1
|
Huang Z, Tan H, Fu Y, Xie H, Tan H, Gao K, Lou H. Neurotransmitter imbalance and amygdala synaptic plasticity in lumbar disc herniation-induced chronic pain and related emotional Disturbances:A multi-omics analysis. Neuropharmacology 2025; 271:110405. [PMID: 40057176 DOI: 10.1016/j.neuropharm.2025.110405] [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: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Chronic pain due to lumbar disc herniation (LDH) significantly impairs quality of life and is often accompanied by emotional disturbances, such as anxiety and depression. Despite the recognition of these comorbidities, the underlying neural mechanisms remain unclear. This study investigates the role of neurotransmitter imbalances and key regulatory molecules in LDH-induced chronic pain and related emotional disturbances, with a focus on synaptic plasticity in the amygdala. A rat model of LDH was developed using male Sprague-Dawley rats. Behavioral assessments were conducted to evaluate pain hypersensitivity, anxiety, and depression-like behaviors. Cerebrospinal fluid (CSF) metabolomics and amygdala transcriptomics were employed to analyze neurotransmitter profiles and gene expression. In vitro experiments were conducted to explore the role of PRKCG in synaptic plasticity. Behavioral tests showed significant pain hypersensitivity and anxiety- and depression-like behavior in LDH rats. Metabolomic analysis revealed altered levels of glutamate and γ-aminobutyric acid (GABA) in the CSF, indicating neurotransmitter imbalances. Transcriptomic profiling identified changes in genes related to synaptic plasticity, including PRKCG. PRKCG knockdown led to reduced CAMKII phosphorylation and GRIA1 expression, supporting its role in modulating synaptic plasticity. This study provides evidence that neurotransmitter imbalances and alterations in synaptic plasticity within the amygdala may contribute to the persistence of chronic pain and associated emotional disturbances in LDH. PRKCG may represent a novel therapeutic target for treating both chronic pain and related emotional disturbances.
Collapse
Affiliation(s)
- Zhenyu Huang
- Ningbo Municipal Hospital of Traditional Chinese Medicine, Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo, 315010, China.
| | - Haibo Tan
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China; The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China.
| | - Yuanfei Fu
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China.
| | - Huanxin Xie
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China.
| | - Huangsheng Tan
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China.
| | - Kun Gao
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China.
| | - Hongkan Lou
- Ningbo Municipal Hospital of Traditional Chinese Medicine, Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo, 315010, China.
| |
Collapse
|
2
|
Nasir A, Afridi M, Afridi OK, Khan MA, Khan A, Zhang J, Qian B. The persistent pain enigma: Molecular drivers behind acute-to-chronic transition. Neurosci Biobehav Rev 2025; 173:106162. [PMID: 40239909 DOI: 10.1016/j.neubiorev.2025.106162] [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: 02/26/2025] [Revised: 03/20/2025] [Accepted: 04/14/2025] [Indexed: 04/18/2025]
Abstract
The transition from acute to chronic pain is a complex and multifactorial process that presents significant challenges in both diagnosis and treatment. Key mechanisms of peripheral and central sensitization, neuroinflammation, and altered synaptic plasticity contribute to the amplification of pain signals and the persistence of pain. Glial cell activation, particularly microglia and astrocytes, is pivotal in developing chronic pain by releasing pro-inflammatory cytokines that enhance pain sensitivity. This review explores the molecular, cellular, and systemic mechanisms underlying the transition from acute to chronic pain, offering new insights into the molecular and neurobiological mechanisms involved, which are often underexplored in existing literature. It also addresses emerging therapeutic strategies beyond traditional pain management, offering valuable perspectives for future research and clinical applications.
Collapse
Affiliation(s)
- Abdul Nasir
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China.
| | - Maryam Afridi
- Department of Pharmacy, Qurtuba University, Peshawar, KP, Pakistan
| | | | | | - Amir Khan
- Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jun Zhang
- Department of Pain, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Bai Qian
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China.
| |
Collapse
|
3
|
Boccella S, Fusco A, Ricciardi F, Morace AM, Bonsale R, Perrone M, Marabese I, De Gregorio D, Belardo C, Posa L, Rullo L, Piscitelli F, di Marzo V, Nicois A, Marfella B, Cristino L, Luongo L, Guida F, Candeletti S, Gobbi G, Romualdi P, Maione S. Acute kappa opioid receptor blocking disrupts the pro-cognitive effect of cannabidiol in neuropathic rats. Neuropharmacology 2025; 266:110265. [PMID: 39674399 DOI: 10.1016/j.neuropharm.2024.110265] [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: 10/08/2024] [Revised: 11/20/2024] [Accepted: 12/10/2024] [Indexed: 12/16/2024]
Abstract
Cannabidiol has been shown to ameliorate neuropathic pain and its affective components. Previous studies highlighted the pharmacological interaction between the CBD and opioid system, particularly the MOR, but the understanding of the interaction between CBD and kappa opioid receptor (KOR), physiologically stimulated by the endogenous opioid dynorphin, remains elusive. We assessed the pharmacological interactions between CBD and nor-BNI, a selective KOR antagonist in a rat neuropathic pain model. We show an increase in dynorphin peptide and its KOR receptors in the hippocampus' dentate gyrus (DG) of neuropathic rats showing allodynia, and memory deficits. Consistent with these findings, neuropathic pain was associated with long-term potentiation (LTP) impairment in the entorhinal cortex-DG, also referred to as the lateral perforant pathway (LPP). Moreover, a downregulation of the endocannabinoid 2-AG and an upregulation of the cannabinoid CB1 receptors in the DG were detected in neuropathic pain animals. Either an acute KOR antagonist administration or one-week CBD treatment normalized dynorphin levels and improved affective symptoms, LTP and receptor expression, whereas only CBD showed an anti-allodynic effect. In addition, CBD normalized the SNI-induced changes in neuroplasticity as well as endocannabinoid and GABA levels in the DG. Noteworthy, the acute blockade of the KOR carried out after CBD repeated administration causes the re-installment of some neuropathic condition symptoms. As a whole, these original results indicate a critical relationship between the adaptive changes in the hippocampus produced by CBD and the need to maintain the recovered physiological dynorphin tone to preserve the therapeutic effect of CBD in neuropathic rats.
Collapse
Affiliation(s)
- Serena Boccella
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Naples, Italy.
| | - Antimo Fusco
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Naples, Italy
| | - Federica Ricciardi
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Naples, Italy
| | - Andrea Maria Morace
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Naples, Italy
| | - Roozbe Bonsale
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Naples, Italy
| | - Michela Perrone
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Naples, Italy
| | - Ida Marabese
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Naples, Italy
| | - Danilo De Gregorio
- IRCCS San Raffaele Scientific Institute, Italy; Vita Salute San Raffaele University, Milan, Italy
| | - Carmela Belardo
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Naples, Italy
| | - Luca Posa
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Laura Rullo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, 40126, Bologna, Italy
| | - Fabiana Piscitelli
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, NA, Italy
| | - Vincenzo di Marzo
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, NA, Italy; Faculty of Medicine and Faculty of Agricultural and Food Sciences, Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Québec City, QC, Canada; Heart and Lung Research Institute of Université Laval, Québec City, QC, Canada; Institute for Nutrition and Functional Foods, Centre NUTRISS, Université Laval, Québec City, QC, Canada
| | - Alessandro Nicois
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, NA, Italy; Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Brenda Marfella
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, NA, Italy; Department of Biology, University of Naples Federico II, 80126, Naples, Italy
| | - Luigia Cristino
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, NA, Italy
| | - Livio Luongo
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Naples, Italy
| | - Sanzio Candeletti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, 40126, Bologna, Italy
| | - Gabriella Gobbi
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, QC, Canada; Research Institute, McGill University Health Center, McGill University, Montreal, QC, Canada
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, 40126, Bologna, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Naples, Italy
| |
Collapse
|
4
|
Tagne AM, Fotio Y, Lee HL, Jung KM, Katz J, Ahmed F, Le J, Bazinet R, Jang C, Piomelli D. Metabolic reprogramming in the spinal cord drives the transition to pain chronicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635746. [PMID: 39975205 PMCID: PMC11838349 DOI: 10.1101/2025.01.30.635746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Acute injuries can progress into painful states that endure long after healing. The mechanism underlying this transition remains unclear, but metabolic adaptations to the bioenergy demands imposed by injury are plausible contributors. Here we show that peripheral injury activates AKT/mTORC1 in afferent segments of the mouse spinal cord, redirecting local core metabolism toward biomass production while simultaneously suppressing autophagy-mediated biomass reclamation. This metabolic shift supports neuroplasticity, but creates a resource bottleneck that depletes critical spinal cord nutrients. Preventing this depletion with a modified diet normalizes biomass generation and autophagy and halts the transition to chronic pain. This effect, observed across multiple pain models, requires activation of the nutrient sensors, sirtuin-1 and AMPK, as well as restoration of autophagy. The findings identify metabolic reprogramming as a key driver of the progression to pain chronicity and point to nutritional and pharmacological interventions that could prevent this progression after surgery or other physical traumas.
Collapse
Affiliation(s)
- Alex Mabou Tagne
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Yannick Fotio
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Hye-Lim Lee
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Kwang-Mook Jung
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Jean Katz
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Faizy Ahmed
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Johnny Le
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Richard Bazinet
- Department of Nutritional Sciences, University of Toronto, ON, Canada
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
5
|
Kim K, Nan G, Kim HY, Cha M, Lee BH. Targeting the insular cortex for neuropathic pain modulation: Insights into synaptic and neuronal mechanisms. FASEB J 2025; 39:e70285. [PMID: 39831885 PMCID: PMC11745213 DOI: 10.1096/fj.202402381r] [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: 10/07/2024] [Revised: 11/26/2024] [Accepted: 12/20/2024] [Indexed: 01/22/2025]
Abstract
Neuropathic pain, caused by nerve damage, greatly affects quality of life. Recent research proposes modulating brain activity, particularly through electrical stimulation of the insular cortex (IC), as a treatment option. This study aimed to understand how IC stimulation (ICS) affects pain modulation. In a rat neuropathy model, researchers used optogenetic and ICS techniques to evaluate changes in mechanical allodynia and synaptic changes, focusing on glutamate receptors (AMPAR, NR2A, NR2B). Optogenetic inhibition of IC neurons relieved pain without altering synaptic plasticity. However, repetitive ICS combined with optogenetic activation diminished the pain-relieving effects of ICS and increased AMPAR and NR2B receptor levels. Additionally, activating inhibitory neurons also reduced pain, while repetitive activation of excitatory neurons lessened the effectiveness of ICS and was associated with heightened receptor expression. These findings suggest that inhibiting excitatory neurons or activating inhibitory neurons in the IC could help modulate pain in neuropathic conditions, shedding light on how ICS can influence pain management through changes in synaptic plasticity.
Collapse
Affiliation(s)
- Kyeongmin Kim
- Department of PhysiologyYonsei University College of MedicineSeoulRepublic of Korea
| | - Guanghai Nan
- Department of PhysiologyYonsei University College of MedicineSeoulRepublic of Korea
- Department of Medical ScienceBrain Korea 21 Project, Yonsei University College of MedicineSeoulRepublic of Korea
| | - Hee Young Kim
- Department of PhysiologyYonsei University College of MedicineSeoulRepublic of Korea
| | - Myeounghoon Cha
- Department of PhysiologyYonsei University College of MedicineSeoulRepublic of Korea
- Department of Physiology, College of MedicineSoonchunhyang UniversityCheonanRepublic of Korea
| | - Bae Hwan Lee
- Department of PhysiologyYonsei University College of MedicineSeoulRepublic of Korea
- Department of Medical ScienceBrain Korea 21 Project, Yonsei University College of MedicineSeoulRepublic of Korea
- Brain Research InstituteYonsei University College of MedicineSeoulRepublic of Korea
| |
Collapse
|
6
|
Bigliassi M, Cabral DF, Evans AC. Improving brain health via the central executive network. J Physiol 2025. [PMID: 39856810 DOI: 10.1113/jp287099] [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: 06/17/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Cognitive and physical stress have significant effects on brain health, particularly through their influence on the central executive network (CEN). The CEN, which includes regions such as the dorsolateral prefrontal cortex, anterior cingulate cortex and inferior parietal lobe, is central to managing the demands of cognitively challenging motor tasks. Acute stress can temporarily reduce connectivity within the CEN, leading to impaired cognitive function and emotional states. However a rebound in these states often follows, driven by motivational signals through the mesocortical and mesolimbic pathways, which help sustain inhibitory control and task execution. Chronic exposure to physical and cognitive challenges leads to long-term improvements in CEN functionality. These changes are supported by neurochemical, structural and systemic adaptations, including mechanisms of tissue crosstalk. Myokines, adipokines, anti-inflammatory cytokines and gut-derived metabolites contribute to a biochemical environment that enhances neuroplasticity, reduces neuroinflammation and supports neurotransmitters such as serotonin and dopamine. These processes strengthen CEN connectivity, improve self-regulation and enable individuals to adopt and sustain health-optimizing behaviours. Long-term physical activity not only enhances inhibitory control but also reduces the risk of age-related cognitive decline and neurodegenerative diseases. This review highlights the role of progressive physical stress through exercise as a practical approach to strengthening the CEN and promoting brain health, offering a strategy to improve cognitive resilience and emotional well-being across the lifespan.
Collapse
Affiliation(s)
- Marcelo Bigliassi
- Department of Teaching and Learning, Florida International University, Miami, Florida, USA
| | - Danylo F Cabral
- Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA
| | - Amanda C Evans
- Functional Flow Solutions LLC, Albuquerque, New Mexico, USA
| |
Collapse
|
7
|
Vitale RM, Morace AM, D'Errico A, Ricciardi F, Fusco A, Boccella S, Guida F, Nasso R, Rading S, Karsak M, Caprioglio D, Iannotti FA, Arcone R, Luongo L, Masullo M, Maione S, Amodeo P. Identification of Cannabidiolic and Cannabigerolic Acids as MTDL AChE, BuChE, and BACE-1 Inhibitors Against Alzheimer's Disease by In Silico, In Vitro, and In Vivo Studies. Phytother Res 2025; 39:233-245. [PMID: 39510547 PMCID: PMC11745148 DOI: 10.1002/ptr.8369] [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: 07/08/2024] [Revised: 09/25/2024] [Accepted: 10/16/2024] [Indexed: 11/15/2024]
Abstract
Cannabidiolic (CBDA) and cannabigerolic (CBGA) acids are naturally occurring compounds from Cannabis sativa plant, previously identified by us as dual PPARα/γ agonists. Since the development of multitarget-directed ligands (MTDL) represents a valuable strategy to alleviate and slow down the progression of multifactorial diseases, we evaluated the potential ability of CBDA and CBGA to also inhibit enzymes involved in the modulation of the cholinergic tone and/or β-amyloid production. A multidisciplinary approach based on computational and biochemical studies was pursued on selected enzymes, followed by behavioral and electrophysiological experiments in an AD mouse model. The β-arrestin assay on GPR109A and qPCR on TRPM7 were also carried out. CBDA and CBGA are effective on both acetyl- and butyryl-cholinesterases (AChE/BuChE), as well as on β-secretase-1 (BACE-1) enzymes in a low micromolar range, and they also prevent aggregation of β-amyloid fibrils. Computational studies provided a rationale for the competitive (AChE) vs. noncompetitive (BuChE) inhibitory profile of the two ligands. The repeated treatment with CBDA and CBGA (10 mg/kg, i.p.) improved the cognitive deficit induced by the β-amyloid peptide. A recovery of the long-term potentiation in the hippocampus was observed, where the treatment with CBGA and CBDA also restored the physiological expression level of TRPM7, a receptor channel involved in neurodegenerative diseases. We also showed that these compounds do not stimulate GPR109A in β-arrestin assay. Collectively, these data broaden the pharmacological profile of CBDA and CBGA and suggest their potential use as novel anti-AD MTDLs.
Collapse
Affiliation(s)
- Rosa Maria Vitale
- Institute of Biomolecular Chemistry (ICB), National Research Council (CNR)Pozzuoli (NA)Italy
| | - Andrea Maria Morace
- Department of Experimental Medicine, Division of PharmacologyUniversity of Campania "Luigi Vanvitelli"NaplesItaly
| | - Antonio D'Errico
- Department of Medical, Human Movement and Well‐Being SciencesUniversity of Naples “Parthenope”NaplesItaly
| | - Federica Ricciardi
- Department of Experimental Medicine, Division of PharmacologyUniversity of Campania "Luigi Vanvitelli"NaplesItaly
| | - Antimo Fusco
- Department of Experimental Medicine, Division of PharmacologyUniversity of Campania "Luigi Vanvitelli"NaplesItaly
| | - Serena Boccella
- Department of Experimental Medicine, Division of PharmacologyUniversity of Campania "Luigi Vanvitelli"NaplesItaly
| | - Francesca Guida
- Department of Experimental Medicine, Division of PharmacologyUniversity of Campania "Luigi Vanvitelli"NaplesItaly
| | - Rosarita Nasso
- Department of Medical, Human Movement and Well‐Being SciencesUniversity of Naples “Parthenope”NaplesItaly
| | - Sebastian Rading
- Neuronal and Cellular Signal Transduction, Center for Molecular Neurobiology Hamburg (ZMNH)University Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
- Institute of Human Genetics, University Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Meliha Karsak
- Neuronal and Cellular Signal Transduction, Center for Molecular Neurobiology Hamburg (ZMNH)University Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
- Institute of Human Genetics, University Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Diego Caprioglio
- Department of Pharmaceutical and Pharmacological SciencesUniversity of Eastern Piedmont "A. Avogadro"NovaraItaly
| | - Fabio Arturo Iannotti
- Institute of Biomolecular Chemistry (ICB), National Research Council (CNR)Pozzuoli (NA)Italy
| | - Rosaria Arcone
- Department of Medical, Human Movement and Well‐Being SciencesUniversity of Naples “Parthenope”NaplesItaly
| | - Livio Luongo
- Department of Experimental Medicine, Division of PharmacologyUniversity of Campania "Luigi Vanvitelli"NaplesItaly
| | - Mariorosario Masullo
- Department of Medical, Human Movement and Well‐Being SciencesUniversity of Naples “Parthenope”NaplesItaly
| | - Sabatino Maione
- Department of Experimental Medicine, Division of PharmacologyUniversity of Campania "Luigi Vanvitelli"NaplesItaly
| | - Pietro Amodeo
- Institute of Biomolecular Chemistry (ICB), National Research Council (CNR)Pozzuoli (NA)Italy
| |
Collapse
|
8
|
Serafini RA, Farzinpour Z, Patel V, Kelley AM, Estill M, Pryce KD, Sakloth F, Teague CD, Torres-Berrio A, Nestler EJ, Shen L, Akbarian S, Karkhanis AN, Blitzer RD, Zachariou V. Nucleus accumbens myocyte enhancer factor 2C mediates the maintenance of peripheral nerve injury-induced physiological and behavioral maladaptations. Pain 2024; 165:2733-2748. [PMID: 38985454 DOI: 10.1097/j.pain.0000000000003316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 04/18/2024] [Indexed: 07/11/2024]
Abstract
ABSTRACT Preclinical and clinical work has demonstrated altered plasticity and activity in the nucleus accumbens (NAc) under chronic pain states, highlighting critical therapeutic avenues for the management of chronic pain conditions. In this study, we demonstrate that myocyte enhancer factor 2C (MEF2C), a master regulator of neuronal activity and plasticity, is repressed in NAc neurons after prolonged spared nerve injury (SNI). Viral-mediated overexpression of Mef2c in NAc neurons partially ameliorated sensory hypersensitivity and emotional behaviors in mice with SNI, while also altering transcriptional pathways associated with synaptic signaling. Mef2c overexpression also reversed SNI-induced potentiation of phasic dopamine release and neuronal hyperexcitability in the NAc. Transcriptional changes induced by Mef2c overexpression were different than those observed after desipramine treatment, suggesting a mechanism of action different from antidepressants. Overall, we show that interventions in MEF2C-regulated mechanisms in the NAc are sufficient to disrupt the maintenance of chronic pain states, providing potential new treatment avenues for neuropathic pain.
Collapse
Affiliation(s)
- Randal A Serafini
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pharmacology, Physiology, & Biophysics, Chobanian and Avedisian School of Medicine at Boston University, Boston, MA, United States
| | - Zahra Farzinpour
- Department of Pharmacology, Physiology, & Biophysics, Chobanian and Avedisian School of Medicine at Boston University, Boston, MA, United States
| | - Vishwendra Patel
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Abigail M Kelley
- Department of Psychology, Binghamton University-SUNY, Binghamton, NY, United States
| | - Molly Estill
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kerri D Pryce
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Farhana Sakloth
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Collin D Teague
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Angelica Torres-Berrio
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Li Shen
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Schahram Akbarian
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Anushree N Karkhanis
- Department of Psychology, Binghamton University-SUNY, Binghamton, NY, United States
| | - Robert D Blitzer
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Venetia Zachariou
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pharmacology, Physiology, & Biophysics, Chobanian and Avedisian School of Medicine at Boston University, Boston, MA, United States
| |
Collapse
|
9
|
Norman-Nott N, Cashin AG, Gustin SM. Psychological, physical and complementary therapies for the management of neuropathic pain. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 179:431-470. [PMID: 39580220 DOI: 10.1016/bs.irn.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
This chapter aims to explain and evaluate the evidence for psychological, physical and complementary therapies as part of a holistic plan for managing neuropathic pain. Psychological therapies refer to interventions targeting mental health, while physical therapies refer to interventions designed to target movement and functional ability, and complementary therapies are those that attempt to target key mechanisms of change to alter brain and body functioning, or thought processes related to the experience of pain. Each therapeutic modality is discussed to narratively report on the evidence and provide implications for clinicians. Where evidence was unavailable for neuropathic pain populations, evidence from chronic pain populations more broadly was considered. Although promising, there is a lack of high-quality evidence investigating the benefits and safety of psychological, physical and complementary therapies for the management of neuropathic pain. The low certainty evidence and lack of evidence across different neuropathic pain conditions impacts the ability to make recommendations for clinical practice. However, there are several potential areas for future research. Psychological therapies that focus on the underlying mechanisms related to emotion regulation may improve mood and pain, while cognitive and behavioural based approaches may improve psychological comorbidities such as anxiety and depression. Physical therapies involving physical activity and exercise, education, and graded motor imagery may improve functioning and reduce pain. Finally, complementary therapies including electroencephalography neurofeedback, acupuncture, virtual reality, hypnosis and transcutaneous electrical nerve stimulation may provide promising reductions in pain. There is a clear need for further high-quality trials to evaluate the benefits and safety of psychological, physical and complementary therapies to guide the management of neuropathic pain.
Collapse
Affiliation(s)
- Nell Norman-Nott
- NeuroRecovery Research Hub, Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia; Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, New South Wales, Australia.
| | - Aidan G Cashin
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, New South Wales, Australia; School of Health Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Sylvia M Gustin
- NeuroRecovery Research Hub, Faculty of Science, University of New South Wales, Sydney, New South Wales, Australia; Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, New South Wales, Australia
| |
Collapse
|
10
|
Song Q, E S, Zhang Z, Liang Y. Neuroplasticity in the transition from acute to chronic pain. Neurotherapeutics 2024; 21:e00464. [PMID: 39438166 PMCID: PMC11585895 DOI: 10.1016/j.neurot.2024.e00464] [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: 06/21/2024] [Revised: 09/10/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
Acute pain is a transient sensation that typically serves as part of the body's defense mechanism. However, in certain patients, acute pain can evolve into chronic pain, which persists for months or even longer. Neuroplasticity refers to the capacity for variation and adaptive alterations in the morphology and functionality of neurons and synapses, and it plays a significant role in the transmission and modulation of pain. In this paper, we explore the molecular mechanisms and signaling pathways underlying neuroplasticity during the transition of pain. We also examine the effects of neurotransmitters, inflammatory mediators, and central sensitization on neuroplasticity, as well as the potential of neuroplasticity as a therapeutic strategy for preventing chronic pain. The aims of this article is to clarify the role of neuroplasticity in the transformation from acute pain to chronic pain, with the hope of providing a novel theoretical basis for unraveling the pathogenesis of chronic pain and offering more effective strategies and approaches for its diagnosis and treatment.
Collapse
Affiliation(s)
- Qingbiao Song
- School of Anesthesiology, Shandong Second Medical University, Weifang 261053, China
| | - Sihan E
- School of Anesthesiology, Shandong Second Medical University, Weifang 261053, China
| | - Zhiyu Zhang
- Department of Orthopedics, Affiliated Hospital of Shandong Second Medical University, Weifang 261035, China
| | - Yingxia Liang
- School of Anesthesiology, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
11
|
Han S, Wang J, Zhang W, Tian X. Chronic Pain-Related Cognitive Deficits: Preclinical Insights into Molecular, Cellular, and Circuit Mechanisms. Mol Neurobiol 2024; 61:8123-8143. [PMID: 38470516 DOI: 10.1007/s12035-024-04073-z] [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: 10/13/2023] [Accepted: 02/23/2024] [Indexed: 03/14/2024]
Abstract
Cognitive impairment is a common comorbidity of chronic pain, significantly disrupting patients' quality of life. Despite this comorbidity being clinically recognized, the underlying neuropathological mechanisms remain unclear. Recent preclinical studies have focused on the fundamental mechanisms underlying the coexistence of chronic pain and cognitive decline. Pain chronification is accompanied by structural and functional changes in the neural substrate of cognition. Based on the developments in electrophysiology and optogenetics/chemogenetics, we summarized the relevant neural circuits involved in pain-induced cognitive impairment, as well as changes in connectivity and function in brain regions. We then present the cellular and molecular alternations related to pain-induced cognitive impairment in preclinical studies, mainly including modifications in neuronal excitability and structure, synaptic plasticity, glial cells and cytokines, neurotransmitters and other neurochemicals, and the gut-brain axis. Finally, we also discussed the potential treatment strategies and future research directions.
Collapse
Affiliation(s)
- Siyi Han
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, Hubei, China
| | - Jie Wang
- Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Wen Zhang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, Hubei, China.
| | - Xuebi Tian
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Avenue, Wuhan, Hubei, China.
| |
Collapse
|
12
|
Sarka BC, Liu S, Banerjee A, Stucky CL, Liu Q, Olsen CM. Neuropathic pain has sex-specific effects on oxycodone-seeking and non-drug-seeking ensemble neurons in the dorsomedial prefrontal cortex of mice. Addict Biol 2024; 29:e13430. [PMID: 39121884 PMCID: PMC11315577 DOI: 10.1111/adb.13430] [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: 05/15/2024] [Revised: 07/06/2024] [Accepted: 07/12/2024] [Indexed: 08/12/2024]
Abstract
Approximately 50 million Americans suffer from chronic pain, and nearly a quarter of chronic pain patients have reported misusing opioid prescriptions. Repeated drug seeking is associated with reactivation of an ensemble of neurons sparsely scattered throughout the dorsomedial prefrontal cortex (dmPFC). Prior research has demonstrated that chronic pain increases intrinsic excitability of dmPFC neurons, which may increase the likelihood of reactivation during drug seeking. We tested the hypothesis that chronic pain would increase oxycodone-seeking behaviour and that the pain state would differentially increase intrinsic excitability in dmPFC drug-seeking ensemble neurons. TetTag mice self-administered intravenous oxycodone. After 7 days of forced abstinence, a drug-seeking session was performed, and the ensemble was tagged. Mice received spared nerve injury (SNI) to induce chronic pain during the period between the first and second seeking session. Following the second seeking session, we performed electrophysiology on individual neurons within the dmPFC to assess intrinsic excitability of the drug-seeking ensemble and non-ensemble neurons. SNI had no impact on sucrose seeking or intrinsic excitability of dmPFC neurons from these mice. In females, SNI increased oxycodone seeking and intrinsic excitability of non-ensemble neurons. In males, SNI had no impact on oxycodone seeking or neuron excitability. Data from females are consistent with clinical reports that chronic pain can promote drug craving and relapse and support the hypothesis that chronic pain itself may lead to neuroadaptations which promote opioid seeking.
Collapse
Affiliation(s)
- Bailey C. Sarka
- Department of Pharmacology and ToxicologyMedical College of WisconsinMilwaukeeWisconsinUSA
- Neuroscience Research CenterMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Shuai Liu
- Department of Pharmacology and ToxicologyMedical College of WisconsinMilwaukeeWisconsinUSA
- Neuroscience Research CenterMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Anjishnu Banerjee
- Division of BiostatisticsMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Cheryl L. Stucky
- Neuroscience Research CenterMedical College of WisconsinMilwaukeeWisconsinUSA
- Department of Cell Biology, Neurobiology and AnatomyMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Qing‐song Liu
- Department of Pharmacology and ToxicologyMedical College of WisconsinMilwaukeeWisconsinUSA
- Neuroscience Research CenterMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Christopher M. Olsen
- Department of Pharmacology and ToxicologyMedical College of WisconsinMilwaukeeWisconsinUSA
- Neuroscience Research CenterMedical College of WisconsinMilwaukeeWisconsinUSA
| |
Collapse
|
13
|
Hu J, Fu J, Cai Y, Chen S, Qu M, Zhang L, Fan W, Wang Z, Zeng Q, Zou J. Bioinformatics and systems biology approach to identify the pathogenetic link of neurological pain and major depressive disorder. Exp Biol Med (Maywood) 2024; 249:10129. [PMID: 38993198 PMCID: PMC11236560 DOI: 10.3389/ebm.2024.10129] [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: 01/25/2024] [Accepted: 06/12/2024] [Indexed: 07/13/2024] Open
Abstract
Neurological pain (NP) is always accompanied by symptoms of depression, which seriously affects physical and mental health. In this study, we identified the common hub genes (Co-hub genes) and related immune cells of NP and major depressive disorder (MDD) to determine whether they have common pathological and molecular mechanisms. NP and MDD expression data was downloaded from the Gene Expression Omnibus (GEO) database. Common differentially expressed genes (Co-DEGs) for NP and MDD were extracted and the hub genes and hub nodes were mined. Co-DEGs, hub genes, and hub nodes were analyzed for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment. Finally, the hub nodes, and genes were analyzed to obtain Co-hub genes. We plotted Receiver operating characteristic (ROC) curves to evaluate the diagnostic impact of the Co-hub genes on MDD and NP. We also identified the immune-infiltrating cell component by ssGSEA and analyzed the relationship. For the GO and KEGG enrichment analyses, 93 Co-DEGs were associated with biological processes (BP), such as fibrinolysis, cell composition (CC), such as tertiary granules, and pathways, such as complement, and coagulation cascades. A differential gene expression analysis revealed significant differences between the Co-hub genes ANGPT2, MMP9, PLAU, and TIMP2. There was some accuracy in the diagnosis of NP based on the expression of ANGPT2 and MMP9. Analysis of differences in the immune cell components indicated an abundance of activated dendritic cells, effector memory CD8+ T cells, memory B cells, and regulatory T cells in both groups, which were statistically significant. In summary, we identified 6 Co-hub genes and 4 immune cell types related to NP and MDD. Further studies are needed to determine the role of these genes and immune cells as potential diagnostic markers or therapeutic targets in NP and MDD.
Collapse
Affiliation(s)
- Jinjing Hu
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Medicine, Southern Medical University, Guangzhou, China
| | - Jia Fu
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Yuxin Cai
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Medicine, Southern Medical University, Guangzhou, China
| | - Shuping Chen
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Medicine, Southern Medical University, Guangzhou, China
| | - Mengjian Qu
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Lisha Zhang
- Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Department of Clinical Medicine, Suzhou Vocational Health College, Suzhou, China
| | - Weichao Fan
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ziyi Wang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Qing Zeng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Medicine, Southern Medical University, Guangzhou, China
| | - Jihua Zou
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Medicine, Southern Medical University, Guangzhou, China
- Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| |
Collapse
|
14
|
Vieira WF, Coelho DRA, Litwiler ST, McEachern KM, Clancy JA, Morales-Quezada L, Cassano P. Neuropathic pain, mood, and stress-related disorders: A literature review of comorbidity and co-pathogenesis. Neurosci Biobehav Rev 2024; 161:105673. [PMID: 38614452 DOI: 10.1016/j.neubiorev.2024.105673] [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: 01/30/2024] [Revised: 04/02/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Neuropathic pain can be caused by multiple factors, and its prevalence can reach 10% of the global population. It is becoming increasingly evident that limited or short-lasting response to treatments for neuropathic pain is associated with psychological factors, which include psychiatric comorbidities known to affect quality of life. It is estimated that 60% of patients with neuropathic pain also experience depression, anxiety, and stress symptoms. Altered mood, including stress, can be a consequence of several painful conditions but can also favor pain chronicization when preexisting. Despite the apparent tight connection between clinical pain and mood/stress disorders, the exact physiological mechanisms remain unclear. This review aims to provide an overview of state-of-the-art research on the mechanisms of pain related to the pathophysiology of depression, anxiety, and stress disorders.
Collapse
Affiliation(s)
- Willians Fernando Vieira
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA; Department of Psychiatry, Harvard Medical School (HMS), Boston, USA; Department of Anatomy, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), São Paulo, Brazil.
| | - David Richer Araujo Coelho
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA; Department of Psychiatry, Harvard Medical School (HMS), Boston, USA; Harvard T. H. Chan School of Public Health (HSPH), Boston, USA
| | - Scott Thomas Litwiler
- Center for Computational and Integrative Biology (CCIB), Massachusetts General Hospital (MGH), Boston, USA
| | - Kayla Marie McEachern
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA
| | - Julie A Clancy
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA
| | - Leon Morales-Quezada
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Boston, USA
| | - Paolo Cassano
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital (MGH), Boston, USA; Department of Psychiatry, Harvard Medical School (HMS), Boston, USA
| |
Collapse
|
15
|
Guida F, Iannotta M, Perrone M, Infantino R, Giorgini G, Fusco A, Marabese I, Manzo I, Belardo C, Di Martino E, Pagano S, Boccella S, Silvestri C, Luongo L, Di Marzo V, Maione S. PEA-OXA restores cognitive impairments associated with vitamin D deficiency-dependent alterations of the gut microbiota. Biomed Pharmacother 2024; 175:116600. [PMID: 38670046 DOI: 10.1016/j.biopha.2024.116600] [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: 02/02/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
There is a growing evidence suggesting the association of vitamin D deficiency (VDD) and cognitive impairment. In this study we evaluated the possible involvement of gut microbiota in the cognitive impairments mediated by VDD and investigated the effects of pharmacological treatment with the oxazoline derivative of the aliamide palmitoylethanolamide, 2-Pentadecyl-2-oxazoline (PEA-OXA). Mice were submitted to behavioural, biochemical and electrophysiological analysis to assess whether their vitamin D status affected cognitive performance together with gut microbiota composition. In VDD mice we found cognitive malfunctioning associated with reduced neuroplasticity, indicated by impaired long term potentiation, and neuroinflammation at the hippocampal level. Importantly, PEA-OXA counteracted the cognitive impairments and modified the biochemical and functional changes induced by VDD. Additionally, PEA-OXA treatment enhanced gut microbiota diversity, which tended to be decreased by VDD only in female mice, elevated the relative abundance of lactic and butyric acid-producing families, i.e. Aerococcaceae and Butyricicoccaceae, and reversed the VDD-induced decrease of butyrate-producing beneficial genera, such as Blautia in female mice, and Roseburia in male mice. These data provide novel insights for a better understanding of the cognitive decline induced by VDD and related gut dysbiosis and its potential therapeutic treatment.
Collapse
Affiliation(s)
- Francesca Guida
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy.
| | - Monica Iannotta
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy
| | - Michela Perrone
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy
| | - Rosmara Infantino
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy
| | - Giada Giorgini
- Centre de Recherche de l'Institut de Cardiologie et Pneumologie de Quebéc (CRIUCPQ) et Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Centre NUTRISS, Department of Medicine and School of Nutrition, Université Laval, Quebec, Canada
| | - Antimo Fusco
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy
| | - Ida Marabese
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy
| | - Iolanda Manzo
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy
| | - Carmela Belardo
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy
| | - Emanuele Di Martino
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy
| | - Salvatore Pagano
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy
| | - Serena Boccella
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy
| | - Cristoforo Silvestri
- Centre de Recherche de l'Institut de Cardiologie et Pneumologie de Quebéc (CRIUCPQ) et Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Centre NUTRISS, Department of Medicine and School of Nutrition, Université Laval, Quebec, Canada
| | - Livio Luongo
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy
| | - Vincenzo Di Marzo
- Centre de Recherche de l'Institut de Cardiologie et Pneumologie de Quebéc (CRIUCPQ) et Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Centre NUTRISS, Department of Medicine and School of Nutrition, Université Laval, Quebec, Canada; Endocannabinoid Research Group, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR), Pozzuoli, Italy; Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, and Joint International Research Unit between the Consiglio Nazionale delle Ricerche and Université Laval on Chemical and Biomolecular Studies on the Microbiome and its Impact on Metabolic Health and Nutrition, Canada
| | - Sabatino Maione
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy.
| |
Collapse
|
16
|
Yao R, Man Y, Lu Y, Su Y, Zhou M, Wang S, Gu X, Wang R, Wu Y, Wang L. Infliximab alleviates memory impairment in rats with chronic pain by suppressing neuroinflammation and restoring hippocampal neurogenesis. Neuropharmacology 2024; 245:109813. [PMID: 38110173 DOI: 10.1016/j.neuropharm.2023.109813] [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: 10/20/2023] [Revised: 11/30/2023] [Accepted: 12/06/2023] [Indexed: 12/20/2023]
Abstract
Patients with chronic pain commonly report impaired memory. Increasing evidence has demonstrated that inhibition of neurogenesis by neuroinflammation plays a crucial role in chronic pain-associated memory impairments. There is currently a lack of treatment strategies for this condition. An increasing number of clinical trials have reported the therapeutic potential of anti-inflammatory therapies targeting tumour necrosis factor-α (TNF-α) for inflammatory diseases. The present study investigated whether infliximab alleviates chronic pain-associated memory impairments in rats with chronic constriction injury (CCI). We demonstrated that infliximab alleviated spatial memory impairment and hyperalgesia induced by CCI. Furthermore, infliximab inhibited the activation of hippocampal astrocytes and microglia and decreased the release of proinflammatory cytokines in CCI rats. Furthermore, infliximab reversed the decrease in the numbers of newborn neurons and mature neurons in the dentate gyrus (DG) caused by chronic pain. Our data provide evidence that infliximab alleviates chronic pain-associated memory impairments, suppresses neuroinflammation and restores hippocampal neurogenesis in a CCI model. These facts indicate that infliximab may be a potential therapeutic agent for the treatment of chronic pain and associated memory impairments.
Collapse
Affiliation(s)
- Rui Yao
- Department of Anesthesiology, Xuzhou First People's Hospital, Xuzhou, 22100, China
| | - Yuanyuan Man
- Department of Respiratory Medicine, Xuzhou Central Hospital, Xuzhou, 22100, China
| | - Yao Lu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 22100, China
| | - Yang Su
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 22100, China
| | - Meiyan Zhou
- Department of Anesthesiology, Xuzhou Central Hospital, Xuzhou, 22100, China
| | - Shuang Wang
- Department of Anesthesiology, Xuzhou Central Hospital, Xuzhou, 22100, China
| | - Xiaoping Gu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Rongguo Wang
- Department of Anesthesiology, Xuzhou Central Hospital, Xuzhou, 22100, China.
| | - Yuqing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 22100, China.
| | - Liwei Wang
- Department of Anesthesiology, Xuzhou Central Hospital, Xuzhou, 22100, China.
| |
Collapse
|
17
|
Han S, Yuan X, Zhao F, Manyande A, Gao F, Wang J, Zhang W, Tian X. Activation of LXRs alleviates neuropathic pain-induced cognitive dysfunction by modulation of microglia polarization and synaptic plasticity via PI3K/AKT pathway. Inflamm Res 2024; 73:157-174. [PMID: 38183431 DOI: 10.1007/s00011-023-01826-9] [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: 08/06/2023] [Revised: 10/20/2023] [Accepted: 11/22/2023] [Indexed: 01/08/2024] Open
Abstract
OBJECTIVE Cognitive dysfunction is a common comorbidity in patients with chronic pain. Activation of Liver X receptors (LXRs) plays a potential role in improving cognitive disorders in central nervous diseases. In this study, we investigated the role of LXRs in cognitive deficits induced by neuropathic pain. METHODS We established the spared nerve injury (SNI) model to investigate pain-induced memory dysfunction. Pharmacological activation of LXRs with T0901317 or inhibition with GSK2033 was applied. PI3K inhibitor LY294002 was administered to explore the underlying mechanism of LXRs. Changes in neuroinflammation, microglia polarization, and synaptic plasticity were assessed using biochemical technologies. RESULTS We found that SNI-induced cognitive impairment was associated with reduced LXRβ expression, increased M1-phenotype microglia, decreased synaptic proteins, and inhibition of PI3K/AKT signaling pathway in the hippocampus. Activation of LXRs using T0901317 effectively alleviated SNI-induced cognitive impairment. Additionally, T0901317 promoted the polarization of microglia from M1 to M2, reduced pro-inflammatory cytokines, and upregulated synaptic proteins in the hippocampus. However, administration of GSK2033 or LY294002 abolished these protective effects of T0901317 in SNI mice. CONCLUSIONS LXRs activation alleviates neuropathic pain-induced cognitive impairment by modulating microglia polarization, neuroinflammation, and synaptic plasticity, at least partly via activation of PI3K/AKT signaling in the hippocampus. LXRs may be promising targets for addressing pain-related cognitive deficits.
Collapse
Affiliation(s)
- Siyi Han
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, China
| | - Xiaoman Yuan
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, China
| | - Fengtian Zhao
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, China
| | - Anne Manyande
- School of Human and Social Sciences, University of West London, London, UK
| | - Feng Gao
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, China
| | - Jie Wang
- Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, Hubei Province, China
| | - Wen Zhang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, China.
| | - Xuebi Tian
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, China.
| |
Collapse
|
18
|
Bonsale R, Infantino R, Perrone M, Marabese I, Ricciardi F, Fusco A, Teweldemedhin MM, Boccella S, Guida F, Rinaldi B. The long-term exercise after traumatic brain injury: Reharmonizing brain by sound body. Brain Res 2023; 1816:148471. [PMID: 37356701 DOI: 10.1016/j.brainres.2023.148471] [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: 03/29/2023] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 06/27/2023]
Abstract
Traumatic brain injuries (TBI) refer to multiple acquired dysfunctions arising from damage to the brain caused by an external force, including rapid acceleration/deceleration and concussion. Among them, mild TBI (mTBI) accounts for most cases (up to 90%) of injuries. It is responsible for a variety of symptoms, including anxiety, depression, and cognitive impairments that remain difficult to be treated. It has been reported that regular physical activity, as well as, improving life quality, display a neuroprotective function, suggesting a possible role in post-traumatic rehabilitation. In this study, we investigated the effects of treadmill exercise in a mice mTBI model by behavioural, electrophysiological and neurochemical analysis. Daily exercise decreased anxiety, aggressive behavior, and depression in mTBI mice. Accordingly, electrophysiological and neurochemical maladaptive rearrangement occurring in the hippocampus of mTBI mice were prevented by the exercise.
Collapse
Affiliation(s)
- Roozbe Bonsale
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Rosmara Infantino
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Michela Perrone
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Ida Marabese
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Federica Ricciardi
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Antimo Fusco
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Milena Melake Teweldemedhin
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Serena Boccella
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Barbara Rinaldi
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy.
| |
Collapse
|
19
|
Amodeo G, Franchi S, D’Agnelli S, Galimberti G, Baciarello M, Bignami EG, Sacerdote P. Supraspinal neuroinflammation and anxio-depressive-like behaviors in young- and older- adult mice with osteoarthritis pain: the effect of morphine. Psychopharmacology (Berl) 2023; 240:2131-2146. [PMID: 37530884 PMCID: PMC10506934 DOI: 10.1007/s00213-023-06436-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/24/2023] [Indexed: 08/03/2023]
Abstract
RATIONALE Asteoarthritis (OA) is a leading cause of chronic pain in the elderly population and is often associated with emotional comorbidities such as anxiety and depression. Despite age is a risk factor for both OA and mood disorders, preclinical studies are mainly conducted in young adult animals. OBJECTIVES Here, using young adult (11-week-old) and older adult (20-month-old) mice, we evaluate in a monosodium-iodoacetate-(MIA)-induced OA model the development of anxio-depressive-like behaviors and whether brain neuroinflammation may underlie the observed changes. We also test whether an effective pain treatment may prevent behavioral and biochemical alterations. METHODS Mechanical allodynia was monitored throughout the experimental protocol, while at the end of protocol (14 days), anxio-depressive-like behaviors and cognitive dysfunction were assessed. Neuroinflammatory condition was evaluated in prefrontal cortex, hippocampus and hypothalamus. Serum IFNγ levels were also measured. Moreover, we test the efficacy of a 1-week treatment with morphine (2.5 mg/kg) on pain, mood alterations and neuroinflammation. RESULTS We observed that young adult and older adult controls (CTRs) mice had comparable allodynic thresholds and developed similar allodynia after MIA injection. Older adult CTRs were characterized by altered behavior in the tests used to assess the presence of depression and cognitive impairment and by elevated neuroinflammatory markers in brain areas compared to younger ones. The presence of pain induced depressive-like behavior and neuroinflammation in adult young mice, anxiety-like behavior in both age groups and worsened neuroinflammation in older adult mice. Morphine treatment counteracted pain, anxio-depressive behaviors and neuroinflammatory activation in both young adult and older adult mice. CONCLUSIONS Here, we demonstrated that the presence of chronic pain in young adult mice induces mood alterations and supraspinal biochemical changes and aggravates the alterations already evident in older adult animals. A treatment with morphine, counteracting the pain, prevents the development of anxio-depressive disorders and reduces neuroinflammation.
Collapse
Affiliation(s)
- Giada Amodeo
- Dipartimento Di Scienze Farmacologiche E Biomolecolari, University of Milan, Via Vanvitelli 32, 20129 Milano, Italy
| | - Silvia Franchi
- Dipartimento Di Scienze Farmacologiche E Biomolecolari, University of Milan, Via Vanvitelli 32, 20129 Milano, Italy
| | - Simona D’Agnelli
- Anesthesiology, Critical Care and Pain Medicine Division, Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Giulia Galimberti
- Dipartimento Di Scienze Farmacologiche E Biomolecolari, University of Milan, Via Vanvitelli 32, 20129 Milano, Italy
| | - Marco Baciarello
- Anesthesiology, Critical Care and Pain Medicine Division, Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Elena Giovanna Bignami
- Anesthesiology, Critical Care and Pain Medicine Division, Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Paola Sacerdote
- Dipartimento Di Scienze Farmacologiche E Biomolecolari, University of Milan, Via Vanvitelli 32, 20129 Milano, Italy
| |
Collapse
|
20
|
Galimberti G, Amodeo G, Magni G, Riboldi B, Balboni G, Onnis V, Ceruti S, Sacerdote P, Franchi S. Prokineticin System Is a Pharmacological Target to Counteract Pain and Its Comorbid Mood Alterations in an Osteoarthritis Murine Model. Cells 2023; 12:2255. [PMID: 37759478 PMCID: PMC10526764 DOI: 10.3390/cells12182255] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent joint disease associated with chronic pain. OA pain is often accompanied by mood disorders. We addressed the role of the Prokineticin (PK) system in pain and mood alterations in a mice OA model induced with monosodium iodoacetate (MIA). The effect of a PK antagonist (PC1) was compared to that of diclofenac. C57BL/6J male mice injected with MIA in the knee joint were characterized by allodynia, motor deficits, and fatigue. Twenty-eight days after MIA, in the knee joint, we measured high mRNA of PK2 and its receptor PKR1, pro-inflammatory cytokines, and MMP13. At the same time, in the sciatic nerve and spinal cord, we found increased levels of PK2, PKR1, IL-1β, and IL-6. These changes were in the presence of high GFAP and CD11b mRNA in the sciatic nerve and GFAP in the spinal cord. OA mice were also characterized by anxiety, depression, and neuroinflammation in the prefrontal cortex and hippocampus. In both stations, we found increased pro-inflammatory cytokines. In addition, PK upregulation and reactive astrogliosis in the hippocampus and microglia reactivity in the prefrontal cortex were detected. PC1 reduced joint inflammation and neuroinflammation in PNS and CNS and counteracted OA pain and emotional disturbances.
Collapse
Affiliation(s)
- Giulia Galimberti
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (G.G.); (G.A.); (G.M.); (B.R.); (S.C.); (P.S.)
| | - Giada Amodeo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (G.G.); (G.A.); (G.M.); (B.R.); (S.C.); (P.S.)
| | - Giulia Magni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (G.G.); (G.A.); (G.M.); (B.R.); (S.C.); (P.S.)
| | - Benedetta Riboldi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (G.G.); (G.A.); (G.M.); (B.R.); (S.C.); (P.S.)
| | - Gianfranco Balboni
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (G.B.); (V.O.)
| | - Valentina Onnis
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (G.B.); (V.O.)
| | - Stefania Ceruti
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (G.G.); (G.A.); (G.M.); (B.R.); (S.C.); (P.S.)
| | - Paola Sacerdote
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (G.G.); (G.A.); (G.M.); (B.R.); (S.C.); (P.S.)
| | - Silvia Franchi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (G.G.); (G.A.); (G.M.); (B.R.); (S.C.); (P.S.)
| |
Collapse
|
21
|
Jin J, Zhang T, Xiong X, Chen H, Jiang Y, He S. A prospective study of chronic postsurgical pain in elderly patients: incidence, characteristics and risk factors. BMC Geriatr 2023; 23:289. [PMID: 37173634 PMCID: PMC10182592 DOI: 10.1186/s12877-023-04006-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Due to the continued growth of surgical procedures in older adults and the significant impact of chronic postsurgical pain (CPSP), it is crucial to improve our understanding of the occurrence of CPSP as well as the appropriate prevention and treatment. We therefore conducted this study to determine the incidence, characteristics and risk factors of CPSP in elderly patients at both 3 and 6 months after surgery. METHODS Elderly patients (aged ≥ 60 years) undergoing elective surgery in our institution between April 2018 and March 2020 were prospectively enrolled in this study. Data on demographics, preoperative psychological well-being, intraoperative surgical and anesthesia management, and acute postoperative pain intensity were collected. At 3 and 6 months after surgery, patients received telephone interview and completed the questionnaires regarding chronic pain characteristics, analgesic consumption, and interference of the pain with activities of daily living (ADL). RESULTS A total of 1065 elderly patients were followed up for 6 postoperative months and included in final analysis. At 3 and 6 months after operation, the incidence of CPSP was 35.6% [95% confidence interval (95% CI) 32.7 - 38.8%] and 21.5% (95% CI 19.0% - 23.9%), respectively. CPSP cause negative impacts on patient's ADL and most particularly on mood. Neuropathic features were found in 45.1% of the patients with CPSP at 3 months. At 6 months, 31.0% of those with CPSP reported that the pain had neuropathic features. Preoperative anxiety [3 months: Odds ratio (OR) 2.244, 95% CI 1.693 to 2.973; 6 months: OR 2.397, 95% CI 1.745 to 3.294], preoperative depression (3 months: OR 1.709, 95% CI 1.292 to 2.261; 6 months: OR 1.565, 95% CI 1.136-2.156), orthopedic surgery (3 months: OR 1.927, 95% CI 1.112 to 3.341; 6 months: OR 2.484, 95% CI 1.220 to 5.061), higher pain severity on movement within postoperative 24 h (3 months: OR 1.317, 95% CI 1.191 to 1.457; 6 months: OR 1.317, 95% CI 1.177 to 1.475) were associated with a higher risk for CPSP independently at both 3 and 6 months after surgery. CONCLUSIONS CPSP is a common postoperative complication in elderly surgical patients. Preoperative anxiety and depression, orthopedic surgery, and greater intensity of acute postoperative pain on movement are associated with an increased risk for CPSP. It should be kept in mind that developing psychological interventions to reduce anxiety and depression and optimizing the management of acute postoperative pain will be effective in reducing the development of CPSP in this population.
Collapse
Affiliation(s)
- Juying Jin
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China.
| | - Ting Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China
| | - Xianwei Xiong
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China
| | - Huan Chen
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China
| | - Yiling Jiang
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China
| | - Shuangyu He
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China
| |
Collapse
|
22
|
Schulte A, Lohner H, Degenbeck J, Segebarth D, Rittner HL, Blum R, Aue A. Unbiased analysis of the dorsal root ganglion after peripheral nerve injury: no neuronal loss, no gliosis, but satellite glial cell plasticity. Pain 2023; 164:728-740. [PMID: 35969236 PMCID: PMC10026836 DOI: 10.1097/j.pain.0000000000002758] [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: 06/04/2022] [Revised: 07/13/2022] [Accepted: 07/26/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT Pain syndromes are often accompanied by complex molecular and cellular changes in dorsal root ganglia (DRG). However, the evaluation of cellular plasticity in the DRG is often performed by heuristic manual analysis of a small number of representative microscopy image fields. In this study, we introduce a deep learning-based strategy for objective and unbiased analysis of neurons and satellite glial cells (SGCs) in the DRG. To validate the approach experimentally, we examined serial sections of the rat DRG after spared nerve injury (SNI) or sham surgery. Sections were stained for neurofilament, glial fibrillary acidic protein (GFAP), and glutamine synthetase (GS) and imaged using high-resolution large-field (tile) microscopy. After training of deep learning models on consensus information of different experts, thousands of image features in DRG sections were analyzed. We used known (GFAP upregulation), controversial (neuronal loss), and novel (SGC phenotype switch) changes to evaluate the method. In our data, the number of DRG neurons was similar 14 d after SNI vs sham. In GFAP-positive subareas, the percentage of neurons in proximity to GFAP-positive cells increased after SNI. In contrast, GS-positive signals, and the percentage of neurons in proximity to GS-positive SGCs decreased after SNI. Changes in GS and GFAP levels could be linked to specific DRG neuron subgroups of different size. Hence, we could not detect gliosis but plasticity changes in the SGC marker expression. Our objective analysis of DRG tissue after peripheral nerve injury shows cellular plasticity responses of SGCs in the whole DRG but neither injury-induced neuronal death nor gliosis.
Collapse
Affiliation(s)
- Annemarie Schulte
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Hannah Lohner
- Department of Anesthesiology, Center for Interdisciplinary Pain Medicine, Intensive Care, Emergency Medicine and Pain Therapy, University Hospital of Würzburg, Würzburg, Germany
| | - Johannes Degenbeck
- Department of Anesthesiology, Center for Interdisciplinary Pain Medicine, Intensive Care, Emergency Medicine and Pain Therapy, University Hospital of Würzburg, Würzburg, Germany
| | - Dennis Segebarth
- Institute of Clinical Neurobiology, University Hospital of Würzburg, Würzburg, Germany
| | - Heike L. Rittner
- Department of Anesthesiology, Center for Interdisciplinary Pain Medicine, Intensive Care, Emergency Medicine and Pain Therapy, University Hospital of Würzburg, Würzburg, Germany
| | - Robert Blum
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Annemarie Aue
- Department of Anesthesiology, Center for Interdisciplinary Pain Medicine, Intensive Care, Emergency Medicine and Pain Therapy, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
23
|
Tian S, Wu L, Zheng H, Zhong X, Yu X, Wu W. Identification of autophagy-related genes in neuropathic pain through bioinformatic analysis. Hereditas 2023; 160:8. [PMID: 36855217 PMCID: PMC9976393 DOI: 10.1186/s41065-023-00269-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 02/16/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Neuropathic pain (NP) is one of the most common types of chronic pain and significantly compromises the quality of life. Autophagy is an intracellular catabolic process that is required to maintain cellular homeostasis in response to various stresses. The role of autophagy-related genes in the diagnosis and treatment of neuropathic pain remains unclear. METHODS We identified autophagy-related differentially expressed genes (ARDEGs) and differentially expressed miRNAs (DE-miRNAs) in neuropathic pain by bioinformatics analysis of the GSE145226 and GSE145199 datasets. These ARDEGs and their co-expressed genes were subjected to Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, Gene Set Enrichment Analysis (GSEA) and friends analysis. Meanwhile, we constructed TFs-ARDEGs, miRNA-ARDEGs regulatory network through ChIPBase database and the HTFtarget database, multiMir R package. Finally, we performed immune infiltration analysis of ARDEGs by Single Sample Gene Set Enrichment Analysis (ssGSEA). RESULTS We identified 2 potential autophagy-related differentially expressed genes (Sirt2 and ST7) that may be closely associated with the pathogenesis of neuropathic pain. GO, KEGG and GSEA analysis revealed that these two ARDEGs were mainly enriched in pyridine nucleotide metabolic process, nicotinamide nucleotide metabolic process, Nicotinate and nicotinamide metabolism, NF-κB pathway, KRAS signaling, P53 pathway. In the TFs-ARDEGs and miRNA-ARDEGs regulatory network, miR-140-5p and Cebpb were predicted to be as crucial regulators in the progression of NP. For the ssGSEA results, Sirt2 was positively correlated with Eosinophil and Effector memory CD8+ T cell infiltration, which suggested that it may be involved in the regulation of neuroimmune-related signaling. CONCLUSION Two autophagy-related differentially expressed genes, especially Sirt2, may be potential biomarkers for NP, providing more evidence about the crucial role of autophagy in neuropathic pain.
Collapse
Affiliation(s)
- Sheng Tian
- grid.412455.30000 0004 1756 5980Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006 China
| | - Lanxiang Wu
- grid.412455.30000 0004 1756 5980Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006 China
| | - Heqing Zheng
- grid.412455.30000 0004 1756 5980Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006 China
| | - Xianhui Zhong
- grid.412455.30000 0004 1756 5980Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006 China
| | - Xinping Yu
- grid.412455.30000 0004 1756 5980Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006 China
| | - Wei Wu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
24
|
Infantino R, Boccella S, Scuteri D, Perrone M, Ricciardi F, Vitale R, Bonsale R, Parente A, Allocca I, Virtuoso A, De Luca C, Belardo C, Amodeo P, Gentile V, Cirillo G, Bagetta G, Luongo L, Maione S, Guida F. 2-pentadecyl-2-oxazoline prevents cognitive and social behaviour impairments in the Amyloid β-induced Alzheimer-like mice model: Bring the α2 adrenergic receptor back into play. Biomed Pharmacother 2022; 156:113844. [DOI: 10.1016/j.biopha.2022.113844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/03/2022] [Accepted: 10/06/2022] [Indexed: 11/15/2022] Open
|
25
|
Sun X, Wang C, Wu J, Chen X, He H. Effect of TGF- β1-Mediated Exercise Analgesia in Spared Nerve Injury Mice. Neural Plast 2022; 2022:7382327. [PMID: 36504685 PMCID: PMC9729053 DOI: 10.1155/2022/7382327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/30/2022] [Accepted: 10/12/2022] [Indexed: 01/07/2023] Open
Abstract
Peripheral nerve injury leads to severe neuropathic pain. Previous studies have highlighted the beneficial effects of physical exercise on alleviating neuropathic pain. Exercise regulating transforming growth factor-β1 (TGF-β1) can improve several diseases and relieve neuropathic pain induced by peripheral nerve injury. Here, we investigated whether exercise could alleviate neuropathic pain by modulating TGF-β1 expression. We assessed mechanical and cold pain behavior and conducted molecular evaluation of the spinal cord. We found that spared nerve injury (SNI) led to mechanical and cold allodynia in the hind paw, elevated the expression of latency-associated peptide- (LAP-) TGF-β1, and activated astroglial in the spinal cord. Exercise decreases allodynia, astroglial activation, and LAP-TGF-β1 in SNI mice. Intrathecal injection of a TGF-type I receptor inhibitor attenuated exercise analgesia and enhanced astroglial activation. These findings demonstrate that exercise induces analgesia by promoting TGF-β1 activation and inhibiting astrogliosis. Our study reveals a new underlying mechanism for exercise-attenuated neuropathic pain in the maintenance stage of neuropathic pain after nerve injury.
Collapse
Affiliation(s)
- Xinzheng Sun
- School of Sports Science, Beijing Sport University, Beijing 100084, China
| | - Chenghao Wang
- School of Sports Science, Beijing Sport University, Beijing 100084, China
| | - Junqi Wu
- School of Sports Science, Beijing Sport University, Beijing 100084, China
| | - Xiaoke Chen
- School of Sports Science, Beijing Sport University, Beijing 100084, China
| | - Hui He
- China Institute of Sports and Health, Beijing Sport University, Beijing 100084, China
| |
Collapse
|
26
|
LANCL1 as the Key Immune Marker in Neuropathic Pain. Neural Plast 2022; 2022:9762244. [PMID: 35510269 PMCID: PMC9061068 DOI: 10.1155/2022/9762244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 03/21/2022] [Indexed: 11/17/2022] Open
Abstract
Objective. This study is to explore key immune markers and changes of immune microenvironment in neuropathic pain (NeuP). Method. The data sets of GSE145199 and GSE145226 in Gene Expression Omnibus (GEO) database was used to analyze, and the key immune markers were verified by GSE70006 and GSE91396, and the infiltration degree of immune cells in different samples were analyzed by CIBERSORT analysis package. Results. In this study, we found a key immune marker, namely, LANCL1. Regulatory axis closely related to LANCL1 has also been found, namely, miR-6325/LANCL1 axis. In the immune infiltration analysis, we also found that the LANCL1 is positively correlated with T cells CD4 naïve (
,
). Conclusion. In this study, we found that LANCL1 may be a protective factor for NeuP, and the miR-6325/LANCL1 axis may be involved in the occurrence and development of NeuP. Cascade reactions including mast cells, macrophages, and T cells may be an important reason for the aggravation of nerve damage.
Collapse
|