dorsal spinal cord expression of neuroexcitatory interleukin (IL)-1ß production and the attendant glutamate transporter GLT-1 decrease, as well as expression of the disinhibitory P2X4R-BDNF axis. The expression of the macrophage marker Iba1 and the chemokine CCL2 (MCP-1), and a neuronal injury marker (activating transcription factor 3), was attenuated by prior running in the ipsilateral lumbar dorsal root ganglia. Prior exercise suppressed macrophage infiltration and/or injury site proliferation, given decreased presence of macrophage markers Iba1, iNOS (M1), and Arg-1 (M2; expression was time dependent). Chronic constriction injury-driven increases in serum proinflammatory chemokines were suppressed by prior running, whereas IL-10 was increased. Peripheral blood mononuclear cells were also stimulated with lipopolysaccharide ex vivo, wherein CCI-induced increases in IL-1ß, nitrite, and IL-10 were suppressed by prior exercise. Last, unrestricted voluntary wheel running, beginning either the day of, or 2 weeks after, CCI, progressively reversed neuropathic pain. This study is the first to investigate the behavioral and neuroimmune consequences of regular exercise terminating before nerve injury. This study suggests that chronic pain should be considered a component of "the diseasome of physical inactivity," and that an active lifestyle may prevent neuropathic pain.


Subject(s)
Exercise Movement Techniques/methods , Neuralgia/prevention & control , Activating Transcription Factor 3/metabolism , Animals , Calcium-Binding Proteins/metabolism , Constriction, Pathologic/complications , Cytokines/metabolism , Disease Models, Animal , Excitatory Amino Acid Transporter 2/metabolism , Functional Laterality , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Hyperalgesia/rehabilitation , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Male , Microfilament Proteins/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuralgia/etiology , Neuralgia/pathology , Nitrites/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X5/metabolism , Sciatic Neuropathy/prevention & control , p21-Activated Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Proc Natl Acad Sci U S A ; 113(24): E3441-50, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27247388

ABSTRACT

Opioid use for pain management has dramatically increased, with little assessment of potential pathophysiological consequences for the primary pain condition. Here, a short course of morphine, starting 10 d after injury in male rats, paradoxically and remarkably doubled the duration of chronic constriction injury (CCI)-allodynia, months after morphine ceased. No such effect of opioids on neuropathic pain has previously been reported. Using pharmacologic and genetic approaches, we discovered that the initiation and maintenance of this multimonth prolongation of neuropathic pain was mediated by a previously unidentified mechanism for spinal cord and pain-namely, morphine-induced spinal NOD-like receptor protein 3 (NLRP3) inflammasomes and associated release of interleukin-1ß (IL-1ß). As spinal dorsal horn microglia expressed this signaling platform, these cells were selectively inhibited in vivo after transfection with a novel Designer Receptor Exclusively Activated by Designer Drugs (DREADD). Multiday treatment with the DREADD-specific ligand clozapine-N-oxide prevented and enduringly reversed morphine-induced persistent sensitization for weeks to months after cessation of clozapine-N-oxide. These data demonstrate both the critical importance of microglia and that maintenance of chronic pain created by early exposure to opioids can be disrupted, resetting pain to normal. These data also provide strong support for the recent "two-hit hypothesis" of microglial priming, leading to exaggerated reactivity after the second challenge, documented here in the context of nerve injury followed by morphine. This study predicts that prolonged pain is an unrealized and clinically concerning consequence of the abundant use of opioids in chronic pain.


Subject(s)
Chronic Pain/metabolism , Inflammasomes/metabolism , Microglia/metabolism , Morphine/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuralgia/metabolism , Spinal Cord Dorsal Horn/metabolism , Animals , Chronic Pain/pathology , Chronic Pain/physiopathology , Clozapine/analogs & derivatives , Clozapine/pharmacology , Interleukin-1beta/metabolism , Male , Microglia/pathology , Neuralgia/pathology , Neuralgia/physiopathology , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Spinal Cord Dorsal Horn/pathology , Spinal Cord Dorsal Horn/physiopathology
15.
Brain Behav Immun ; 48: 115-22, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25774010

ABSTRACT

Despite dramatic improvement in cardiopulmonary resuscitation (CPR) and other techniques for cardiac arrest (CA), the majority of survivors continue to show signs of decreased memory or executive cognitive function. Such memory impairment may be due to hippocampal CA1 neuronal death, which is delayed by several days after CA/CPR. Classical microgliosis in the CA1 region may contribute to neuronal death, yet the role of a key activation receptor Toll Like Receptor 4 (TLR4) has not been previously investigated for such neuronal death after CA/CPR. We show that (+)-naltrexone was neuroprotective after CA/CPR. TLR4 blockade was associated with decreased expression of markers for microglial/macrophage activation and T cell and B cell infiltration, as well as decreased pro-inflammatory cytokine levels. Notably, IL-10 expression was elevated in response to CA/CPR, but was not attenuated by (+)-naltrexone, suggesting that the local monocyte/microglial phenotype had shifted towards alternative activation. This was confirmed by elevated expression of Arginase-1, and decreased expression of NFκB p65 subunit. Thus, (+)-naltrexone and other TLR4 antagonists may represent a novel therapeutic strategy to alleviate the substantial burden of memory or executive cognitive function impairment after CA/CPR.


Subject(s)
Cell Death/drug effects , Heart Arrest/pathology , Hippocampus/drug effects , Naltrexone/pharmacology , Neuroprotective Agents/pharmacology , Animals , Cardiopulmonary Resuscitation , Heart Arrest/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Interleukin-10/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology
16.
Brain Behav Immun ; 46: 50-4, 2015 May.
Article in English | MEDLINE | ID: mdl-25653191

ABSTRACT

A single intrathecal dose of adenosine 2A receptor (A2AR) agonist was previously reported to produce a multi-week reversal of allodynia in two different models of neuropathic pain in addition to downregulating glial activation markers in the spinal cord. We aimed to determine whether a single intrathecal administration of an A2AR agonist was able to attenuate motor symptoms induced by experimental autoimmune encephalopathy. Two A2AR agonists (CGS21680 and ATL313) significantly attenuated progression of motor symptoms following a single intrathecal administration at the onset of motor symptoms. OX-42, a marker of microglial activation, was significantly attenuated in the lumbar spinal cord following A2AR administration compared to vehicle. Therefore, A2AR agonists attenuate motor symptoms of EAE by acting on A2AR in the spinal cord.


Subject(s)
Adenosine A2 Receptor Agonists/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Paralysis/drug therapy , Spinal Cord/drug effects , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine/therapeutic use , Adenosine A2 Receptor Agonists/pharmacology , Animals , Male , Microglia/drug effects , Phenethylamines/pharmacology , Phenethylamines/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , Rats
17.
J Pain ; 15(2): 121-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24287315

ABSTRACT

UNLABELLED: Decreased voluntary wheel running has recently been proposed as a preclinical pain measure for inflammatory pain, but whether this reflects pain evoked by use of the affected limbs is unknown. To assess the role of inflammation site as a determinant of this measure, complete Freund's adjuvant (CFA), formalin, or equivolume vehicle was subcutaneously injected into the plantar surface of the hind paws (bilateral) or L1 dorsum dermatome (leaving paws unaffected) of male Sprague Dawley rats. CFA-induced hind paw mechanical allodynia (P < .001) did not correlate with reduced voluntary wheel running. Intraplantar formalin did not attenuate voluntary running, despite eliciting robust licking/writhing/flinching behavior and hind paw mechanical allodynia (P < .001). Subcutaneous L1 dorsum dermatome formalin, but not CFA, induced licking/writhing/flinching behavior (P < .001), but neither induced hind paw mechanical allodynia or attenuated voluntary running. That voluntary running is decreased by hind paw CFA, but not by L1 dorsum CFA, implies that the behavior is a measure of CFA-induced pain evoked by use of the affected limbs rather than supraspinal pain processing that is independent of inflammation site. Furthermore, the results suggest that interpretation of voluntary wheel running data cannot simply be explained by correlation with mechanical allodynia. PERSPECTIVE: Whether decreased voluntary running is dependent on inflammation site is unknown. We show that intraplantar, but not L1 dorsum, CFA suppressed voluntary running and formalin-induced licking/writhing/flinching behavior but had no effect on voluntary running. These data suggest that suppressed voluntary running by CFA likely reflects pain evoked by use of the affected limbs.


Subject(s)
Hindlimb/physiopathology , Hyperalgesia/etiology , Hyperalgesia/physiopathology , Inflammation/physiopathology , Motor Activity , Animals , Formaldehyde , Freund's Adjuvant , Hyperalgesia/diagnosis , Lumbosacral Region/physiology , Male , Pain/diagnosis , Pain/etiology , Pain/physiopathology , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Time Factors , Touch
18.
Brain Behav Immun ; 33: 112-22, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23811314

ABSTRACT

A single intrathecal dose of adenosine 2A receptor (A2AR) agonist was previously reported to produce a multi-week reversal of allodynia in a chronic constriction injury (CCI) model of neuropathic pain. We aimed to determine if this long-term reversal was induced by A2AR agonism versus more generalized across adenosine receptor subtypes, and begin to explore the intracellular signaling cascades involved. In addition, we sought to identify whether the enduring effect could be extended to other models of neuropathic pain. We tested an A1R and A2BR agonist in CCI and found the same long duration effect with A2BR but not A1R agonism. An A2AR agonist (ATL313) produced a significant long-duration reversal of mechanical allodynia induced by long established CCI (administered 6 weeks after surgery), spinal nerve ligation and sciatic inflammatory neuropathy. To determine if ATL313 had a direct effect on glia, ATL313 was coadministered with lipopolysaccharide to neonatal microglia and astrocytes in vitro. ATL313 significantly attenuated TNFα production in both microglia and astrocytes but had no effect on LPS induced IL-10. Protein kinase C significantly reversed the ATL313 effects on TNFα in vitro in microglia and astrocytes, while a protein kinase A inhibitor only effected microglia. Both intrathecal PKA and PKC inhibitors significantly reversed the effect of the A2AR agonist on neuropathic allodynia. Therefore, A2AR agonists administered IT remain an exciting novel target for the treatment of neuropathic pain.


Subject(s)
Adenosine A2 Receptor Agonists/therapeutic use , Cyclic AMP-Dependent Protein Kinases/physiology , Hyperalgesia/metabolism , Protein Kinase C/physiology , Sciatic Neuropathy/drug therapy , Sciatic Neuropathy/enzymology , Signal Transduction/immunology , Adenosine A2 Receptor Agonists/administration & dosage , Animals , Cells, Cultured , Chronic Disease , Constriction, Pathologic/drug therapy , Constriction, Pathologic/enzymology , Constriction, Pathologic/pathology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Hyperalgesia/enzymology , Hyperalgesia/pathology , Inflammation/drug therapy , Inflammation/enzymology , Inflammation/pathology , Injections, Spinal , Ligation , Male , Piperidines/administration & dosage , Piperidines/therapeutic use , Protein Kinase C/antagonists & inhibitors , Random Allocation , Rats , Rats, Sprague-Dawley , Sciatic Neuropathy/pathology , Signal Transduction/drug effects
19.
FASEB J ; 27(7): 2713-22, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23568774

ABSTRACT

Rifampin has been used for the treatment of bacterial infections for many years. Clinically, rifampin has been found to possess immunomodulatory effects. However, the molecular target responsible for the immunosuppressive effects of rifampin is not known. Herein, we show that rifampin binds to myeloid differentiation protein 2 (MD-2), the key coreceptor for innate immune TLR4. Rifampin blocked TLR4 signaling induced by LPS, including NF-κB activation and the overproduction of proinflammatory mediators nitric oxide, interleukin 1ß, and tumor necrosis factor α in mouse microglia BV-2 cells and macrophage RAW 264.7 cells. Rifampin's inhibition of TLR4 signaling was also observed in immunocompetent rat primary macrophage, microglia, and astrocytes. Further, we show that rifampin (75 or 100 mg/kg b.i.d. for 3 d, intraperitoneal) suppressed allodynia induced by chronic constriction injury of the sciatic nerve and suppressed nerve injury-induced activation of microglia. Our findings indicate that MD-2 is a important target of rifampin in its inhibition of innate immune function and contributes to its clinically observed immune-suppressive effect. The results also suggest that rifampin may be repositioned as an agent for the treatment of neuropathic pain.


Subject(s)
Lymphocyte Antigen 96/metabolism , Neuralgia/prevention & control , Rifampin/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Animals , Animals, Newborn , Blotting, Western , Cell Line , Cells, Cultured , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Hyperalgesia/prevention & control , Interleukin-1beta/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , NF-kappa B/metabolism , Neuralgia/metabolism , Neuralgia/physiopathology , Nitric Oxide/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Rifampin/administration & dosage , Rifampin/metabolism , Tumor Necrosis Factor-alpha/metabolism
20.
J Pain ; 13(12): 1162-71, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23182225

ABSTRACT

UNLABELLED: Alpha-7 nicotinic acetylcholine receptor (α7 nAChR) agonists attenuate pain and inflammation in preclinical models. This study tested whether systemic delivery of an α7 nAChR agonist attenuates neuropathic pain and associated immune-mediated pro-inflammation. Hind paw response thresholds to mechanical stimuli in male Sprague Dawley rats were assessed before and after sciatic chronic constriction injury (CCI) or sham surgery. Osmotic mini-pumps containing TC-7020, an α7 nAChR selective agonist, were implanted 10 to 14 days after surgery. TC-7020 (1, 3, and 10 mg/kg/d; s.c.) significantly attenuated CCI-induced allodynia, which lasted through 2 weeks of test compound administration. Spinal cords were collected after 2 weeks and processed for microglial and astrocyte activation markers within the ipsilateral L4-L6 dorsal horn. In addition, ipsilateral L4-5 dorsal root ganglia (DRGs) were processed for neuronal injury and satellite cell activation markers. CCI-induced central glial cell activation markers were not suppressed by TC-7020, even though TC-7020 is mildly blood-brain barrier permeable. However, TC-7020 downregulated the integrated density of activation transcription factor 3 (ATF3) but not the number of ATF positive cells. TC-7020 also downregulated phosphorylated extracellular signal kinase (p-ERK) and satellite cell activation in the CCI-affected DRGs. Therefore, systemic α7 nAChR agonist may be effective in treating neuropathic pain via reducing neuronal injury and immune cells activation occurring in the periphery. PERSPECTIVE: These studies demonstrated that TC-7020, an alpha7 nicotinic acetylcholine receptor agonist with partial blood-brain barrier permeability, reversed neuropathic pain in rats, likely via attenuation of inflammation in the DRG and/or the site of sciatic injury.


Subject(s)
Neuralgia/drug therapy , Nicotinic Agonists/administration & dosage , Quinuclidines/administration & dosage , Receptors, Nicotinic/physiology , Thiophenes/administration & dosage , Animals , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Male , Neuralgia/metabolism , Neuralgia/pathology , Rats , Rats, Sprague-Dawley , alpha7 Nicotinic Acetylcholine Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...