1
|
Silva A, Prior R, D'Antonio M, Swinnen JV, Van Den Bosch L. Lipid metabolism alterations in peripheral neuropathies. Neuron 2025:S0896-6273(25)00262-4. [PMID: 40311611 DOI: 10.1016/j.neuron.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/14/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025]
Abstract
Alterations in lipid metabolism are increasingly recognized as central pathological hallmarks of inherited and acquired peripheral neuropathies. Correct lipid balance is critical for cellular homeostasis. However, the mechanisms linking lipid disturbances to cellular dysfunction and whether these changes are primary drivers or secondary effects of disease remain unresolved. This is particularly relevant in the peripheral nervous system, where the lipid-rich myelin integrity is critical for axonal function, and even subtle perturbations can cause widespread effects. This review explores the role of lipids as structural components as well as signaling molecules, emphasizing their metabolic role in peripheral neurons and Schwann cells. Additionally, we explore the genetic and environmental connections in both inherited and acquired peripheral neuropathies, respectively, which are known to affect lipid metabolism in peripheral neurons or Schwann cells. Overall, we highlight how understanding lipid-centric mechanisms could advance biomarker discovery and therapeutic interventions for peripheral nerve disorders.
Collapse
Affiliation(s)
- Alessio Silva
- KU Leuven, University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.
| | - Robert Prior
- KU Leuven, University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium; Department of Ophthalmology, University Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Maurizio D'Antonio
- Biology of Myelin Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Johannes V Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Ludo Van Den Bosch
- KU Leuven, University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.
| |
Collapse
|
2
|
Warda M, Tekin S, Gamal M, Khafaga N, Çelebi F, Tarantino G. Lipid rafts: novel therapeutic targets for metabolic, neurodegenerative, oncological, and cardiovascular diseases. Lipids Health Dis 2025; 24:147. [PMID: 40247292 PMCID: PMC12004566 DOI: 10.1186/s12944-025-02563-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
Lipid rafts are specialized microdomains within cellular membranes enriched with cholesterol and sphingolipids that play key roles in cellular organization, signaling, and homeostasis. This review highlights their involvement in protein clustering, energy metabolism, oxidative stress responses, inflammation, autophagy, and apoptosis. These findings clarify their influence on signaling, trafficking, and adhesion while interacting with the extracellular matrix, cytoskeleton, and ion channels, making them pivotal in the progression of various diseases. This review further addresses their contributions to immune responses, including autoimmune diseases, chronic inflammation, and cytokine storms. Additionally, their role as entry points for pathogens has been demonstrated, with raft-associated receptors being exploited by viruses and bacteria to increase infectivity and evade immune defenses. Disruptions in lipid raft dynamics are linked to oxidative stress and cellular signaling defects, which contribute to metabolic, neurodegenerative, and cardiovascular diseases. This review underscores their potential as therapeutic targets, discussing innovations such as engineered lipid raft transplantation. Advances in analytical techniques such as mass spectrometry have expanded our understanding of lipid raft composition and dynamics, opening new directions for research. By consolidating the current insights, we highlight the therapeutic potential of lipid rafts and highlight the need for further exploration of their molecular mechanisms.
Collapse
Affiliation(s)
- Mohamad Warda
- Department of Physiology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
- Department of Biochemistry, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt.
| | - Samet Tekin
- Department of Physiology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Mahmoud Gamal
- Department of Biochemistry, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Nagwa Khafaga
- Food Hygiene Department, Animal Health Research Institute (AHRI), Agricultural Research Center (ARC), Dokki, Egypt
| | - Fikret Çelebi
- Department of Physiology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Giovanni Tarantino
- Department of Clinical Medicine and Surgery, Federico II University Medical School of Naples, Naples, Italy.
| |
Collapse
|
3
|
Borges Paes Lemes J, Panichkina A, Franco Malange K, Morado-Urbina CE, Dochnal SA, Jadhav S, Dolmat M, Pagliusi M, Navia-Pealez JM, Corr M, Miller YI, Yaksh TL. Chronic Pain Induced by Social Defeat Stress in Juvenile Mice Depends on TLR4. Cells 2025; 14:350. [PMID: 40072079 PMCID: PMC11898947 DOI: 10.3390/cells14050350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/07/2025] [Accepted: 02/18/2025] [Indexed: 03/15/2025] Open
Abstract
A significant portion of adolescents suffer from mental illnesses and persistent pain due to repeated stress. The components of the nervous system that link stress and pain in early life remain unclear. Prior studies in adult mice implicated the innate immune system, specifically Toll-like receptors (TLRs), as critical for inducing long-term anxiety and pain-like behaviors in social defeat stress (SDS) models. In this work, we investigated the pain and anxiety behavioral phenotypes of wild-type and TLR4-deficient juvenile mice subjected to repeated SDS and evaluated the engagement of TLR4 by measuring dimerization in the spinal cord, dorsal root ganglia, and prefrontal cortex. Male juvenile (4-week-old) mice (C57BL/6J or Tlr4-/-) underwent six social defeat sessions with adult aggressor (CD1) mice. In WT mice, SDS promotes chronic mechanical allodynia and thermal hyperalgesia assessed via von Frey testing and the Hargreaves test, respectively. In parallel, the stressed WT mice exhibited transient anxiety-like behavior and long-lasting locomotor activity reduction in the open-field test. Tlr4-/--stressed animals were resistant to the induction of pain-like behavior but had a remnant of anxious behavior, spending less time in the center of the arena. In WT SDS, there were concordant robust increases in TLR4 dimerization in dorsal root ganglia macrophages and spinal cord microglia, indicating TLR4 activation. These results suggest that the chronic pain phenotype and locomotor impairment induced by SDS in juvenile mice depends on TLR4 engagement evidenced by dimerization in immune cells of the dorsal root ganglia and spinal cord.
Collapse
Affiliation(s)
- Julia Borges Paes Lemes
- Department of Anesthesiology, University of California, San Diego, CA 92093, USA; (A.P.); (K.F.M.); (C.E.M.-U.); (S.A.D.); (S.J.); (T.L.Y.)
| | - Alisa Panichkina
- Department of Anesthesiology, University of California, San Diego, CA 92093, USA; (A.P.); (K.F.M.); (C.E.M.-U.); (S.A.D.); (S.J.); (T.L.Y.)
| | - Kaue Franco Malange
- Department of Anesthesiology, University of California, San Diego, CA 92093, USA; (A.P.); (K.F.M.); (C.E.M.-U.); (S.A.D.); (S.J.); (T.L.Y.)
| | - Carlos E. Morado-Urbina
- Department of Anesthesiology, University of California, San Diego, CA 92093, USA; (A.P.); (K.F.M.); (C.E.M.-U.); (S.A.D.); (S.J.); (T.L.Y.)
| | - Sara Anna Dochnal
- Department of Anesthesiology, University of California, San Diego, CA 92093, USA; (A.P.); (K.F.M.); (C.E.M.-U.); (S.A.D.); (S.J.); (T.L.Y.)
| | - Saee Jadhav
- Department of Anesthesiology, University of California, San Diego, CA 92093, USA; (A.P.); (K.F.M.); (C.E.M.-U.); (S.A.D.); (S.J.); (T.L.Y.)
| | - Maksim Dolmat
- Department of Chemical and Nano Engineering, University of California, San Diego, CA 92093, USA;
| | - Marco Pagliusi
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Sao Paulo 14049-900, Brazil;
| | - Juliana M. Navia-Pealez
- Department of Pharmacology and Physiology, Saint Louis University, Saint Louis, MO 63104, USA;
| | - Maripat Corr
- Department of Medicine, University of California, San Diego, CA 92093, USA; (M.C.); (Y.I.M.)
| | - Yury I. Miller
- Department of Medicine, University of California, San Diego, CA 92093, USA; (M.C.); (Y.I.M.)
| | - Tony L. Yaksh
- Department of Anesthesiology, University of California, San Diego, CA 92093, USA; (A.P.); (K.F.M.); (C.E.M.-U.); (S.A.D.); (S.J.); (T.L.Y.)
| |
Collapse
|
4
|
Li Y, Uhelski ML, North RY, Farson LB, Bankston CB, Roland GH, Fan DH, Sheffield KN, Jia A, Orlando D, Heles M, Yaksh TL, Miller YI, Kosten TA, Dougherty PM. ApoA-I binding protein (AIBP) regulates transient receptor potential vanilloid 1 (TRPV1) activity in rat dorsal root ganglion neurons by selective disruption of toll-like receptor 4 (TLR4)-lipid rafts. Brain Behav Immun 2025; 123:644-655. [PMID: 39414176 DOI: 10.1016/j.bbi.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/24/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024] Open
Abstract
Toll-like receptor 4 (TLR4) and the transient receptor potential vanilloid subtype 1 (TRPV1) are both upregulated and play key roles in the induction and expression of paclitaxel-related chemotherapy-induced peripheral neuropathy (CIPN). Using Apolipoprotein A-I binding protein, non-specific cholesterol depletion, TLR4 mis-sense rats and a TLR4 inhibitor, we demonstrate that co-localization of TRPV1 with TLR4 to cholesterol-rich lipid membrane rafts in nociceptors is essential for its normal activation as well as for its exaggerated activation that underlies the development and expression of CIPN. The findings suggest that TLR4-lipid rafts may have an essential role in numerous neuroinflammatory and neuropathic pain conditions. This mechanism is also generalized to female rats for the first time.
Collapse
Affiliation(s)
- Yan Li
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, the United States of America
| | - Megan L Uhelski
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, the United States of America
| | - Robert Y North
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, the United States of America
| | - Luke B Farson
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | - Christopher B Bankston
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | - Gavin H Roland
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | - Dwight H Fan
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | | | - Amy Jia
- Northwestern University, Evanston, IL 60208, the United States of America
| | - Dana Orlando
- The University of Texas Health Science Center, Houston, TX 77030, the United States of America
| | - Mario Heles
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, the United States of America
| | - Tony L Yaksh
- The Department of Anesthesiology, the University of California San Diego, La Jolla, CA, 92093, the United States of America
| | - Yury I Miller
- Department of Medicine, the University of California San Diego, La Jolla, CA, 92093, the United States of America
| | - Therese A Kosten
- Department of Psychology, Health Building 1, 4349 Martin Luther King Blvd, Houston, TX 77204, the United States of America
| | - Patrick M Dougherty
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, the United States of America.
| |
Collapse
|
5
|
Tao Z, Chen Z, Zeng X, Cui J, Quan M. An emerging aspect of cancer neuroscience: A literature review on chemotherapy-induced peripheral neuropathy. Cancer Lett 2024; 611:217433. [PMID: 39736454 DOI: 10.1016/j.canlet.2024.217433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 01/01/2025]
Abstract
The nervous system governs both ontogeny and oncology. Foundational discoveries have clarified the direct communication of neurotransmitters with tumors and indirect interactions through neural effects on the immune system and the tumor microenvironment. Meantime, the nervous system is susceptible to cancer and its treatment. Chemotherapy-induced peripheral neuropathy (CIPN) is the most common side effects that significantly reduce the efficacy of anti-cancer treatment and patients' quality of life by leading to dose reduction or early cessation of chemotherapy. However, there are no effective strategies to reverse or treat CIPN. A better understanding of the mechanisms is expected to enable the development of the next generation of therapies. Here, we summarize the recent important studies on clinical manifestations, risk factors, prediction, pathogenesis, prevention, and treatment of CIPN. We also provide perspectives and insights regarding the rationales of bidirectional interactions between cancer and the nervous system.
Collapse
Affiliation(s)
- Zhirui Tao
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Zhiqin Chen
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Xiaochen Zeng
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Jiujie Cui
- Department of Oncology and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China.
| | - Ming Quan
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China.
| |
Collapse
|
6
|
Dos Santos GG, Jiménez-Andrade JM, Muñoz-Islas E, Candanedo-Quiroz ME, Cardenas AG, Drummond B, Pham P, Stilson G, Hsu CC, Delay L, Navia-Pelaez JM, Lemes JP, Miller YI, Yaksh TL, Corr M. Role of TLR4 activation and signaling in bone remodeling, and afferent sprouting in serum transfer arthritis. Arthritis Res Ther 2024; 26:212. [PMID: 39696684 DOI: 10.1186/s13075-024-03424-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 10/23/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND In the murine K/BxN serum transfer rheumatoid arthritis (RA) model, tactile allodynia persists after resolution of inflammation in male and partially in female wild type (WT) mice, which is absent in Toll-like receptor (TLR)4 deficient animals. We assessed the role of TLR4 on allodynia, bone remodeling and afferent sprouting in this model of arthritis. METHODS K/BxN sera were injected into male and female mice with conditional or stable TLR4 deletion and controls. Paw swelling was scored and allodynia assessed by von Frey filaments. At day 28, synovial neural fibers were visualized with confocal microscopy and bone density assayed with microCT. Microglial activity and TLR4 dimerization in spinal cords were examined by immunofluorescence and flow cytometry. RESULTS In the synovium, K/BxN injected WT male and female mice showed robust increases in calcitonin gene related-peptide (CGRP+), tyrosine hydroxylase (TH)+ and GAP43+ nerve fibers. Trabecular bone density by microCT was significantly decreased in K/BxN WT female but not in WT male mice. The number of osteoclasts increased in both sexes of WT mice, but not in Tlr4-/- K/BxN mice. We used conditional strains with Cre drivers for monocytes/osteoclasts (lysozyme M), microglia (Tmem119 and Cx3CR1), astrocytes (GFAP) and sensory neurons (advillin) for Tlr4f/f disruption. All strains developed similar arthritis scores after K/BxN serum injection with the exception being the Tlr4Tmem119 mice which showed a reduction. Both sexes of Tlr4Lyz2, Tlr4Tmem119 and Tlr4Cx3cr1 mice displayed a partial reversal of the chronic pain phenotype but not in Tlr4Avil, and Tlr4Gfap mice. WT K/BxN male mice showed increases in spinal Iba1, but not GFAP, compared to Tlr4-/- male mice. To determine whether spinal TLR4 was indeed activated in the K/BxN mice, flow cytometry of lumbar spinal cords of WT K/BxN male mice was performed and revealed that TLR4 in microglia cells (CD11b+ /TMEM119+) demonstrated dimerization (e.g. activation) and a characteristic increase in lipid rafts. CONCLUSION These results demonstrated a complex chronic allodynia phenotype associated with TLR4 in microglia and monocytic cell lineages, and a parallel spinal TLR4 activation. However, TLR4 is dispensable for the development of peripheral nerve sprouting in this model.
Collapse
Affiliation(s)
| | | | - Enriqueta Muñoz-Islas
- Unidad Académica Multidisciplinaria Reynosa-Aztlán, UAT, Reynosa, Tamaulipas, México
| | | | - Andrea Gonzalez Cardenas
- Department of Anesthesiology and Pharmacology, University of California, La Jolla, San Diego, CA, USA
| | - Bronwen Drummond
- Department of Anesthesiology and Pharmacology, University of California, La Jolla, San Diego, CA, USA
| | - Peter Pham
- Department of Medicine, University of California, 9500 Gilman Dr. MC 0663, La Jolla, San Diego, CA, USA
| | - Gwendalynn Stilson
- Department of Medicine, University of California, 9500 Gilman Dr. MC 0663, La Jolla, San Diego, CA, USA
| | - Chao-Chin Hsu
- Department of Medicine, University of California, 9500 Gilman Dr. MC 0663, La Jolla, San Diego, CA, USA
| | - Lauriane Delay
- Department of Anesthesiology and Pharmacology, University of California, La Jolla, San Diego, CA, USA
| | - Juliana M Navia-Pelaez
- Department of Medicine, University of California, 9500 Gilman Dr. MC 0663, La Jolla, San Diego, CA, USA
| | - Julia Paes Lemes
- Department of Anesthesiology and Pharmacology, University of California, La Jolla, San Diego, CA, USA
| | - Yury I Miller
- Department of Medicine, University of California, 9500 Gilman Dr. MC 0663, La Jolla, San Diego, CA, USA
| | - Tony L Yaksh
- Department of Anesthesiology and Pharmacology, University of California, La Jolla, San Diego, CA, USA
| | - Maripat Corr
- Department of Medicine, University of California, 9500 Gilman Dr. MC 0663, La Jolla, San Diego, CA, USA.
| |
Collapse
|
7
|
Zhang WJ, Chen D. Mesenchymal stem cell transplantation plays a role in relieving cancer pain. Front Pharmacol 2024; 15:1483716. [PMID: 39679363 PMCID: PMC11637888 DOI: 10.3389/fphar.2024.1483716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
Tumors can invade, compress, and damage nerves, leading to persistent pain and seriously affecting the quality of life of patients. However, their treatment is challenging. Sensitization of peripheral receptors, abnormal activity of primary sensory neurons, activation of glial cells, enhanced inflammatory responses, and sensory information transmission contribute towards cancer pain. Therefore, considerable attention has been paid to exploring prospective methods to inhibit the occurrence of these factors and relieve cancer pain. Studies on different types of pains have revealed that the transplantation of functionally active cells into the host has the pharmacological effect of producing analgesia. Mesenchymal stem cells (MSCs) can act as small active pumps to reduce the expression of pain-related molecules and produce analgesic effects. Moreover, MSCs can establish complex communication networks with non-tumor and cancer cells in the microenvironment, interact with each other, and can be used as destinations for inflammation and tumor sites, affecting their potential for invasion and metastasis. This emphasizes the key role of MSCs in cancer and pain management. The pain relief mechanisms of MSCs include neuronutrition, neural protection, neural network reconstruction, immune regulation, and improvement of the inflammatory microenvironment around the nerve injury. All of these are beneficial for the recovery of injured or stimulated nerves and the reconstruction of neural function, and play a role in relieving pain. The pain treatment strategy of cell transplantation is to repair injured nerves and produce analgesic pharmacological properties that are different from those of painkillers and other physiotherapies. Although the therapeutic role of MSCs in cancer and pain is in its early stages, the therapeutic value of MSCs for cancer pain has great prospects. Therefore, in this study, we explored the possible mechanism between MSCs and cancer pain, the potential therapeutic role of therapeutic cells in cancer pain, and some problems and challenges.
Collapse
Affiliation(s)
- Wen-Jun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Jiangxi Medical college, Nanchang, China
| | - Dingyi Chen
- Emergency department, The Second Affiliated Hospital, Nanchang University, Jiangxi Medical college, Nanchang, China
| |
Collapse
|
8
|
Handlin LJ, Macchi NL, Dumaire NLA, Salih L, Lessie EN, McCommis KS, Moutal A, Dai G. Membrane lipid nanodomains modulate HCN pacemaker channels in nociceptor DRG neurons. Nat Commun 2024; 15:9898. [PMID: 39548079 PMCID: PMC11568329 DOI: 10.1038/s41467-024-54053-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Cell membranes consist of heterogeneous lipid nanodomains that influence key cellular processes. Using FRET-based fluorescent assays and fluorescence lifetime imaging microscopy (FLIM), we find that the dimension of cholesterol-enriched ordered membrane domains (OMD) varies considerably, depending on specific cell types. Particularly, nociceptor dorsal root ganglion (DRG) neurons exhibit large OMDs. Disruption of OMDs potentiated action potential firing in nociceptor DRG neurons and facilitated the opening of native hyperpolarization-activated cyclic nucleotide-gated (HCN) pacemaker channels. This increased neuronal firing is partially due to an increased open probability and altered gating kinetics of HCN channels. The gating effect on HCN channels is likely due to a direct modulation of their voltage sensors by OMDs. In animal models of neuropathic pain, we observe reduced OMD size and a loss of HCN channel localization within OMDs. Additionally, cholesterol supplementation inhibited HCN channels and reduced neuronal hyperexcitability in pain models. These findings suggest that disturbances in lipid nanodomains play a critical role in regulating HCN channels within nociceptor DRG neurons, influencing pain modulation.
Collapse
Affiliation(s)
- Lucas J Handlin
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Natalie L Macchi
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Nicolas L A Dumaire
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Lyuba Salih
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Erin N Lessie
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Kyle S McCommis
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Aubin Moutal
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Gucan Dai
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA.
| |
Collapse
|
9
|
Kim L, Nan G, Kim HY, Cha M, Lee BH. Modulation of chemotherapy-induced peripheral neuropathy by JZL195 through glia and the endocannabinoid system. Biomed Pharmacother 2024; 180:117515. [PMID: 39362070 DOI: 10.1016/j.biopha.2024.117515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/12/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) used to treat cancer, is a significant side effect with a complex pathophysiology, and its mechanisms remain unclear. Recent research highlights neuroinflammation, which is modulated by the endocannabinoid system (ECS) and associated with glial activation, and the role of toll-like receptor 4 (TLR4) in CIPN. This study aimed to investigate the effects of JZL195, an inhibitor of fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL), and explore the connection between cannabinoid receptors and TLR4 in glial cells. A CIPN animal model was developed using cisplatin-injected male C57BL/6 mice. Mechanical and cold allodynia were assessed through von Frey and acetone tests. Western blot analysis was used to examine the expression of catabolic enzymes, cannabinoid receptors, glial cells, and neuroinflammatory factors in the dorsal root ganglia (DRGs) and spinal cord. Immunohistochemistry was used to investigate the colocalization of cannabinoid receptors and TLR4 in glial cells. JZL195 alleviated pain by inhibiting FAAH/MAGL, modulating the ECS and neuroinflammatory factors, and suppressing glial cell activity. Additionally, cannabinoid receptors and TLR4 colocalized with astrocytes and microglia in the spinal cord. This study highlights the therapeutic potential of JZL195 in modulating the ECS and suggests a correlation between cannabinoid receptors and TLR4 in spinal glial cells, providing insight into alleviating pain and neuroinflammation in CIPN.
Collapse
Affiliation(s)
- Leejeong Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Department of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Guanghai Nan
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Department of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hee Young Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Department of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Myeounghoon Cha
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Bae Hwan Lee
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Department of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| |
Collapse
|
10
|
Babu N, Gadepalli A, Akhilesh, Sharma D, Singh AK, Chouhan D, Agrawal S, Tiwari V. TLR-4: a promising target for chemotherapy-induced peripheral neuropathy. Mol Biol Rep 2024; 51:1099. [PMID: 39466456 DOI: 10.1007/s11033-024-10038-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) affects a significant majority of cancer patients, with up to 80% experiencing this severe and dose-limiting side effect while undergoing anti-cancer treatment. CIPN can be induced by a variety of drugs commonly employed in the management of both solid tumors and hematologic cancers. The inadequacies in comprehending the pharmacological interventions associated with CIPN and the subsequent signaling pathways have significantly contributed to the disappointing outcomes of several drugs in clinical trials. Recent investigations in pain research have demonstrated a growing inclination toward addressing neuro-inflammation as a strategy for managing chronic pain conditions. Notably, toll-like receptor-4 (TLR-4) has emerged as a key player in immune system activation and is undergoing extensive research. In this review, we emphasize the potential role of TLR-4 in neuropathic pain, highlighting its promise as a target for CIPN treatment. Furthermore, we explore and analyse the intricate interplay between TLR-4, diverse immune cells, downstream pathways, and receptors within the context of CIPN. A comprehensive exploration of these interactions provides valuable insights into the central role of TLR-4 in CIPN development, paving the way for potential ground-breaking therapeutic approaches to alleviate this debilitating condition.
Collapse
Affiliation(s)
- Nagendra Babu
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Anagha Gadepalli
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Akhilesh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Dilip Sharma
- Amity Institute of Pharmacy, Amity University of Haryana, Gurgaon, India
| | - Anurag Kumar Singh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Deepak Chouhan
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Somesh Agrawal
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India.
| |
Collapse
|
11
|
Kim YS, Choi SH, Kim KY, Navia-Pelaez JM, Perkins GA, Choi S, Kim J, Nazarenkov N, Rissman RA, Ju WK, Ellisman MH, Miller YI. AIBP controls TLR4 inflammarafts and mitochondrial dysfunction in a mouse model of Alzheimer's disease. J Neuroinflammation 2024; 21:245. [PMID: 39342323 PMCID: PMC11439205 DOI: 10.1186/s12974-024-03214-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 08/29/2024] [Indexed: 10/01/2024] Open
Abstract
Microglia-driven neuroinflammation plays an important role in the development of Alzheimer's disease. Microglia activation is accompanied by the formation and chronic expression of TLR4 inflammarafts, defined as enlarged and cholesterol-rich lipid rafts serving as an assembly platform for TLR4 dimers and complexes of other inflammatory receptors. The secreted apoA-I binding protein (APOA1BP or AIBP) binds TLR4 and selectively targets cholesterol depletion machinery to TLR4 inflammaraft-expressing inflammatory, but not homeostatic microglia. Here we demonstrated that amyloid-beta (Aβ) induced formation of TLR4 inflammarafts in microglia in vitro and in the brain of APP/PS1 mice. Mitochondria in Apoa1bp-/- APP/PS1 microglia were hyperbranched and cupped, which was accompanied by increased reactive oxygen species and the dilated endoplasmic reticulum. The size and number of Aβ plaques and neuronal cell death were significantly increased, and the animal survival was decreased in Apoa1bp-/-APP/PS1 compared to APP/PS1 female mice. These results suggest that AIBP exerts control of TLR4 inflammarafts and mitochondrial dynamics in microglia and plays a protective role in Alzheimer's disease associated oxidative stress and neurodegeneration.
Collapse
Affiliation(s)
- Yi Sak Kim
- Department of Medicine, University of California, San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Soo-Ho Choi
- Department of Medicine, University of California, San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Keun-Young Kim
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Juliana M Navia-Pelaez
- Department of Medicine, University of California, San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Guy A Perkins
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Seunghwan Choi
- Viterbi Family Department of Ophthalmology, Hamilton Glaucoma Center and Shiley Eye Institute, University of California San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Jungsu Kim
- Department of Medicine, University of California, San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Nicolaus Nazarenkov
- Department of Medicine, University of California, San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Won-Kyu Ju
- Viterbi Family Department of Ophthalmology, Hamilton Glaucoma Center and Shiley Eye Institute, University of California San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Yury I Miller
- Department of Medicine, University of California, San Diego, La Jolla, San Diego, CA, 92093, USA.
| |
Collapse
|
12
|
Handlin LJ, Macchi NL, Dumaire NLA, Salih L, Lessie EN, McCommis KS, Moutal A, Dai G. Membrane Lipid Nanodomains Modulate HCN Pacemaker Channels in Nociceptor DRG Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.02.556056. [PMID: 37732182 PMCID: PMC10508734 DOI: 10.1101/2023.09.02.556056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Cell membranes consist of heterogeneous lipid nanodomains that influence key cellular processes. Using FRET-based fluorescent assays and fluorescence lifetime imaging microscopy (FLIM), we found that the dimension of cholesterol-enriched ordered membrane domains (OMD) varies considerably, depending on specific cell types. Particularly, nociceptor dorsal root ganglion (DRG) neurons exhibit large OMDs. Disruption of OMDs potentiated action potential firing in nociceptor DRG neurons and facilitated the opening of native hyperpolarization-activated cyclic nucleotide-gated (HCN) pacemaker channels. This increased neuronal firing is partially due to an increased open probability and altered gating kinetics of HCN channels. The gating effect on HCN channels was likely due to a direct modulation of their voltage sensors by OMDs. In animal models of neuropathic pain, we observed reduced OMD size and a loss of HCN channel localization within OMDs. Additionally, cholesterol supplementation inhibited HCN channels and reduced neuronal hyperexcitability in pain models. These findings suggest that disturbances in lipid nanodomains play a critical role in regulating HCN channels within nociceptor DRG neurons, influencing pain modulation.
Collapse
|
13
|
Mattar M, Umutoni F, Hassan MA, Wamburu MW, Turner R, Patton JS, Chen X, Lei W. Chemotherapy-Induced Peripheral Neuropathy: A Recent Update on Pathophysiology and Treatment. Life (Basel) 2024; 14:991. [PMID: 39202733 PMCID: PMC11355765 DOI: 10.3390/life14080991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/29/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a major long-lasting side effect of some chemotherapy drugs, which threatens cancer survival rate. CIPN mostly affects sensory neurons and occasionally motor neurons, causing numbness, tingling, discomfort, and burning pain in the upper and lower extremities. The pathophysiology of CIPN is not completely understood; however, it is believed that chemotherapies induce peripheral neuropathy via directly damaging mitochondria, impairing the function of ion channels, triggering immunological mechanisms, and disrupting microtubules. The treatment of CIPN is a medical challenge, and there are no approved pharmacological options. Currently, duloxetine and other antidepressants, antioxidant, anti-inflammatory, and ion-channel targeted therapies are commonly used in clinics to relieve the symptoms of CIPN. Several other types of drugs, such as cannabinoids, sigma-1 receptor antagonists, and nicotinamides ribose, are being evaluated in preclinical and clinical studies. This paper summarizes the information related to the physiology of CIPN and medicines that could be used for treating this condition.
Collapse
Affiliation(s)
- Marina Mattar
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, Clinton, SC 29325, USA; (M.M.); (M.A.H.)
| | - Florence Umutoni
- Department of Pharmaceutical and Graduate Life Sciences, College of Health Sciences, Nursing, and Pharmacy, Manchester University, Fort Wayne, IN 46845, USA; (F.U.); (J.S.P.)
| | - Marwa A. Hassan
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, Clinton, SC 29325, USA; (M.M.); (M.A.H.)
| | - M. Wambui Wamburu
- Department of Pharmacy Practice, College of Health Sciences, Nursing, and Pharmacy, Manchester University, Fort Wayne, IN 46845, USA;
| | - Reagan Turner
- Department of Biology, Presbyterian College, Clinton, SC 29325, USA;
| | - James S. Patton
- Department of Pharmaceutical and Graduate Life Sciences, College of Health Sciences, Nursing, and Pharmacy, Manchester University, Fort Wayne, IN 46845, USA; (F.U.); (J.S.P.)
| | - Xin Chen
- Department of Pharmaceutical and Clinical Sciences, College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC 27506, USA;
| | - Wei Lei
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, Clinton, SC 29325, USA; (M.M.); (M.A.H.)
- Department of Pharmaceutical and Graduate Life Sciences, College of Health Sciences, Nursing, and Pharmacy, Manchester University, Fort Wayne, IN 46845, USA; (F.U.); (J.S.P.)
| |
Collapse
|
14
|
Cong J, Lv H, Xu Y. The role of nociceptive neurons in allergic rhinitis. Front Immunol 2024; 15:1430760. [PMID: 39185421 PMCID: PMC11341422 DOI: 10.3389/fimmu.2024.1430760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/25/2024] [Indexed: 08/27/2024] Open
Abstract
Allergic rhinitis (AR) is a chronic, non-infectious condition affecting the nasal mucosa, primarily mediated mainly by IgE. Recent studies reveal that AR is intricately associated not only with type 2 immunity but also with neuroimmunity. Nociceptive neurons, a subset of primary sensory neurons, are pivotal in detecting external nociceptive stimuli and modulating immune responses. This review examines nociceptive neuron receptors and elucidates how neuropeptides released by these neurons impact the immune system. Additionally, we summarize the role of immune cells and inflammatory mediators on nociceptive neurons. A comprehensive understanding of the dynamic interplay between nociceptive neurons and the immune system augments our understanding of the neuroimmune mechanisms underlying AR, thereby opening novel avenues for AR treatment modalities.
Collapse
Affiliation(s)
- Jianchao Cong
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hao Lv
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yu Xu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, Wuhan, China
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan, China
| |
Collapse
|
15
|
Xu YS, Xiang J, Lin SJ. Functional role of P2X7 purinergic receptor in cancer and cancer-related pain. Purinergic Signal 2024:10.1007/s11302-024-10019-w. [PMID: 38771429 DOI: 10.1007/s11302-024-10019-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/08/2024] [Indexed: 05/22/2024] Open
Abstract
Numerous studies have revealed that the ATP-gated ion channel purinergic 2X7 receptor (P2X7R) plays an important role in tumor progression and the pathogenesis of cancer pain. P2X7R requires activation by extracellular ATP to perform its regulatory role functions. During tumor development or cancer-induced pain, ATP is released from tumor cells or other cells in the tumor microenvironment (such as tumor-associated immune cells), which activates P2X7R, opens ion channels on the cell membrane, affects intracellular molecular metabolism, and regulates the activity of tumor cells. Furthermore, peripheral organs and receptors can be damaged during tumor progression, and P2X7R expression in nerve cells (such as microglia) is significantly upregulated, enhancing sensory afferent information, sensitizing the central nervous system, and inducing or exacerbating pain. These findings reveal that the ATP-P2X7R signaling axis plays a key regulatory role in the pathogenesis of tumors and cancer pain and also has a therapeutic role. Accordingly, in this study, we explored the role of P2X7R in tumors and cancer pain, discussed the pharmacological properties of inhibiting P2X7R activity (such as the use of antagonists) or blocking its expression in the treatment of tumor and cancer pain, and provided an important evidence for the treatment of both in the future.
Collapse
Affiliation(s)
- Yong-Sheng Xu
- The Second Affiliated Hospital, Nanchang University, Nanchang City, 343000, Jiangxi Province, China
| | - Jun Xiang
- The Second Affiliated Hospital, Nanchang University, Nanchang City, 343000, Jiangxi Province, China
| | - Si-Jian Lin
- Department of Rehabilitation Medicine, the Second Affiliated Hospital, Nanchang University, Nanchang City, 343000, Jiangxi Province, China.
| |
Collapse
|
16
|
Kacem H, Cimini A, d’Angelo M, Castelli V. Molecular and Cellular Involvement in CIPN. Biomedicines 2024; 12:751. [PMID: 38672107 PMCID: PMC11048589 DOI: 10.3390/biomedicines12040751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Many anti-cancer drugs, such as taxanes, platinum compounds, vinca alkaloids, and proteasome inhibitors, can cause chemotherapy-induced peripheral neuropathy (CIPN). CIPN is a frequent and harmful side effect that affects the sensory, motor, and autonomic nerves, leading to pain, numbness, tingling, weakness, and reduced quality of life. The causes of CIPN are not fully known, but they involve direct nerve damage, oxidative stress, inflammation, DNA damage, microtubule dysfunction, and altered ion channel activity. CIPN is also affected by genetic, epigenetic, and environmental factors that modulate the risk and intensity of nerve damage. Currently, there are no effective treatments or prevention methods for CIPN, and symptom management is mostly symptomatic and palliative. Therefore, there is a high demand for better understanding of the cellular and molecular mechanisms involved in CIPN, as well as the development of new biomarkers and therapeutic targets. This review gives an overview of the current knowledge and challenges in the field of CIPN, focusing on the biological and molecular mechanisms underlying this disorder.
Collapse
Affiliation(s)
| | | | - Michele d’Angelo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (H.K.); (A.C.); (V.C.)
| | | |
Collapse
|
17
|
Kim YS, Choi SH, Kim KY, Navia-Pelaez JM, Perkins GA, Choi S, Kim J, Nazarenkov N, Rissman RA, Ju WK, Ellisman MH, Miller YI. AIBP controls TLR4 inflammarafts and mitochondrial dysfunction in a mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.16.580751. [PMID: 38586011 PMCID: PMC10996524 DOI: 10.1101/2024.02.16.580751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Microglia-driven neuroinflammation plays an important role in the development of Alzheimer's disease (AD). Microglia activation is accompanied by the formation and chronic maintenance of TLR4 inflammarafts, defined as enlarged and cholesterol-rich lipid rafts serving as an assembly platform for TLR4 dimers and complexes of other inflammatory receptors. The secreted apoA-I binding protein (APOA1BP or AIBP) binds TLR4 and selectively targets cholesterol depletion machinery to TLR4 inflammaraft expressing inflammatory, but not homeostatic microglia. Here we demonstrated that amyloid-beta (Aβ) induced formation of TLR4 inflammarafts in microglia in vitro and in the brain of APP/PS1 mice. Mitochondria in Apoa1bp-/- APP/PS1 microglia were hyperbranched and cupped, which was accompanied by increased ROS and the dilated ER. The size and number of Aβ plaques and neuronal cell death were significantly increased, and the animal survival was decreased in Apoa1bp-/- APP/PS1 compared to APP/PS1 female mice. These results suggest that AIBP exerts control of TLR4 inflammarafts and mitochondrial dynamics in microglia and plays a protective role in AD associated oxidative stress and neurodegeneration.
Collapse
Affiliation(s)
- Yi Sak Kim
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Soo-Ho Choi
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Keun-Young Kim
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
| | | | - Guy A. Perkins
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Seunghwan Choi
- Hamilton Glaucoma Center and Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jungsu Kim
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Nicolaus Nazarenkov
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Robert A. Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Won-Kyu Ju
- Hamilton Glaucoma Center and Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mark H. Ellisman
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yury I. Miller
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
18
|
Choi S, Choi SH, Bastola T, Park Y, Oh J, Kim KY, Hwang S, Miller YI, Ju WK. AIBP: A New Safeguard against Glaucomatous Neuroinflammation. Cells 2024; 13:198. [PMID: 38275823 PMCID: PMC10814024 DOI: 10.3390/cells13020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/18/2024] [Accepted: 01/20/2024] [Indexed: 01/27/2024] Open
Abstract
Glaucoma is a group of ocular diseases that cause irreversible blindness. It is characterized by multifactorial degeneration of the optic nerve axons and retinal ganglion cells (RGCs), resulting in the loss of vision. Major components of glaucoma pathogenesis include glia-driven neuroinflammation and impairment of mitochondrial dynamics and bioenergetics, leading to retinal neurodegeneration. In this review article, we summarize current evidence for the emerging role of apolipoprotein A-I binding protein (AIBP) as an important anti-inflammatory and neuroprotective factor in the retina. Due to its association with toll-like receptor 4 (TLR4), extracellular AIBP selectively removes excess cholesterol from the plasma membrane of inflammatory and activated cells. This results in the reduced expression of TLR4-associated, cholesterol-rich lipid rafts and the inhibition of downstream inflammatory signaling. Intracellular AIBP is localized to mitochondria and modulates mitophagy through the ubiquitination of mitofusins 1 and 2. Importantly, elevated intraocular pressure induces AIBP deficiency in mouse models and in human glaucomatous retina. AIBP deficiency leads to the activation of TLR4 in Müller glia, triggering mitochondrial dysfunction in both RGCs and Müller glia, and compromising visual function in a mouse model. Conversely, restoring AIBP expression in the retina reduces neuroinflammation, prevents RGCs death, and protects visual function. These results provide new insight into the mechanism of AIBP function in the retina and suggest a therapeutic potential for restoring retinal AIBP expression in the treatment of glaucoma.
Collapse
Affiliation(s)
- Seunghwan Choi
- Hamilton Glaucoma Center and Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA 92093, USA; (S.C.); (T.B.); (Y.P.)
| | - Soo-Ho Choi
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Tonking Bastola
- Hamilton Glaucoma Center and Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA 92093, USA; (S.C.); (T.B.); (Y.P.)
| | - Younggun Park
- Hamilton Glaucoma Center and Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA 92093, USA; (S.C.); (T.B.); (Y.P.)
- Department of Ophthalmology and Visual Science, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jonghyun Oh
- Hamilton Glaucoma Center and Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA 92093, USA; (S.C.); (T.B.); (Y.P.)
- Department of Ophthalmology, Dongguk University Ilsan Hospital, Goyang 10326, Republic of Korea
| | - Keun-Young Kim
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Sinwoo Hwang
- Hamilton Glaucoma Center and Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA 92093, USA; (S.C.); (T.B.); (Y.P.)
| | - Yury I. Miller
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Won-Kyu Ju
- Hamilton Glaucoma Center and Shiley Eye Institute, Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, CA 92093, USA; (S.C.); (T.B.); (Y.P.)
| |
Collapse
|
19
|
Akhilesh, Chouhan D, Ummadisetty O, Verma N, Tiwari V. Bergenin ameliorates chemotherapy-induced neuropathic pain in rats by modulating TRPA1/TRPV1/NR2B signalling. Int Immunopharmacol 2023; 125:111100. [PMID: 38149571 DOI: 10.1016/j.intimp.2023.111100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/26/2023] [Accepted: 10/18/2023] [Indexed: 12/28/2023]
Abstract
Chemotherapy-induced neuropathic pain (CINP) is one of the most prominent and incapacitating complication associated with chemotherapeutic regimens. The exact mechanisms underlying CINP are not fully understood yet, which hampers the development of effective therapeutics. The current study has been designed to investigate the effect of bergenin on CINP and dissect the underlying cellular and molecular mechanisms. Behavioural responsiveness assays were conducted in rats before and after CINP induction and at different time points post-bergenin treatment. We also measured alterations in tight junction proteins, pro-inflammatory cytokines, microglia activity, transient receptor potential (TRP) channels (TRPV1, TRPA1 and TRPM8) and N-methyl-D-aspartate receptor subtype 2 (NR2B) in dorsal root ganglion (DRG) and spinal tissues of neuropathic rats. Bergenin treatment leads to a significant and dose-dependent reduction in evoked and spontaneous ongoing pain without causing central side effects in neuropathic rats. Furthermore, treatment with bergenin and gabapentin did not affect the baseline pain threshold in healthy, non-chemotherapy-treated rats, as evaluated through tail-flick and tail-clip assays. Chemotherapy administration leads to a significant activation of TRP channels, concurrent with microglial activation, disruption of spinal cord tight junction proteins, and subsequent infiltration of pro-inflammatory cytokines, as well as NR2B activation. Notably, bergenin treatment effectively reversed all of these alterations, with the exception of TRPM8, in both the DRG and spinal cord of neuropathic rats. Findings from the present study suggests that bergenin mitigates neuropathic pain by modulating the TRPA1/TRPV1/NR2B signalling and presents a promising therapeutic avenue for the treatment of chemotherapy-induced neuropathic pain.
Collapse
Affiliation(s)
- Akhilesh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Deepak Chouhan
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Obulapathi Ummadisetty
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Nivedita Verma
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India.
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Advances in single cell techniques revealed a remarkable diversity in macrophage gene expression profiles in atherosclerosis. However, the diversity of functional processes at the macrophage plasma membrane remains less studied. This review summarizes recent advances in characterization of lipid rafts, where inflammatory receptors assemble, in macrophages that undergo reprogramming in atherosclerotic lesions and in vitro under conditions relevant to the development of atherosclerosis. RECENT FINDINGS The term inflammarafts refers to enlarged lipid rafts with increased cholesterol content, hosting components of inflammatory receptor complexes assembled in close proximity, including TLR4-TLR4, TLR2-TLR1 and TLR2-CD36 dimers. Macrophages decorated with inflammarafts maintain chronic inflammatory gene expression and are primed to an augmented response to additional inflammatory stimuli. In mouse atherosclerotic lesions, inflammarafts are expressed primarily in nonfoamy macrophages and less in lipid-laden foam cells. This agrees with the reported suppression of inflammatory programs in foam cells. In contrast, nonfoamy macrophages expressing inflammarafts are the major inflammatory population in atherosclerotic lesions. Discussed are emerging reports that help understand formation and persistence of inflammarafts and the potential of inflammarafts as a novel therapeutic target. SUMMARY Chronic maintenance of inflammarafts in nonfoamy macrophages serves as an effector mechanism of inflammatory macrophage reprogramming in atherosclerosis.
Collapse
Affiliation(s)
- Shenglin Li
- Department of Medicine, University of California, San Diego, California, USA
| | | | | | | |
Collapse
|