Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
PLoS One ; 19(2): e0298396, 2024.
Article in English | MEDLINE | ID: mdl-38330029

ABSTRACT

Chemotherapy is often a life-saving treatment, but the development of intractable pain caused by chemotherapy-induced peripheral neuropathy (CIPN) is a major dose-limiting toxicity that restricts cancer survival rates. Recent reports demonstrate that paclitaxel (PTX) robustly increases anti-inflammatory CD4+ T cells in the dorsal root ganglion (DRG), and that T cells and anti-inflammatory cytokines are protective against CIPN. However, the mechanism by which CD4+ T cells are activated, and the extent cytokines released by CD4+ T cells target DRG neurons are unknown. Here, we are the first to detect major histocompatibility complex II (MHCII) protein in mouse DRG neurons and to find CD4+ T cells breaching the satellite glial cell barrier to be in close proximity to neurons, together suggesting CD4+ T cell activation and targeted cytokine release. MHCII protein is primarily expressed in small nociceptive neurons in male and female mouse DRG but increased after PTX in small nociceptive neurons in only female DRG. Reducing one copy of MHCII in small nociceptive neurons decreased anti-inflammatory IL-10 and IL-4 producing CD4+ T cells in naïve male DRG and increased their hypersensitivity to cold. Administration of PTX to male and female mice that lacked one copy of MHCII in nociceptive neurons decreased anti-inflammatory CD4+ T cells in the DRG and increased the severity of PTX-induced cold hypersensitivity. Collectively, our results demonstrate expression of MHCII protein in mouse DRG neurons, which modulates cytokine producing CD4+ T cells in the DRG and attenuates cold hypersensitivity during homeostasis and after PTX treatment.


Subject(s)
Cryopyrin-Associated Periodic Syndromes , Paclitaxel , Peripheral Nervous System Diseases , Rats , Mice , Male , Female , Animals , Paclitaxel/toxicity , Paclitaxel/metabolism , Ganglia, Spinal/metabolism , Hyperalgesia/etiology , Rats, Sprague-Dawley , Peripheral Nervous System Diseases/chemically induced , Cytokines/metabolism , Neurons/metabolism , Anti-Inflammatory Agents/therapeutic use
2.
Pain ; 165(4): 725-726, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-37975869

Subject(s)
Immunotherapy , Pain , Humans
3.
bioRxiv ; 2023 Sep 21.
Article in English | MEDLINE | ID: mdl-37066176

ABSTRACT

Chemotherapy is often a life-saving treatment, but the development of intractable pain caused by chemotherapy-induced peripheral neuropathy (CIPN) is a major dose-limiting toxicity that restricts survival rates. Recent reports demonstrate that paclitaxel (PTX) robustly increases anti-inflammatory CD4+ T cells in the dorsal root ganglion (DRG), and that T cells and anti-inflammatory cytokines are protective against CIPN. However, the mechanism by which CD4+ T cells are activated, and the extent cytokines released by CD4+ T cells target DRG neurons are unknown. Here, we found novel expression of functional major histocompatibility complex II (MHCII) protein in DRG neurons, and CD4+ T cells in close proximity to DRG neurons, together suggesting CD4+ T cell activation and targeted cytokine release. MHCII protein is primarily expressed in small nociceptive neurons in male mouse DRG regardless of PTX, while MHCII is induced in small nociceptive neurons in female DRG after PTX. Accordingly, reducing MHCII in small nociceptive neurons increased hypersensitivity to cold only in naive male mice, but increased severity of PTX-induced cold hypersensitivity in both sexes. Collectively, our results demonstrate expression of MHCII on DRG neurons and a functional role during homeostasis and inflammation.

4.
J Neuroimmunol ; 367: 577878, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35509138

ABSTRACT

Chemotherapy is often dose limiting due to the emergence of a debilitating neuropathy. IL-10 and IL-4 are protective against peripheral neuropathy, yet the contribution by CD4+ T cells is unknown. Using flow cytometry, we found that naïve females had a greater frequency of anti-inflammatory CD4+ T cells in the dorsal root ganglion (DRG) compared to males. In response to paclitaxel, females had reduced mechanical hypersensitivity and a greater frequency of anti-inflammatory CD4+ T cells (FoxP3, IL-10, IL-4) in the DRG than male and ovariectomized female mice. These findings support a model in which estrogen promotes anti-inflammatory CD4+ T cells in female DRG to suppress peripheral neuropathy.


Subject(s)
Ganglia, Spinal , Peripheral Nervous System Diseases , Animals , Anti-Inflammatory Agents/therapeutic use , CD4-Positive T-Lymphocytes , Female , Humans , Interleukin-10/therapeutic use , Interleukin-4 , Male , Mice , Ovariectomy , Paclitaxel/toxicity , Peripheral Nervous System Diseases/chemically induced , Rats
5.
J Neurosci ; 41(13): 2883-2898, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33593853

ABSTRACT

Gαs-coupled receptors signaling through cAMP provide a key mechanism for the sensitization of nociceptive sensory neurons, and the cAMP effector Epac has been implicated in the transition from acute to chronic pain. Epac exerts its effects through Rap1 and protein kinase C (PKC). To identify targets of Epac-PKC signaling in sensory neurons of the mouse dorsal root ganglion (DRG), we profiled PKC substrate proteins phosphorylated in response to the activation of Epac with the proinflammatory prostaglandin E2 (PGE2). A prominent Epac-dependent phospho-protein band induced by PGE2 was identified by mass spectrometry as the mitochondrial enzyme pyruvate dehydrogenase (Pdha1). In dissociated DRG from both males and females, the recruitment of Pdha1 to phospho-protein fractions was rapidly induced by PGE2 and prevented by selective inhibition of Epac2. Epac activation increased mitochondrial respiration, consistent with an increase in Pdha1 function mediated by Epac2. Hindpaw injection of PGE2 induced heat hyperalgesia in males and females, but Pdha1 phosphorylation occurred only in males. Hyperalgesia was attenuated in males but not in females by systemic inhibition of Epac2, and also by a mitochondrial membrane potential uncoupler, dinitrophenol, supporting a role for mitochondrial regulation in acute hyperalgesia. These findings identify a mechanism for the regulation of mitochondrial function by Epac2 that contributes to acute inflammatory hyperalgesia in male mice. Systemic administration of the cyclooxygenase 2 inhibitor celecoxib suppressed both PGE2-induced heat hyperalgesia and Pdha1 phosphorylation in DRG of males but not females, suggesting that prostaglandin synthesis within the DRG mediates the phosphorylation of Pdha1 in response to hindpaw insult.SIGNIFICANCE STATEMENT There has been extensive investigation of mitochondrial dysfunction as a causative factor in neuropathic pain disorders. In contrast, results reported here implicate enhanced mitochondrial function as a contributing factor in the development of acute inflammatory hyperalgesia. We describe a mechanism in which Epac2 activation by prostaglandin receptors leads to phosphorylation of pyruvate dehydrogenase and an increase in mitochondrial respiration in peripheral sensory neurons. Although Epac2 activation leads to Pdha1 (pyruvate dehydrogenase) phosphorylation in dissociated neurons from mice of both sexes, induction of this pathway in vivo by hindpaw insult is restricted to males and appears to require intraganglionic prostaglandin synthesis. These findings support a model in which Gs-coupled receptor modulation of mitochondrial function promotes acute nociceptive signaling and inflammatory hyperalgesia.


Subject(s)
Ganglia, Spinal/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Hyperalgesia/metabolism , Mitochondria/metabolism , Pain Measurement/methods , Animals , Enzyme Inhibitors/pharmacology , Female , Ganglia, Spinal/drug effects , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Nociceptors/metabolism , Pain Measurement/drug effects , Pyruvate Dehydrogenase (Lipoamide)/antagonists & inhibitors , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism
6.
Mol Cell Biochem ; 456(1-2): 167-178, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30739223

ABSTRACT

Exchange protein directly activated by cAMP (Epac) and protein kinase A are effectors for cAMP with distinct actions and regulatory mechanisms. Epac is a Rap guanine nucleotide exchange factor that activates Rap1; protein kinase C (PKC) is a major downstream target of Epac-Rap1 signaling that has been implicated in a variety of pathophysiological processes, including cardiac hypertrophy, cancer, and nociceptor sensitization leading to chronic pain. Despite the implication of both Epac and PKC in these processes, few downstream targets of Epac-PKC signaling have been identified. This study characterized the regulation of PKC activity downstream of Epac activation. Using an antibody that recognizes phospho-serine residues within the consensus sequence phosphorylated by PKC, we analyzed the 1-dimensional banding profile of PKC substrate protein phosphorylation from the Neuro2A mouse neuroblastoma cell line. Activation of Epac either indirectly by prostaglandin PGE2, or directly by 8-pCPT-2-O-Me-cAMP-AM (8pCpt), produced distinct PKC phospho-substrate protein bands that were suppressed by co-administration of the Epac inhibitor ESI09. Different PKC isoforms contributed to the induction of individual phospho-substrate bands, as determined using isoform-selective PKC inhibitors. Moreover, the banding profile after Epac activation was altered by disruption of the cytoskeleton, suggesting that the orchestration of Epac-dependent PKC signaling is regulated in part by interactions with the cytoskeleton. The approach described here provides an effective means to characterize Epac-dependent PKC activity.


Subject(s)
Cytoskeleton/enzymology , Dinoprostone/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Protein Kinase C/metabolism , Signal Transduction , rap1 GTP-Binding Proteins/metabolism , Animals , Cell Line, Tumor , Cytoskeleton/genetics , Guanine Nucleotide Exchange Factors/genetics , Mice , Phosphorylation , Protein Kinase C/genetics , rap1 GTP-Binding Proteins/genetics
7.
Immunity ; 34(5): 729-40, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21530327

ABSTRACT

CD28 and CTLA-4 are cell surface cosignaling molecules essential for the control of T cell activation upon the engagement of their ligands B7-1 and B7-2 from antigen-presenting cells. By employing a receptor array assay, we have demonstrated that B7-H2, best known as the ligand of inducible costimulator, was a ligand for CD28 and CTLA-4 in human, whereas these interactions were not conserved in mouse. B7-H2 and B7-1 or B7-2 interacted with CD28 through distinctive domains. B7-H2-CD28 interaction was essential for the costimulation of human T cells' primary responses to allogeneic antigens and memory recall responses. Similar to B7-1 and B7-2, B7-H2 costimulation via CD28 induced survival factor Bcl-xL, downregulated cell cycle inhibitor p27(kip1), and triggered signaling cascade of ERK and AKT kinase-dependent pathways. Our findings warrant re-evaluation of CD28 and CTLA-4's functions previously attributed exclusively to B7-1 and B7-2 and have important implications in therapeutic interventions against human diseases.


Subject(s)
Antigens, CD/immunology , CD28 Antigens/immunology , Animals , Antigens, CD/chemistry , Antigens, Differentiation, T-Lymphocyte/immunology , Binding Sites , CD28 Antigens/chemistry , CTLA-4 Antigen , Cell Line , Cell Proliferation , Humans , Inducible T-Cell Co-Stimulator Ligand , Inducible T-Cell Co-Stimulator Protein , Ligands , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Structure, Quaternary , T-Lymphocytes/cytology , T-Lymphocytes/immunology
8.
Proc Natl Acad Sci U S A ; 106(49): 20877-82, 2009 Dec 08.
Article in English | MEDLINE | ID: mdl-19933330

ABSTRACT

Both activated and resting CD4(+) T cells in mucosal tissues play important roles in the earliest phases of infection after sexual transmission of HIV-1, a process that is inefficient. HIV-1 gp120 binds to integrin alpha(4)beta(7) (alpha(4)beta(7)), the gut mucosal homing receptor. We find that alpha(4)beta(7)(high) CD4(+) T cells are more susceptible to productive infection than are alpha(4)beta(7)(low-neg) CD4(+) T cells in part because this cellular subset is enriched with metabolically active CD4(+) T cells. alpha(4)beta(7)(high) CD4(+) T cells are CCR5(high) and CXCR4(low); on these cells, alpha(4)beta(7) appears in a complex with CD4. The specific affinity of gp120 for alpha(4)beta(7) provides a mechanism for HIV-1 to target activated cells that are critical for efficient virus propagation and dissemination following sexual transmission.


Subject(s)
CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , Integrins/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/virology , Antibodies, Monoclonal/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Cell Membrane/drug effects , Cell Membrane/immunology , Cell Membrane/virology , Female , Genitalia, Female/drug effects , Genitalia, Female/immunology , HIV-1/drug effects , HIV-1/physiology , Humans , Immunoprecipitation , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Receptors, CCR5/metabolism , T-Lymphocyte Subsets/drug effects , Virus Replication/drug effects
9.
Nat Immunol ; 9(3): 301-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18264102

ABSTRACT

Infection with human immunodeficiency virus 1 (HIV-1) results in the dissemination of virus to gut-associated lymphoid tissue. Subsequently, HIV-1 mediates massive depletion of gut CD4+ T cells, which contributes to HIV-1-induced immune dysfunction. The migration of lymphocytes to gut-associated lymphoid tissue is mediated by integrin alpha4beta7. We demonstrate here that the HIV-1 envelope protein gp120 bound to an activated form of alpha4beta7. This interaction was mediated by a tripeptide in the V2 loop of gp120, a peptide motif that mimics structures presented by the natural ligands of alpha4beta7. On CD4+ T cells, engagement of alpha4beta7 by gp120 resulted in rapid activation of LFA-1, the central integrin involved in the establishment of virological synapses, which facilitate efficient cell-to-cell spreading of HIV-1.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Envelope Protein gp120/metabolism , HIV Infections/immunology , HIV-1/immunology , Integrins/metabolism , Intestinal Mucosa/immunology , CD4-Positive T-Lymphocytes/virology , Cell Movement/immunology , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/virology , Flow Cytometry , Humans , Intestinal Mucosa/virology , Killer Cells, Natural/immunology , Ligands , Lymphocyte Function-Associated Antigen-1/metabolism , Protein Binding/immunology , Signal Transduction/immunology
10.
Proc Natl Acad Sci U S A ; 104(9): 3396-401, 2007 Feb 27.
Article in English | MEDLINE | ID: mdl-17360657

ABSTRACT

Plasmacytoid dendritic cells (pDCs) play a central role in innate and adaptive immune responses against viral infections. pDCs secrete type I IFNs and proinflammatory cytokines upon stimulation by either TLR7 or TLR9. Throughout the course of HIV infection, the production of type-I IFNs is profoundly impaired, and total pDC cell counts in peripheral blood correlates inversely with viral load and positively with CD4(+) T cell count. The origin of these defects is unclear. pDCs express CD4, CCR5, and CXCR4, the primary receptor and coreceptors, respectively, for the HIV envelope; yet little is known concerning the effects of the viral envelope on these cells. Here, we show that exposure of pDCs to gp120 results in the suppression of activation of these cells. This suppression is specific for TLR9-mediated responses, because TLR7-mediated responses are unaffected by gp120. gp120 also suppressed TLR9-mediated induction of proinflammatory cytokines and expression of CD83, a marker of DC activation. Finally, gp120 suppressed pDC-induced cytolytic activity of natural killer cells. Taken together, these data demonstrate that the direct interaction of HIV-1 gp120 with pDCs interferes with TLR9 activation resulting in a decreased ability of pDCs to secrete antiviral and inflammatory factors that play a central role in initiating host immune responses against invading pathogens.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , HIV Envelope Protein gp120/metabolism , HIV-1/metabolism , Immunity, Cellular/immunology , Interferon-alpha/metabolism , Toll-Like Receptor 9/antagonists & inhibitors , Antigens, CD/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulins/metabolism , Killer Cells, Natural/immunology , Membrane Glycoproteins/metabolism , CD83 Antigen
SELECTION OF CITATIONS
SEARCH DETAIL
...