1
|
Xue T, Song Y, Zhao J, Fan G, Liu Z. Inhibition of S100A4 decreases neurotoxic astrocyte reactivity and attenuates neuropathic pain via the TLR4/NF-κB pathway in a rat model of spinal nerve ligation. J Headache Pain 2025; 26:97. [PMID: 40312684 PMCID: PMC12044810 DOI: 10.1186/s10194-025-02045-9] [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: 03/07/2025] [Accepted: 04/23/2025] [Indexed: 05/03/2025] Open
Abstract
S100A4 participates in inflammation and immune reactions in the central nervous system and is involved in the pathogenesis of multiple neurological disorders. It can affect the functions of astrocytes, microglia, infiltrating cells and neurons and further modulates neuronal plasticity and survival in the central nervous system. However, its impact on astrocyte phenotypes and neuropathic pain and the intrinsic mechanisms involved remain poorly understood. Here, we showed that S100A4 was markedly upregulated after spinal nerve ligation and was mainly expressed in neurons in the spinal dorsal horn. Transcriptional inhibition of S100A4 with niclosamide attenuated neuropathic pain after surgery. We found that astrocytes differentiated into C3-positive reactive populations, so-called neurotoxic (A1) astrocytes and identified differentially expressed genes and specific molecular expression signatures after ligation. Neurotoxic astrocyte reactivity is regulated by exogenous S100A4 in vitro, and targeted inhibition of S100A4 suppresses neurotoxic astrocyte proliferation in rats. Finally, we reported that TLR4/NF-κB signaling pathway is a downstream of S100A4 activation, and that specific depletion this pathway suppresses deleterious A1 astrocyte activation and further attenuates the development and maintenance of neuropathic pain after spinal nerve ligation. Thus, S100A4 in neurons plays a key role in neurotoxic astrocyte reactivity and can be targeted for treatment to prevent and alleviate neuropathic pain.
Collapse
Affiliation(s)
- Tao Xue
- Department of Orthopedics, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213003, China
| | - Yu Song
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China
| | - Jie Zhao
- Department of Orthopedics, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213003, China
| | - Guiyong Fan
- Department of Orthopedics, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, 215028, China
| | - Zhiyuan Liu
- Department of Orthopedics, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213003, China.
- The Wujin Clinical College of Xuzhou Medical University, Changzhou, 213003, China.
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Changzhou, 213003, China.
| |
Collapse
|
2
|
Carvalhas-Almeida C, Sehgal A. Glia: the cellular glue that binds circadian rhythms and sleep. Sleep 2025; 48:zsae314. [PMID: 39812780 PMCID: PMC11893543 DOI: 10.1093/sleep/zsae314] [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: 09/16/2024] [Revised: 11/19/2024] [Indexed: 01/16/2025] Open
Abstract
Glia are increasingly appreciated as serving an important function in the control of sleep and circadian rhythms. Glial cells in Drosophila and mammals regulate daily rhythms of locomotor activity and sleep as well as homeostatic rebound following sleep deprivation. In addition, they contribute to proposed functions of sleep, with different functions mapping to varied glial subtypes. Here, we discuss recent findings in Drosophila and rodent models establishing a role of glia in circadian or sleep regulation of synaptic plasticity, brain metabolism, removal of cellular debris, and immune challenges. These findings underscore the relevance of glia for benefits attributed to sleep and have implications for understanding the neurobiological mechanisms underlying sleep and associated disorders.
Collapse
Affiliation(s)
- Catarina Carvalhas-Almeida
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Amita Sehgal
- Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Howard Hughes Medical Institute, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
3
|
Wu F, Fu C, Li Y, Wang H. TNF-α Enhanced Activity of Sympathetic Neurons in Superior Cervical Ganglion to Promote Chronic Sleep Deprivation-Related Hyperalgesia. Mol Neurobiol 2025:10.1007/s12035-025-04790-z. [PMID: 39992586 DOI: 10.1007/s12035-025-04790-z] [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: 04/23/2024] [Accepted: 02/17/2025] [Indexed: 02/26/2025]
Abstract
The mechanisms underlying the association between sleep deprivation (SD) and hyperalgesia remain incompletely understood. In this study, the Modified Horizontal Platform Method was employed to induce chronic SD. Neuropathic pain was induced using chronic constriction injury of the sciatic nerve. Pain-like behaviors were assessed through measurements of mechanical allodynia and thermal hyperalgesia, while gait analysis was used to evaluate motor function. Immunofluorescence and western blot analyses were conducted to examine the expression of TNF-α, Iba-1, TH, neurons and c-Fos. Apoptosis was assessed using TUNEL staining. To explore anatomical connections, anterograde and retrograde tracer viruses were injected into the superior cervical ganglion (SCG) and the spinal cord, respectively. Local injection of 6-OHDA was used to ablate sympathetic neurons in the SCG, and R-7050 was administrated to block the TNF-α receptor. We found that chronic SD induced hyperalgesia in both normal and neuropathic pain model, accompanied by significant infiltration of microglia in the dorsal horn. TH expression and apoptotic cells were increased in the SCG following chronic SD. Viral tracer results demonstrated the existence of anatomical connections between the SCG and the spinal cord. Ablation of sympathetic innervation improved pain-like behaviors and reduced microglia, without affecting movement. Furthermore, chronic SD led to increased expression of TNF-α in sympathetic neurons, which was associated with heightened SCG activity. Blocking the TNF-α receptor ameliorated pain-like behaviors, decreased microglia, reduced apoptosis, lowered SCG activity. In conclusion, TNF-α enhanced the activity of sympathetic neurons in the SCG, promoting hyperalgesia related to chronic SD.
Collapse
Affiliation(s)
- Fancan Wu
- Department of Anesthesiology, The First People's Hospital of Foshan, Number 81, North of Ling Nan Road, Foshan, 528000, Guangdong, China
| | - Chao Fu
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Number 613, The West of Huangpu Avenue, Tianhe Region, Guangzhou, 510630, Guangdong Province, China
| | - Yalan Li
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Number 613, The West of Huangpu Avenue, Tianhe Region, Guangzhou, 510630, Guangdong Province, China.
| | - Hanbing Wang
- Department of Anesthesiology, The First People's Hospital of Foshan, Number 81, North of Ling Nan Road, Foshan, 528000, Guangdong, China.
| |
Collapse
|
4
|
Chen S, Xie Y, Liang Z, Liu J, Wang J, Mao Y, Xing F, Wei X, Wang Z, Yang J, Yuan J. Sleep deprivation affects pain sensitivity by increasing oxidative stress and apoptosis in the medial prefrontal cortex of rats via the HDAC2-NRF2 pathway. Biomed J 2025:100826. [PMID: 39755172 DOI: 10.1016/j.bj.2024.100826] [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: 02/16/2024] [Revised: 09/03/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025] Open
Abstract
Sleep is crucial for sustaining normal physiological functions, and sleep deprivation has been associated with increased pain sensitivity. The histone deacetylases (HDACs) are known to significantly regulate in regulating neuropathic pain, but their involvement in nociceptive hypersensitivity during sleep deprivation is still not fully understood. Utilizing a modified multi-platform water environment technique to establish a sleep deprivation model. We measured the expression levels of HDAC1/2 in the medial prefrontal cortex (mPFC) through immunoblotting and real-time quantitative PCR. The presence of pyroptosis was determined using a TUNEL assay. Suberoylanilide hydroxamic acid (SAHA), an HDAC inhibitor employed clinically, was injected into the peritoneal cavity to inhibit HDAC2 expression. Animal pain behaviors were evaluated by measuring paw withdrawal thresholds (PWTs) and paw withdrawal latencies (PWLs). Our findings indicate that sleep deprivation leads to increased nociceptive hypersensitivity, an upregulation of HDAC2 expression in the mPFC, a downregulation of the expression of nuclear factor erythroid 2-related factor 2 (NRF2), and changes in markers of oxidative stress in rats. SAHA, the HDAC inhibitor, enhanced NRF2 expression by inhibiting HDAC2, which consequently ameliorated oxidative stress and mitigated nociceptive hypersensitivity in rats. The incidence of apoptosis was found to be higher in the mPFC tissues of sleep deprivation rats, and the intraperitoneal administration of SAHA decreased this apoptosis. The co-injection of SAHA and the NRF2 inhibitor ML385 into sleep deprivation rats negated the beneficial effects of SAHA. In conclusion, HDAC2 is implicated in the induction of oxidative stress and apoptosis by suppressing NRF2 levels, thereby exacerbating nociceptive hypersensitivity in sleep deprivation rats.
Collapse
Affiliation(s)
- Shuhan Chen
- Department of Anesthesiology, Perioperative and Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450000, China; Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, Henan Province 450000, China
| | - Yanle Xie
- Department of Anesthesiology, Perioperative and Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450000, China; Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, Henan Province 450000, China
| | - Zenghui Liang
- Department of Anesthesiology, Perioperative and Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450000, China; Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, Henan Province 450000, China
| | - Jing Liu
- Department of Anesthesiology, Perioperative and Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450000, China; Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, Henan Province 450000, China
| | - Jingping Wang
- Massachusetts General Hospital Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA
| | - Yuanyuan Mao
- Department of Anesthesiology, Perioperative and Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450000, China; Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, Henan Province 450000, China
| | - Fei Xing
- Department of Anesthesiology, Perioperative and Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450000, China; Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, Henan Province 450000, China
| | - Xin Wei
- Department of Anesthesiology, Perioperative and Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450000, China; Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, Henan Province 450000, China
| | - Zhongyu Wang
- Department of Anesthesiology, Perioperative and Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450000, China; Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, Henan Province 450000, China
| | - Jianjun Yang
- Department of Anesthesiology, Perioperative and Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450000, China; Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, Henan Province 450000, China
| | - Jingjing Yuan
- Department of Anesthesiology, Perioperative and Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450000, China; Henan Province International Joint Laboratory of Pain, Cognition and Emotion, Zhengzhou, Henan Province 450000, China.
| |
Collapse
|
5
|
Martínez-Magaña CJ, Murbartián J. Estrogen receptor α regulates the IKKs/NF-kB activity involved in the development of mechanical allodynia induced by REM sleep deprivation in rats. Brain Res 2024; 1845:149269. [PMID: 39384127 DOI: 10.1016/j.brainres.2024.149269] [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: 07/04/2024] [Revised: 09/11/2024] [Accepted: 10/06/2024] [Indexed: 10/11/2024]
Abstract
Several signaling pathways that converge in NF-kB activation have been linked to developing and maintaining different types of pathological pain. In addition, some mechanisms implied in the establishment of chronic pain have been demonstrated to have a sex-dependent correlation. This study aimed to determine if the IKKs/NF-kB signaling pathway is involved in establishing REM sleep deprivation (REMSD) induced mechanical allodynia in rats and its possible regulation depending on estradiol and estrogen receptors. Intrathecal administration of BMS-345541 or minocycline, two drugs that reduce the IKKs/NF-kB activity, avoided the development of mechanical allodynia in female but not in male rats subjected to 48 h of REMSD. Ovariectomy in female rats abolished the effect of BMS-345541 and minocycline. Meanwhile, the 17-β-estradiol restitution restored it. Intrathecal administration of MPP, a selective ERα antagonist, but not PHTPP, a selective ERβ antagonist, avoided the effect of BMS-345541 in female rats without hormonal manipulation. In addition, the transient run-down of ERα in female rats abolished the effect of BMS-345541. All data suggest an important role of ERα as a regulator of the IKKs/NF-kB activity. REMSD increased the ERα protein expression in the dorsal root ganglia and the dorsal spinal cord in females but not in male rats. Interestingly, ERα activation or ERα overexpression allowed the effect of BMS-345541 in male rats. Data suggest an important regulatory role of ERα in the IKKs/NF-kB activity on establishing mechanical allodynia induced by REMSD in female rats.
Collapse
Affiliation(s)
| | - Janet Murbartián
- Pharmacobiology Department, Cinvestav, South Campus, Mexico City, Mexico.
| |
Collapse
|
6
|
Rodríguez-Palma EJ, Huerta de la Cruz S, Islas-Espinoza AM, Castañeda-Corral G, Granados-Soto V, Khanna R. Nociplastic pain mechanisms and toll-like receptors as promising targets for its management. Pain 2024; 165:2150-2164. [PMID: 38595206 DOI: 10.1097/j.pain.0000000000003238] [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: 11/06/2023] [Accepted: 02/29/2024] [Indexed: 04/11/2024]
Abstract
ABSTRACT Nociplastic pain, characterized by abnormal pain processing without an identifiable organic cause, affects a significant portion of the global population. Unfortunately, current pharmacological treatments for this condition often prove ineffective, prompting the need to explore new potential targets for inducing analgesic effects in patients with nociplastic pain. In this context, toll-like receptors (TLRs), known for their role in the immune response to infections, represent promising opportunities for pharmacological intervention because they play a relevant role in both the development and maintenance of pain. Although TLRs have been extensively studied in neuropathic and inflammatory pain, their specific contributions to nociplastic pain remain less clear, demanding further investigation. This review consolidates current evidence on the connection between TLRs and nociplastic pain, with a specific focus on prevalent conditions like fibromyalgia, stress-induced pain, sleep deprivation-related pain, and irritable bowel syndrome. In addition, we explore the association between nociplastic pain and psychiatric comorbidities, proposing that modulating TLRs can potentially alleviate both pain syndromes and related psychiatric disorders. Finally, we discuss the potential sex differences in TLR signaling, considering the higher prevalence of nociplastic pain among women. Altogether, this review aims to shed light on nociplastic pain, its underlying mechanisms, and its intriguing relationship with TLR signaling pathways, ultimately framing the potential therapeutic role of TLRs in addressing this challenging condition.
Collapse
Affiliation(s)
- Erick J Rodríguez-Palma
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, United States
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | | | - Ana M Islas-Espinoza
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | | | - Vinicio Granados-Soto
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Rajesh Khanna
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
7
|
Chen S, Xie Y, Liang Z, Lu Y, Wang J, Xing F, Mao Y, Wei X, Wang Z, Yang J, Yuan J. A Narrative Review of the Reciprocal Relationship Between Sleep Deprivation and Chronic Pain: The Role of Oxidative Stress. J Pain Res 2024; 17:1785-1792. [PMID: 38799272 PMCID: PMC11122178 DOI: 10.2147/jpr.s455621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 05/10/2024] [Indexed: 05/29/2024] Open
Abstract
Sleep is crucial for human health, insufficient sleep or poor sleep quality may negatively affect sleep function and lead to a state of sleep deprivation. Sleep deprivation can result in various health problems, including chronic pain. The intricate relationship between sleep and pain is complex and intertwined, with daytime pain affecting sleep quality and poor sleep increasing pain intensity. The article first describes the influence of sleep on the onset and development of pain, and then explores the impact of daytime pain intensity on nighttime sleep quality and subsequent pain thresholds. However, the primary emphasis is placed on the pivotal role of oxidative stress in this bidirectional relationship. Although the exact mechanisms underlying sleep and chronic pain are unclear, this review focuses on the role of oxidative stress. Numerous studies on sleep deprivation have demonstrated that it can lead to varying degrees of increased pain sensitivity, while chronic pain leads to sleep deprivation and further exacerbates pain. Further research on the role of oxidative stress in the mechanism of sleep deprivation-induced pain sensitization seems reasonable. This article comprehensively reviews the current research on the interrelationship between sleep deprivation, pain and the crucial role of oxidative stress.
Collapse
Affiliation(s)
- Shuhan Chen
- Department of Anesthesiology, Pain and Perioperative Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Yanle Xie
- Department of Anesthesiology, Pain and Perioperative Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Zenghui Liang
- Department of Anesthesiology, Pain and Perioperative Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Yu Lu
- Department of Orthopedics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Jingping Wang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Fei Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Yuanyuan Mao
- Department of Anesthesiology, Pain and Perioperative Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Xin Wei
- Department of Anesthesiology, Pain and Perioperative Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Zhongyu Wang
- Department of Anesthesiology, Pain and Perioperative Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Jianjun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| | - Jingjing Yuan
- Department of Anesthesiology, Pain and Perioperative Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
- Henan Province International Joint Laboratory of Pain, Cognition and Emotion, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People’s Republic of China
| |
Collapse
|
8
|
Nguyen T, Nguyen N, Cochran AG, Smith JA, Al-Juboori M, Brumett A, Saxena S, Talley S, Campbell EM, Obukhov AG, White FA. Repeated closed-head mild traumatic brain injury-induced inflammation is associated with nociceptive sensitization. J Neuroinflammation 2023; 20:196. [PMID: 37635235 PMCID: PMC10464478 DOI: 10.1186/s12974-023-02871-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 08/03/2023] [Indexed: 08/29/2023] Open
Abstract
BACKGROUND Individuals who have experienced mild traumatic brain injuries (mTBIs) suffer from several comorbidities, including chronic pain. Despite extensive studies investigating the underlying mechanisms of mTBI-associated chronic pain, the role of inflammation in long-term pain after mTBIs is not fully elucidated. Given the shifting dynamics of inflammation, it is important to understand the spatial-longitudinal changes in inflammatory processes following mTBIs and their effects on TBI-related pain. METHODS We utilized a recently developed transgenic caspase-1 luciferase reporter mouse model to monitor caspase-1 activation through a thinned skull window in the in vivo setting following three closed-head mTBI events. Organotypic coronal brain slice cultures and acutely dissociated dorsal root ganglion (DRG) cells provided tissue-relevant context of inflammation signal. Mechanical allodynia was assessed by mechanical withdrawal threshold to von Frey and thermal hyperalgesia withdrawal latency to radiant heat. Mouse grimace scale (MGS) was used to detect spontaneous or non-evoked pain. In some experiments, mice were prophylactically treated with MCC950, a potent small molecule inhibitor of NLRP3 inflammasome assembly to inhibit injury-induced inflammatory signaling. Bioluminescence spatiotemporal dynamics were quantified in the head and hind paws, and caspase-1 activation was confirmed by immunoblot. Immunofluorescence staining was used to monitor the progression of astrogliosis and microglial activation in ex vivo brain tissue following repetitive closed-head mTBIs. RESULTS Mice with repetitive closed-head mTBIs exhibited significant increases of the bioluminescence signals within the brain and paws in vivo for at least one week after each injury. Consistently, immunoblotting and immunofluorescence experiments confirmed that mTBIs led to caspase-1 activation, astrogliosis, and microgliosis. Persistent changes in MGS and hind paw withdrawal thresholds, indicative of pain states, were observed post-injury in the same mTBI animals in vivo. We also observed enhanced inflammatory responses in ex vivo brain slice preparations and DRG for at least 3 days following mTBIs. In vivo treatment with MCC950 significantly reduced caspase-1 activation-associated bioluminescent signals in vivo and decreased stimulus-evoked and non-stimulus evoked nociception. CONCLUSIONS Our findings suggest that the inflammatory states in the brain and peripheral nervous system following repeated mTBIs are coincidental with the development of nociceptive sensitization, and that these events can be significantly reduced by inhibition of NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Tyler Nguyen
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Natalie Nguyen
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ashlyn G Cochran
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jared A Smith
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mohammed Al-Juboori
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew Brumett
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Saahil Saxena
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sarah Talley
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL, USA
| | - Edward M Campbell
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL, USA
- Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Alexander G Obukhov
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy, Cellular Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fletcher A White
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA.
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
9
|
Zhu M, Huang H. The Underlying Mechanisms of Sleep Deprivation Exacerbating Neuropathic Pain. Nat Sci Sleep 2023; 15:579-591. [PMID: 37533626 PMCID: PMC10392808 DOI: 10.2147/nss.s414174] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023] Open
Abstract
Pain disrupts sleep, and sleep deprivation or interference can alter pain perception in animals and humans, for example by increasing sensitivity to pain. However, the mechanism by which sleep affects neuropathic pain remains unclear. In this review, we discuss the available evidence from the epidemiologic, clinical, and human, as well as laboratory studies. In previous studies, we have found that sleep deprivation affects various injurious systems, including opioids, dopaminergic, immune, orexins, hypothalamic-pituitary-adrenal axis, and adenosine. At the same time, these systems play a crucial role in neuropathic pain regulation. In the complex interactions between these neurobiological systems, there may be potential regulatory pathways through which sleep deprivation amplifies neuropathic pain. Because of the impact sleep problems and neuropathic pain can have on the patients' quality of life, studying the link between sleep and neuropathic pain is important for neuropathic pain prevention and public health.
Collapse
Affiliation(s)
- Manmin Zhu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Guizhou, People’s Republic of China
| | - Hao Huang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Guizhou, People’s Republic of China
| |
Collapse
|
10
|
Li YJ, He XL, Zhang JY, Liu XJ, Liang JL, Zhou Q, Zhou GH. 8-O-acetyl shanzhiside methylester protects against sleep deprivation-induced cognitive deficits and anxiety-like behaviors by regulating NLRP3 and Nrf2 pathways in mice. Metab Brain Dis 2023; 38:641-655. [PMID: 36456714 DOI: 10.1007/s11011-022-01132-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022]
Abstract
Sleep deprivation (SD) is prevalent throughout the world, which has negative effects on cognitive abilities, and causing mood alterations. 8-O-acetyl shanzhiside methylester (8-OaS), a chief component in Lamiophlomis rotata (L. rotata) Kudo, possesses potent neuroprotective properties and analgesic effects. Here, we evaluated the alleviative effects of 8-OaS on memory impairment and anxiety in mice subjected to SD (for 72-h). Our results demonstrated that 8-OaS (0.2, 2, 20 mg/kg) administration dose-dependently ameliorated behavioral abnormalities in SD mice, accompanied with restored synaptic plasticity and reduced shrinkage and loss of hippocampal neurons. 8-OaS reduced the inflammatory response and oxidative stress injury in hippocampus caused by SD, which may be related to inhibition of NLRP3 inflammasome-mediated inflammatory process and activation of the Nrf2/HO-1 pathway. SD also led to increases in the expressions of TLR-4/MyD88, active NF-κB, pro-IL-1β, TNFα and MDA, as well as a decrease in the level of SOD in mice hippocampus, which were reversed by 8-OaS administration. Moreover, our molecular docking analyses showed that 8-OaS also has good affinity for NLRP3 and Nrf2 signaling pathways. These results suggested that 8-OaS could be used as a novel herbal medicine for the treatment of sleep loss and for use as a structural base for developing new drugs.
Collapse
Affiliation(s)
- Yu-Jiao Li
- Department of Clinical Pharmacy, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu Province, China
| | - Xiao-Lu He
- Department of Clinical Pharmacy, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu Province, China
| | - Jie-Yu Zhang
- Department of Clinical Pharmacy, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu Province, China
| | - Xue-Jiao Liu
- Department of Clinical Pharmacy, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu Province, China
| | - Jia-Long Liang
- No.946 Hospital of PLA land Force, Yining, 835000, Xinjiang Uygur Autonomous Regions, China.
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Air Force Medical University, Xi'an, 710032, Shaanxi Province, China.
| | - Qing Zhou
- Department of Clinical Pharmacy, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu Province, China.
| | - Guo-Hua Zhou
- Department of Clinical Pharmacy, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu Province, China
| |
Collapse
|
11
|
Kourbanova K, Alexandre C, Latremoliere A. Effect of sleep loss on pain-New conceptual and mechanistic avenues. Front Neurosci 2022; 16:1009902. [PMID: 36605555 PMCID: PMC9807925 DOI: 10.3389/fnins.2022.1009902] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction Sleep disturbances increase pain sensitivity in clinical and preclinical settings, but the precise mechanisms are unknown. This represents a major public health issue because of the growing sleep deficiency epidemic fueled by modern lifestyle. To understand the neural pathways at the intersection between sleep and pain processes, it is critical to determine the precise nature of the sleep disruptions that increase pain and the specific component of the pain response that is targeted. Methods We performed a review of the literature about sleep disturbances and pain sensitivity in humans and rodents by taking into consideration the targeted sleep stage (REMS, non-NREMS, or both), the amount of sleep lost, and the different types of sleep disruptions (partial or total sleep loss, duration, sleep fragmentation or interruptions), and how these differences might affect distinct components of the pain response. Results We find that the effects of sleep disturbances on pain are highly conserved among species. The major driver for pain hypersensitivity appears to be the total amount of sleep lost, while REMS loss by itself does not seem to have a direct effect on pain sensitivity. Sleep loss caused by extended wakefulness preferentially increases pain perception, whereas interrupted and limited sleep strongly dysregulates descending controls such as DNIC, especially in women. Discussion We discuss the possible mechanisms involved, including an increase in inflammatory processes, a loss of nociceptive inhibitory pathways, and a defect in the cognitive processing of noxious input.
Collapse
Affiliation(s)
- Kamila Kourbanova
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Chloe Alexandre
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Alban Latremoliere
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, United States
| |
Collapse
|