1
|
Brengel EK, Axe B, Maheswari A, Abeer MI, Ortiz RJ, Woodward TJ, Walhof R, Utama R, Sawada C, Balaji S, Kulkarni PP, Bradshaw HB, Gitcho MA, Ferris CF. Psilocybin as a Treatment for Repetitive Mild Head Injury: Evidence from Neuroradiology and Molecular Biology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636248. [PMID: 39975204 PMCID: PMC11838531 DOI: 10.1101/2025.02.03.636248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Repetitive mild head injuries incurred while playing organized sports, during car accidents and falls, or in active military service are a major health problem. These head injuries induce cognitive, motor, and behavioral deficits that can last for months and even years with an increased risk of dementia, Parkinson's disease, and chronic traumatic encephalopathy. There is no approved medical treatment for these types of head injuries. To this end, we tested the healing effects of the psychedelic psilocybin, as it is known to reduce neuroinflammation and enhance neuroplasticity. Using a model of mild repetitive head injury in adult female rats, we provide unprecedented data that psilocybin can reduce vasogenic edema, restore normal vascular reactivity and functional connectivity, reduce phosphorylated tau buildup, enhance levels of brain-derived neurotrophic factor and its receptor TrkB, and modulate lipid signaling molecules.
Collapse
Affiliation(s)
- Eric K Brengel
- Dept Psychology, Northeastern Univ, Boston, MA, USA
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA
| | - Bryce Axe
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA
| | - Ashwath Maheswari
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA
| | | | | | - Taylor J Woodward
- Psychological & Brain Sciences, Program in Neuroscience, Indiana Univ, Bloomington, IN
| | - Reagan Walhof
- Psychological & Brain Sciences, Program in Neuroscience, Indiana Univ, Bloomington, IN
| | - Rachel Utama
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA
| | - Courtney Sawada
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA
| | - Shreyas Balaji
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA
| | - Praveen P Kulkarni
- Dept Psychology, Northeastern Univ, Boston, MA, USA
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA
| | - Heather B Bradshaw
- Psychological & Brain Sciences, Program in Neuroscience, Indiana Univ, Bloomington, IN
| | | | - Craig F Ferris
- Dept Psychology, Northeastern Univ, Boston, MA, USA
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA
- Dept Pharmaceutical Sciences, Northeastern Univ, Boston, MA
| |
Collapse
|
2
|
Tian L, Li X, Zhao Y, Yi H, Liu X, Yao R, Hou X, Zhu X, Huo F, Chen T, Liang L. DNMT3a Downregulation Ttriggered Upregulation of GABA A Receptor in the mPFC Promotes Paclitaxel-Induced Pain and Anxiety in Male Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407387. [PMID: 39679872 PMCID: PMC11791956 DOI: 10.1002/advs.202407387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/25/2024] [Indexed: 12/17/2024]
Abstract
Chemotherapeutic agents, such as paclitaxel (PTX), induce neuroplastic changes and alter gene expression in the prefrontal cortex (PFC), which may be associated with chemotherapy-induced pain and negative emotions. Notably, DNA methylation undergoes adaptive changes in neurological disorders, emerging as a promising target for neuromodulation. In this study, systemic administration of PTX leads to a decrease in the expression of the DNA methyltransferase DNMT3a, while concurrently upregulating the expression of Gabrb1 mRNA and its encoded GABAARβ1 protein in the medial PFC (mPFC) of male mice. Overexpression of DNMT3a in the mPFC alleviates PTX-induced pain hypersensitivity, and anxiety-like behavior in these mice. Additionally, it reverses the PTX-induced increase in inhibitory synaptic transmission in the pyramidal neurons of the mPFC. Mechanistically, the upregulation of GABAARβ1 in the mPFC is linked to the reduced expression of DNMT3a and DNA hypomethylation at the promoter region of the Gabrb1 gene. Furthermore, a long-term diet rich in methyl donors alleviates PTX-induced pain hypersensitivity and anxiety-like behavior in mice. These findings suggest that the DNMT3a-mediated upregulation of GABAARβ1 in the mPFC contributes to PTX-induced neuropathic pain and anxiety, highlighting DNA methylation-dependent epigenetic regulation as a potential therapeutic target for addressing chemotherapy-induced cortical dysfunction.
Collapse
Affiliation(s)
- Lixia Tian
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesInstitute of NeuroscienceTranslational Medicine InstituteXi'an Jiaotong University Health Science CenterXi'anShaanxi710061P. R. China
| | - Xu‐Hui Li
- Center for Neuron and DiseaseFrontier Institutes of Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710061P. R. China
| | - Yu‐Long Zhao
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesInstitute of NeuroscienceTranslational Medicine InstituteXi'an Jiaotong University Health Science CenterXi'anShaanxi710061P. R. China
| | - Hui‐Yuan Yi
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesInstitute of NeuroscienceTranslational Medicine InstituteXi'an Jiaotong University Health Science CenterXi'anShaanxi710061P. R. China
| | - Xue‐Ru Liu
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesInstitute of NeuroscienceTranslational Medicine InstituteXi'an Jiaotong University Health Science CenterXi'anShaanxi710061P. R. China
| | - Rongrong Yao
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesInstitute of NeuroscienceTranslational Medicine InstituteXi'an Jiaotong University Health Science CenterXi'anShaanxi710061P. R. China
| | - Xue‐Mei Hou
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesInstitute of NeuroscienceTranslational Medicine InstituteXi'an Jiaotong University Health Science CenterXi'anShaanxi710061P. R. China
| | - Xuan Zhu
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesInstitute of NeuroscienceTranslational Medicine InstituteXi'an Jiaotong University Health Science CenterXi'anShaanxi710061P. R. China
| | - Fu‐Quan Huo
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesInstitute of NeuroscienceTranslational Medicine InstituteXi'an Jiaotong University Health Science CenterXi'anShaanxi710061P. R. China
| | - Tao Chen
- Department of Anatomy and K.K. Leung Brain Research CentreFourth Military Medical UniversityXi'an710032P. R. China
| | - Lingli Liang
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesInstitute of NeuroscienceTranslational Medicine InstituteXi'an Jiaotong University Health Science CenterXi'anShaanxi710061P. R. China
| |
Collapse
|
3
|
van de Wetering R, Vu LY, Kornberger LD, Luo D, Scouller B, Hong S, Paton K, Prisinzano TE, Kivell BM. Effects of Biased Analogues of the Kappa Opioid Receptor Agonist, U50,488, in Preclinical Models of Pain and Side Effects. Molecules 2025; 30:604. [PMID: 39942708 PMCID: PMC11820436 DOI: 10.3390/molecules30030604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Kappa opioid receptor (KOR) agonists have well-established antinociceptive effects. However, many KOR agonists have negative side effects, which limit their therapeutic potential. Some researchers have suggested that the development of biased agonists that preferentially stimulate KOR G-protein pathways over β-arrestin pathways may yield drugs with fewer adverse side effects. This was investigated in the current study. We describe the synthesis and characterization of three U50,488 analogues, 1, 2, and 3. We evaluated the acute and chronic antinociceptive effects of these compounds in mice using the warm-water tail flick assay and in a paclitaxel-induced neuropathic pain model. Side effects were investigated using open-field, passive wire hang, rotarod, elevated zero maze, conditioned place aversion, and whole-body plethysmography, with some tests being conducted in KOR or β-arrestin2 knock out mice. All compounds were highly potent, full agonists of the KOR, with varying signaling biases in vitro. In the warm-water tail withdrawal assay, these agonists were ~10 times more potent than U50,488, but not more efficacious. All KOR agonists reversed paclitaxel-induced neuropathic pain, without tolerance. Compound 3 showed no significant side effects on any test. Signaling bias did not correlate with the antinociceptive or side effects of any compounds and knockout of β-arrestin2 had no effect on U50,488-induced sedation or motor incoordination. These findings highlight the therapeutic potential of 3, with its lack of side effects typically associated with KOR agonists, and also suggest that G-protein signaling bias is a poor predictor of KOR agonist-induced side effects.
Collapse
MESH Headings
- Animals
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/metabolism
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology
- Mice
- Disease Models, Animal
- Male
- Mice, Knockout
- Neuralgia/drug therapy
- Neuralgia/chemically induced
- Paclitaxel/adverse effects
- Paclitaxel/pharmacology
- Analgesics/pharmacology
- Analgesics/chemistry
- Humans
- Analgesics, Opioid/pharmacology
- Analgesics, Opioid/adverse effects
Collapse
Affiliation(s)
- Ross van de Wetering
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand (B.S.)
| | - Loan Y. Vu
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA; (L.Y.V.); (L.D.K.); (D.L.)
| | - Lindsay D. Kornberger
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA; (L.Y.V.); (L.D.K.); (D.L.)
| | - Dan Luo
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA; (L.Y.V.); (L.D.K.); (D.L.)
| | - Brittany Scouller
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand (B.S.)
| | - Sheein Hong
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand (B.S.)
| | - Kelly Paton
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand (B.S.)
| | - Thomas E. Prisinzano
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA; (L.Y.V.); (L.D.K.); (D.L.)
| | - Bronwyn M. Kivell
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand (B.S.)
| |
Collapse
|
4
|
Pondelick AM, Moncayo LV, Donvito G, McLane VD, Gillespie JC, Hauser KF, Spiegel S, Lichtman AH, Sim-Selley LJ, Selley DE. Dissociation between the anti-allodynic effects of fingolimod (FTY720) and desensitization of S1P 1 receptor-mediated G-protein activation in a mouse model of sciatic nerve injury. Neuropharmacology 2024; 261:110165. [PMID: 39303855 DOI: 10.1016/j.neuropharm.2024.110165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/28/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Sphingosine-1-phosphate (S1P) receptor (S1PR) agonists, such as fingolimod (FTY720), alleviate nociception in preclinical pain models by either activation (agonism) or inhibition (functional antagonism) of S1PR type-1 (S1PR1). However, the dose-dependence and temporal relationship between reversal of nociception and modulation of S1PR1 signaling has not been systematically investigated. This study examined the relationship between FTY720-induced antinociception and S1PR1 adaptation using a sciatic nerve chronic constriction injury (CCI) model of neuropathic pain in male and female C57Bl/6J mice. Daily injections of FTY720 for 14 days dose-dependently reversed CCI-induced mechanical allodynia without tolerance development, and concomitantly resulted in a dose-dependent reduction of G-protein activation by the S1PR1-selective agonist SEW2871 in the lumbar spinal cord and brain. These findings indicate FTY720-induced desensitization of S1PR1 signaling coincides with its anti-allodynic effects. Consistent with this finding, a single injection of FTY720 reversed mechanical allodynia while concomitantly producing partial desensitization of S1PR1-stimulated G-protein activation in the CNS. However, mechanical allodynia returned 24-hr post injection, despite S1PR1 desensitization at that time, demonstrating a dissociation between these measures. Furthermore, CCI surgery led to elevations of sphingolipid metabolites, including S1P, which were unaffected by daily FTY720 administration, suggesting FTY720 reversed mechanical allodynia by targeting S1PR1 rather than sphingolipid metabolism. Supporting this hypothesis, acute administration of the S1PR1-selective agonist CYM-5442 mimicked the anti-allodynic effect of FTY720. In contrast, the S1PR1-selective antagonist NIBR-0213 prevented the anti-allodynic effect of FTY720, but NIBR-0213 given alone did not affect nociception. These results indicate that FTY720 alleviates CCI-induced allodynia through a mechanism distinct from functional antagonism.
Collapse
Affiliation(s)
- Abby M Pondelick
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Lauren V Moncayo
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Giulia Donvito
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Virginia D McLane
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - James C Gillespie
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Aron H Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Laura J Sim-Selley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Dana E Selley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| |
Collapse
|
5
|
Liu Z, Liu S, Zhao Y, Wang Q. Biological Mediators and Partial Regulatory Mechanisms on Neuropathic Pain Associated With Chemotherapeutic Agents. Physiol Res 2024; 73:333-341. [PMID: 39027951 PMCID: PMC11299781 DOI: 10.33549/physiolres.935162] [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: 06/15/2023] [Accepted: 09/13/2023] [Indexed: 07/27/2024] Open
Abstract
One of the most common issues caused by antineoplastic agents is chemotherapy-induced peripheral neuropathy (CIPN). In patients, CIPN is a sensory neuropathy accompanied by various motor and autonomic changes. With a high prevalence of cancer patients, CIPN is becoming a major problem for both cancer patients and for their health care providers. Nonetheless, there are lacking effective interventions preventing CIPN and treating the CIPN symptoms. A number of studies have demonstrated the cellular and molecular signaling pathways leading to CIPN using experimental models and the beneficial effects of some interventions on the CIPN symptoms related to those potential mechanisms. This review will summarize results obtained from recent human and animal studies, which include the abnormalities in mechanical and temperature sensory responses following chemotherapy such as representative bortezomib, oxaliplatin and paclitaxel. The underlying mechanisms of CIPN at cellular and molecular levels will be also discussed for additional in-depth studies needed to be better explored. Overall, this paper reviews the basic picture of CIPN and the signaling mechanisms of the most common antineoplastic agents in the peripheral and central nerve systems. A better understanding of the risk factors and fundamental mechanisms of CIPN is needed to develop effective preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Z Liu
- Tumor Center, The First Hospital of Jilin University, Changchun, Jilin, China.
| | | | | | | |
Collapse
|
6
|
Cunha M, Tavares I, Costa-Pereira JT. Centralizing the Knowledge and Interpretation of Pain in Chemotherapy-Induced Peripheral Neuropathy: A Paradigm Shift towards Brain-Centric Approaches. Brain Sci 2024; 14:659. [PMID: 39061400 PMCID: PMC11274822 DOI: 10.3390/brainsci14070659] [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: 05/14/2024] [Revised: 06/17/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a side effect of cancer treatment, often linked with pain complaints. Patients report mechanical and thermal hypersensitivity that may emerge during chemotherapy treatment and may persist after cancer remission. Whereas the latter situation disturbs the quality of life, life itself may be endangered by the appearance of CIPN during cancer treatment. The causes of CIPN have almost entirely been ascribed to the neurotoxicity of chemotherapeutic drugs in the peripheral nervous system. However, the central consequences of peripheral neuropathy are starting to be unraveled, namely in the supraspinal pain modulatory system. Based on our interests and experience in the field, we undertook a review of the brain-centered alterations that may underpin pain in CIPN. The changes in the descending pain modulation in CIPN models along with the functional and connectivity abnormalities in the brain of CIPN patients are analyzed. A translational analysis of preclinical findings about descending pain regulation during CIPN is reviewed considering the main neurochemical systems (serotoninergic and noradrenergic) targeted in CIPN management in patients, namely by antidepressants. In conclusion, this review highlights the importance of studying supraspinal areas involved in descending pain modulation to understand the pathophysiology of CIPN, which will probably allow a more personalized and effective CIPN treatment in the future.
Collapse
Affiliation(s)
- Mário Cunha
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (M.C.); (J.T.C.-P.)
| | - Isaura Tavares
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (M.C.); (J.T.C.-P.)
- I3S—Institute of Investigation and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - José Tiago Costa-Pereira
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (M.C.); (J.T.C.-P.)
- I3S—Institute of Investigation and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Faculty of Nutrition and Food Sciences, University of Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal
| |
Collapse
|
7
|
Chen X, Gan Y, Au NPB, Ma CHE. Current understanding of the molecular mechanisms of chemotherapy-induced peripheral neuropathy. Front Mol Neurosci 2024; 17:1345811. [PMID: 38660386 PMCID: PMC11039947 DOI: 10.3389/fnmol.2024.1345811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is the most common off-target adverse effects caused by various chemotherapeutic agents, such as cisplatin, oxaliplatin, paclitaxel, vincristine and bortezomib. CIPN is characterized by a substantial loss of primary afferent sensory axonal fibers leading to sensory disturbances in patients. An estimated of 19-85% of patients developed CIPN during the course of chemotherapy. The lack of preventive measures and limited treatment options often require a dose reduction or even early termination of life-saving chemotherapy, impacting treatment efficacy and patient survival. In this Review, we summarized the current understanding on the pathogenesis of CIPN. One prominent change induced by chemotherapeutic agents involves the disruption of neuronal cytoskeletal architecture and axonal transport dynamics largely influenced by the interference of microtubule stability in peripheral neurons. Due to an ineffective blood-nerve barrier in our peripheral nervous system, exposure to some chemotherapeutic agents causes mitochondrial swelling in peripheral nerves, which lead to the opening of mitochondrial permeability transition pore and cytochrome c release resulting in degeneration of primary afferent sensory fibers. The exacerbated nociceptive signaling and pain transmission in CIPN patients is often linked the increased neuronal excitability largely due to the elevated expression of various ion channels in the dorsal root ganglion neurons. Another important contributing factor of CIPN is the neuroinflammation caused by an increased infiltration of immune cells and production of inflammatory cytokines. In the central nervous system, chemotherapeutic agents also induce neuronal hyperexcitability in the spinal dorsal horn and anterior cingulate cortex leading to the development of central sensitization that causes CIPN. Emerging evidence suggests that the change in the composition and diversity of gut microbiota (dysbiosis) could have direct impact on the development and progression of CIPN. Collectively, all these aspects contribute to the pathogenesis of CIPN. Recent advances in RNA-sequencing offer solid platform for in silico drug screening which enable the identification of novel therapeutic agents or repurpose existing drugs to alleviate CIPN, holding immense promises for enhancing the quality of life for cancer patients who undergo chemotherapy and improve their overall treatment outcomes.
Collapse
Affiliation(s)
- Xinyu Chen
- Department of Neuroscience, Hong Kong Special Administrative Region (HKSAR), City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Yumeng Gan
- Department of Neuroscience, Hong Kong Special Administrative Region (HKSAR), City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Ngan Pan Bennett Au
- Department of Neuroscience, Hong Kong Special Administrative Region (HKSAR), City University of Hong Kong, Kowloon, Hong Kong SAR, China
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
- Institute of Life Sciences and Healthcare, University of Portsmouth, Portsmouth, United Kingdom
| | - Chi Him Eddie Ma
- Department of Neuroscience, Hong Kong Special Administrative Region (HKSAR), City University of Hong Kong, Kowloon, Hong Kong SAR, China
| |
Collapse
|
8
|
Sun S, Xu J, Lin L, Jia M, Xue X, Wang Q, Chen D, Huang Z, Wang Y. Chemotherapeutic drug elemene induces pain and anxiety-like behaviors by activating GABAergic neurons in the lateral septum of mice. Biochem Biophys Res Commun 2024; 699:149548. [PMID: 38281329 DOI: 10.1016/j.bbrc.2024.149548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/30/2023] [Accepted: 01/17/2024] [Indexed: 01/30/2024]
Abstract
Most chemotherapeutic drugs are potent and have a very narrow range of dose safety and efficacy, most of which can cause many side effects. Chemotherapy-induced peripheral neuropathy (CIPN) is the most common and serious side effect of chemotherapy for cancer treatment. However, its mechanism of action is yet to be fully elucidated. In the present study, we found that the treatment of the chemotherapy drug elemene induced hyperalgesia accompanied by anxiety-like emotions in mice based on several pain behavioral assays, such as mechanical allodynia and thermal hyperalgesia tests. Second, immunostaining for c-fos (a marker of activated neurons) further showed that elemene treatment activated several brain regions, including the lateral septum (LS), cingulate cortex (ACC), paraventricular nucleus of the thalamus (PVT), and dorsomedial hypothalamic nucleus (DMH), most notably in the GABAergic neurons of the lateral septum (LS). Finally, we found that both chemogenetic inhibition and apoptosis of LS neurons significantly reduced pain- and anxiety-like behaviors in mice treated with elemene. Taken together, these findings suggest that LS is involved in the regulation of elemene-induced chemotherapy pain and anxiety-like behaviors, providing a new target for the treatment of chemotherapy pain induced by elemene.
Collapse
Affiliation(s)
- Shanshan Sun
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Jiayun Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Lin Lin
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Mengxian Jia
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiumin Xue
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Qian Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Danni Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Zhihui Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| | - Yongjie Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| |
Collapse
|
9
|
Reddy P, Vasudeva J, Shah D, Prajapati JN, Harikumar N, Barik A. A Deep-Learning Driven Investigation of the Circuit Basis for Reflexive Hypersensitivity to Thermal Pain. Neuroscience 2023; 530:158-172. [PMID: 37640138 DOI: 10.1016/j.neuroscience.2023.08.023] [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: 02/22/2023] [Revised: 08/14/2023] [Accepted: 08/15/2023] [Indexed: 08/31/2023]
Abstract
Objectively measuring animal behavior is vital to understanding the neural circuits underlying pain. Recent progress in machine vision has presented unprecedented scope in behavioral analysis. Here, we apply DeepLabCut (DLC) to dissect mouse behavior on the thermal-plate test - a commonly used paradigm to ascertain supraspinal contributions to noxious thermal sensation and pain hypersensitivity. We determine the signature characteristics of the pattern of mouse movement and posture in 3D in response to a range of temperatures from innocuous to noxious on the thermal-plate test. Next, we test how acute chemical and chronic inflammatory injuries sensitize mouse behaviors. Repeated exposure to noxious temperatures on the thermal plate can induce learning. In this study, we design a novel assay and formulate an analytical pipeline to facilitate the dissection of plasticity mechanisms in pain circuits in the brain. Last, we record and test how activating Tacr1 expressing PBN neurons (PBNTacr1) - a population responsive to sustained noxious stimuli- affects mouse behavior on the thermal plate test. Taken together, we demonstrate that by tracking a single body part of a mouse, we can reveal the behavioral signatures of mice exposed to noxious surface temperatures, report the alterations of the same when injured, and determine if a molecularly and anatomically defined pain-responsive circuit plays a role in the reflexive hypersensitivity to thermal pain.
Collapse
Affiliation(s)
- Prannay Reddy
- Center for Neuroscience, Division of Biological Sciences, Indian Institute of Science, Gulmohar Marg, Bengaluru, Karnataka 560012, India
| | - Jayesh Vasudeva
- Center for Neuroscience, Division of Biological Sciences, Indian Institute of Science, Gulmohar Marg, Bengaluru, Karnataka 560012, India
| | - Devanshi Shah
- Center for Neuroscience, Division of Biological Sciences, Indian Institute of Science, Gulmohar Marg, Bengaluru, Karnataka 560012, India
| | - Jagat Narayan Prajapati
- Center for Neuroscience, Division of Biological Sciences, Indian Institute of Science, Gulmohar Marg, Bengaluru, Karnataka 560012, India
| | - Nikhila Harikumar
- Center for Neuroscience, Division of Biological Sciences, Indian Institute of Science, Gulmohar Marg, Bengaluru, Karnataka 560012, India
| | - Arnab Barik
- Center for Neuroscience, Division of Biological Sciences, Indian Institute of Science, Gulmohar Marg, Bengaluru, Karnataka 560012, India.
| |
Collapse
|
10
|
Kenkel WM, Ortiz RJ, Yee JR, Perkeybile AM, Kulkarni P, Carter CS, Cushing BS, Ferris CF. Neuroanatomical and functional consequences of oxytocin treatment at birth in prairie voles. Psychoneuroendocrinology 2023; 150:106025. [PMID: 36709631 PMCID: PMC10064488 DOI: 10.1016/j.psyneuen.2023.106025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 01/06/2023] [Accepted: 01/07/2023] [Indexed: 01/15/2023]
Abstract
Birth is a critical period for the developing brain, a time when surging hormone levels help prepare the fetal brain for the tremendous physiological changes it must accomplish upon entry into the 'extrauterine world'. A number of obstetrical conditions warrant manipulations of these hormones at the time of birth, but we know little of their possible consequences on the developing brain. One of the most notable birth signaling hormones is oxytocin, which is administered to roughly 50% of laboring women in the United States prior to / during delivery. Previously, we found evidence for behavioral, epigenetic, and neuroendocrine consequences in adult prairie vole offspring following maternal oxytocin treatment immediately prior to birth. Here, we examined the neurodevelopmental consequences in adult prairie vole offspring following maternal oxytocin treatment prior to birth. Control prairie voles and those exposed to 0.25 mg/kg oxytocin were scanned as adults using anatomical and functional MRI, with neuroanatomy and brain function analyzed as voxel-based morphometry and resting state functional connectivity, respectively. Overall, anatomical differences brought on by oxytocin treatment, while widespread, were generally small, while differences in functional connectivity, particularly among oxytocin-exposed males, were larger. Analyses of functional connectivity based in graph theory revealed that oxytocin-exposed males in particular showed markedly increased connectivity throughout the brain and across several parameters, including closeness and degree. These results are interpreted in the context of the organizational effects of oxytocin exposure in early life and these findings add to a growing literature on how the perinatal brain is sensitive to hormonal manipulations at birth.
Collapse
Affiliation(s)
- William M Kenkel
- Department of Psychological & Brain Sciences, University of Delaware, Newark, DE, USA; Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA, USA.
| | - Richard J Ortiz
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA, USA; Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, NM, USA; Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA
| | - Jason R Yee
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA, USA; Institute of Animal Welfare Science, University of Veterinary Medicine, Vienna, Austria
| | - Allison M Perkeybile
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA, USA; Department of Psychology, University of Virginia, Charlottesville, VA, USA
| | - Praveen Kulkarni
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA, USA
| | - C Sue Carter
- Department of Psychology, University of Virginia, Charlottesville, VA, USA
| | - Bruce S Cushing
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX, USA
| | - Craig F Ferris
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA, USA
| |
Collapse
|
11
|
Costa-Pereira JT, Oliveira R, Guadilla I, Guillén MJ, Tavares I, López-Larrubia P. Neuroimaging uncovers neuronal and metabolic changes in pain modulatory brain areas in a rat model of chemotherapy-induced neuropathy - MEMRI and ex vivo spectroscopy studies. Brain Res Bull 2023; 192:12-20. [PMID: 36328144 DOI: 10.1016/j.brainresbull.2022.10.018] [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: 07/20/2022] [Revised: 10/20/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
Abstract
Chemotherapy-induced neuropathy (CIN) is one of the most common complications of cancer treatment with sensory dysfunctions which frequently include pain. The mechanisms underlying pain during CIN are starting to be uncovered. Neuroimaging allows the identification of brain circuitry involved in pain processing and modulation and has recently been used to unravel the disruptions of that circuitry by neuropathic pain. The present study evaluates the effects of paclitaxel, a cytostatic drug frequently used in cancer treatment, at the neuronal function in the anterior cingulate cortex (ACC), hypothalamus and periaqueductal gray (PAG) using manganese-enhanced magnetic resonance imaging (MEMRI). We also studied the metabolic profile at the prefrontal cortex (PFC) and hypothalamus using ex vivo spectroscopy. Wistar male rats were intraperitoneal injected with paclitaxel or vehicle solution (DMSO). The evaluation of mechanical sensitivity using von Frey test at baseline (BL), 21 (T21), 28 (T28), 49 (T49) and 56 days (T56) after CIN induction showed that paclitaxel-injected rats presented mechanical hypersensitivity from T21 until T56 after CIN induction. The evaluation of the locomotor activity and exploratory behaviors using open-field test at T28 and T56 after the first injection of paclitaxel revealed that paclitaxel-injected rats walked higher distance with higher velocity at late point of CIN accompanied with a sustained exhibition of anxiety-like behaviors. Imaging studies performed using MEMRI at T28 and T56 showed that paclitaxel treatment increased the neuronal activation in the hypothalamus and PAG at T56 in comparison with the control group. The analysis of data from ex vivo spectroscopy demonstrated that at T28 paclitaxel-injected rats presented an increase of N-acetyl aspartate (NAA) levels in the PFC and an increase of NAA and decrease of lactate (Lac) concentration in the hypothalamus compared to the control group. Furthermore, at T56 the paclitaxel-injected rats presented lower NAA and higher taurine (Tau) levels in the PFC. Together, MEMRI and metabolomic data indicate that CIN is associated with neuroplastic changes in brain areas involved in pain modulation and suggests that other events involving glial cells may be happening.
Collapse
Affiliation(s)
- José Tiago Costa-Pereira
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; IBMC-Institute of Molecular and Cell Biology, University of Porto, Portugal; I3S, Institute of Investigation and Innovation in Health, University of Porto, Portugal; Faculty of Nutrition and Food Sciences, University of Porto, Portugal
| | - Rita Oliveira
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; IBMC-Institute of Molecular and Cell Biology, University of Porto, Portugal; I3S, Institute of Investigation and Innovation in Health, University of Porto, Portugal
| | - Irene Guadilla
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | - Maria Jose Guillén
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain
| | - Isaura Tavares
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; IBMC-Institute of Molecular and Cell Biology, University of Porto, Portugal; I3S, Institute of Investigation and Innovation in Health, University of Porto, Portugal
| | - Pilar López-Larrubia
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain.
| |
Collapse
|
12
|
Lin X, Xu Z, Carey L, Romero J, Makriyannis A, Hillard CJ, Ruggiero E, Dockum M, Houk G, Mackie K, Albrecht PJ, Rice FL, Hohmann AG. A peripheral CB2 cannabinoid receptor mechanism suppresses chemotherapy-induced peripheral neuropathy: evidence from a CB2 reporter mouse. Pain 2022; 163:834-851. [PMID: 35001054 PMCID: PMC8942871 DOI: 10.1097/j.pain.0000000000002502] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 01/10/2023]
Abstract
ABSTRACT CB2 cannabinoid receptors (CB2) are a promising therapeutic target that lacks unwanted side effects of CB1 activation. However, the cell types expressing CB2 that mediate these effects remain poorly understood. We used transgenic mice with CB2 promoter-driven expression of enhanced green fluorescent protein (EGFP) to study cell types that express CB2 and suppress neuropathic nociception in a mouse model of chemotherapy-induced peripheral neuropathy. Structurally distinct CB2 agonists (AM1710 and LY2828360) suppressed paclitaxel-induced mechanical and cold allodynia in CB2EGFP reporter mice with established neuropathy. Antiallodynic effects of AM1710 were blocked by SR144528, a CB2 antagonist with limited CNS penetration. Intraplantar AM1710 administration suppressed paclitaxel-induced neuropathic nociception in CB2EGFP but not CB2 knockout mice, consistent with a local site of antiallodynic action. mRNA expression levels of the anti-inflammatory cytokine interleukin-10 were elevated in the lumbar spinal cord after intraplantar AM1710 injection along with the proinflammatory cytokine tumor necrosis factor alpha and chemokine monocyte chemoattractant protein-1. CB2EGFP, but not wildtype mice, exhibited anti-GFP immunoreactivity in the spleen. However, the anti-GFP signal was below the threshold for detection in the spinal cord and brain of either vehicle-treated or paclitaxel-treated CB2EGFP mice. EGFP fluorescence was coexpressed with CB2 immunolabeling in stratified patterns among epidermal keratinocytes. EGFP fluorescence was also expressed in dendritic cells in the dermis, Langerhans cells in the epidermis, and Merkel cells. Quantification of the EGFP signal revealed that Langerhans cells were dynamically increased in the epidermis after paclitaxel treatment. Our studies implicate CB2 expressed in previously unrecognized populations of skin cells as a potential target for suppressing chemotherapy-induced neuropathic nociception.
Collapse
Affiliation(s)
- Xiaoyan Lin
- Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Zhili Xu
- Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Lawrence Carey
- Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
- Program in Neuroscience, Indiana University, Bloomington, IN, United States
| | - Julian Romero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, Madrid, Spain
| | - Alexandros Makriyannis
- School of Pharmacy, Bouvé College of Health Sciences, Center for Drug Discovery, College of Science, Health Sciences Entrepreneurs, Northeastern University, Boston, MA, United States
| | - Cecilia J. Hillard
- Department of Pharmacology and Toxicology, Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | | | - Marilyn Dockum
- Integrated Tissue Dynamics LLC, Rensselaer, NY, United States
| | - George Houk
- Integrated Tissue Dynamics LLC, Rensselaer, NY, United States
| | - Ken Mackie
- Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
- Program in Neuroscience, Indiana University, Bloomington, IN, United States
- Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States
| | | | - Frank L. Rice
- Integrated Tissue Dynamics LLC, Rensselaer, NY, United States
| | - Andrea G. Hohmann
- Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
- Program in Neuroscience, Indiana University, Bloomington, IN, United States
- Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States
| |
Collapse
|
13
|
Coleman JR, Madularu D, Ortiz RJ, Athanassiou M, Knudsen A, Alkislar I, Cai X, Kulkarni PP, Cushing BS, Ferris CF. Changes in brain structure and function following chronic exposure to inhaled vaporised cannabis during periadolescence in female and male mice: A multimodal MRI study. Addict Biol 2022; 27:e13169. [PMID: 35470553 DOI: 10.1111/adb.13169] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND AIMS Social norms and legality surrounding the use of medical and recreational cannabis are changing rapidly. The prevalence of cannabis use in adolescence is increasing. The aim of this study was to assess any sex-based neurobiological effects of chronically inhaled, vaporised cannabis on adolescent female and male mice. METHODS Female and male mice were exposed daily to vaporised cannabis (10.3% Δ-9-tetrahydrocannabinol [THC] and 0.05% cannabidiol [CBD]) or placebo from postnatal day 23 to day 51. Following cessation of treatment, mice were examined for changes in brain structure and function using noninvasive multimodal magnetic resonance imaging (MRI). Data from voxel-based morphometry, diffusion weighted imaging and rest state functional connectivity were registered to and analysed with a 3D mouse atlas with 139 brain areas. Following imaging, mice were tested for their preference for a novel object. RESULTS The effects were sexually dimorphic with females showing a unique distribution and inverse correlation between measures of fractional anisotropy and apparent diffusion coefficient localised to the forebrain and hindbrain. In contrast males displayed significant increased functional coupling with the thalamus, hypothalamus and brainstem reticular activating system as compared with controls. Cannabis males also presented with altered hippocampal coupling and deficits in cognitive function. CONCLUSION Chronic exposure to inhaled vaporised cannabis had significant effects on brain structure and function in early adulthood corroborating much of the literature. Females presented with changes in grey matter microarchitecture, while males showed altered functional connectivity in hippocampal circuitry and deficits in object recognition.
Collapse
Affiliation(s)
- James R. Coleman
- Center for Translational NeuroImaging Northeastern University Boston Massachusetts USA
| | - Dan Madularu
- Center for Translational NeuroImaging Northeastern University Boston Massachusetts USA
| | - Richard J. Ortiz
- Department of Biological Sciences University of Texas at El Paso El Paso Texas USA
| | - Maria Athanassiou
- Centre de recherche de l'Institut Universitaire en Santé Mentale de Montréal Montreal Québec Canada
| | - Alexa Knudsen
- Center for Translational NeuroImaging Northeastern University Boston Massachusetts USA
| | - Ilayda Alkislar
- Center for Translational NeuroImaging Northeastern University Boston Massachusetts USA
| | - Xuezhu Cai
- Center for Translational NeuroImaging Northeastern University Boston Massachusetts USA
| | - Praveen P. Kulkarni
- Center for Translational NeuroImaging Northeastern University Boston Massachusetts USA
| | - Bruce S. Cushing
- Department of Biological Sciences University of Texas at El Paso El Paso Texas USA
| | - Craig F. Ferris
- Center for Translational NeuroImaging Northeastern University Boston Massachusetts USA
| |
Collapse
|
14
|
Peña-Ortega F, Robles-Gómez ÁA, Xolalpa-Cueva L. Microtubules as Regulators of Neural Network Shape and Function: Focus on Excitability, Plasticity and Memory. Cells 2022; 11:cells11060923. [PMID: 35326374 PMCID: PMC8946818 DOI: 10.3390/cells11060923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/09/2022] [Accepted: 02/17/2022] [Indexed: 12/19/2022] Open
Abstract
Neuronal microtubules (MTs) are complex cytoskeletal protein arrays that undergo activity-dependent changes in their structure and function as a response to physiological demands throughout the lifespan of neurons. Many factors shape the allostatic dynamics of MTs and tubulin dimers in the cytosolic microenvironment, such as protein–protein interactions and activity-dependent shifts in these interactions that are responsible for their plastic capabilities. Recently, several findings have reinforced the role of MTs in behavioral and cognitive processes in normal and pathological conditions. In this review, we summarize the bidirectional relationships between MTs dynamics, neuronal processes, and brain and behavioral states. The outcomes of manipulating the dynamicity of MTs by genetic or pharmacological approaches on neuronal morphology, intrinsic and synaptic excitability, the state of the network, and behaviors are heterogeneous. We discuss the critical position of MTs as responders and adaptative elements of basic neuronal function whose impact on brain function is not fully understood, and we highlight the dilemma of artificially modulating MT dynamics for therapeutic purposes.
Collapse
|
15
|
Liu J, Li D, Huang J, Cao J, Cai G, Guo Y, Wang G, Zhao S, Wang X, Wu S. Glutamatergic Neurons in the Amygdala Are Involved in Paclitaxel-Induced Pain and Anxiety. Front Psychiatry 2022; 13:869544. [PMID: 35492735 PMCID: PMC9049739 DOI: 10.3389/fpsyt.2022.869544] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/21/2022] [Indexed: 11/18/2022] Open
Abstract
Paclitaxel is widely used as a first-line chemotherapy agent to treat malignant tumors. However, paclitaxel causes peripheral nerve fiber damage and neuropathic pain in some patients. In addition, patients received paclitaxel chemotherapy are often accompanied by negative emotions such as anxiety. The amygdala is critically involved in regulating pain signals, as well as anxiety. The purpose of this study is to clarify the role of Ca2+/calmodulin-dependent protein kinase II (CaMKII)-positive glutamatergic neurons in the amygdala in paclitaxel-induced pain and negative affective symptoms. Intraperitoneal injection of paclitaxel into mice caused mechanical and thermal allodynia, as measured by Von Frey test and Hargreaves test, and anxiety, as measured by open field test and elevated plus maze test. Immunofluorescence staining revealed that c-fos-positive neurons were significantly more in the basolateral amygdala (BLA) and central amygdala (CeA) in paclitaxel-treated mice than untreated mice. Furthermore, part of c-fos-positive neurons in the BLA were immunoreactive of CaMKII. Engineered Designer receptors exclusively activated by designer drugs (DREADD) receptor hM4Di or hM3Dq was selectively expressed on CaMKII neurons by injection of adeno-associated virus (AAV) vectors containing CaMKII and hM4Di or hM3Dq. Administration of DREADD agonist CNO to selectively inhibit the CaMKII neurons in the BLA significantly increased the paw withdrawal thresholds and paw withdrawal latencies. In addition, selectively inhibition of CaMKII neurons in the BLA alleviated anxiety behavior without affecting the motor activity. In summary, our findings suggest that CaMKII neurons in the amygdala are critical for neuropathic pain and anxiety behaviors induced by paclitaxel chemotherapy.
Collapse
Affiliation(s)
- Jiaxin Liu
- Department of Anesthesiology, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Dangchao Li
- Department of Anesthesiology, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jing Huang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Jing Cao
- Department of Anesthesiology, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guohong Cai
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yuexian Guo
- Department of Surgery, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guiying Wang
- Department of Surgery, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shuang Zhao
- Department of Anesthesiology, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiuli Wang
- Department of Anesthesiology, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
16
|
Whole brain in vivo neuropathology: Imaging site-specific changes in brain structure over time following trimethyltin exposure in rats. Toxicol Lett 2021; 352:54-60. [PMID: 34600096 DOI: 10.1016/j.toxlet.2021.09.009] [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: 04/22/2021] [Revised: 09/09/2021] [Accepted: 09/27/2021] [Indexed: 11/21/2022]
Abstract
Presented is a diffusion weighted imaging protocol with measures of apparent diffusion coefficient which when registered to a 3D MRI rat brain atlas provides site-specific information on 173 different brain areas. This protocol coined "in vivo neuropathology" was used to follow the progressive neurotoxic effects of trimethyltin on global gray matter microarchitecture. Four rats were given an IP injection of 7 mg/kg of the neurotoxin trimethyltin and imaged for changes in water diffusivity at 3- and 7-days post injections. At 3 days, there was a significant decrease in apparent diffusion coefficient, a proxy for cytotoxic edema, in several cortical areas and cerebellum. At 7 days the level of injury expanded to include most of the cerebral cortex, hippocampus, olfactory system, and cerebellum/brainstem corroborating much of the work done with traditional histopathology. Analysis is achieved with a minimum number of rats adhering to the laws and regulations around the humane care and use of laboratory animals, providing an alternative to the traditional tests for assessing drug neurotoxicity. "In vivo neuropathology" can minimize the cost, expedite the process, and identify subtle changes in site-specific brain microarchitecture across the entire brain.
Collapse
|
17
|
Vermeer CJC, Hiensch AE, Cleenewerk L, May AM, Eijkelkamp N. Neuro-immune interactions in paclitaxel-induced peripheral neuropathy. Acta Oncol 2021; 60:1369-1382. [PMID: 34313190 DOI: 10.1080/0284186x.2021.1954241] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Paclitaxel is a taxane-based chemotherapeutic agent used as a treatment in breast cancer. There is no effective prevention or treatment strategy for the most common side effect of peripheral neuropathy. In this manuscript, we reviewed the molecular mechanisms that contribute to paclitaxel-induced peripheral neuropathy (PIPN) with an emphasis on immune-related processes. METHODS A systematic search of the literature was conducted in PubMed, EMBASE and Cochrane Library. The SYRCLE's risk of bias tool was used to assess internal validity. RESULTS 156 studies conducted with rodent models were included. The risk of bias was high due to unclear methodology. Paclitaxel induces changes in myelinated axons, mitochondrial dysfunction, and mechanical hypersensitivity by affecting ion channels expression and function and facilitating spinal transmission. Paclitaxel-induced inflammatory responses are important contributors to PIPN. CONCLUSION Immune-related processes are an important mechanism contributing to PIPN. Studies in humans that validate these mechanistic data are highly needed to facilitate the development of therapeutic strategies.
Collapse
Affiliation(s)
- Cornelia J. C. Vermeer
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Anouk E. Hiensch
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Laurence Cleenewerk
- Center of Translational Immunology (CTI), University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Anne M. May
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Niels Eijkelkamp
- Center of Translational Immunology (CTI), University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
18
|
Chmielewski-Stivers N, Petit B, Ollivier J, Monceau V, Tsoutsou P, Quintela Pousa A, Lin X, Limoli C, Vozenin MC. Sex-Specific Differences in Toxicity Following Systemic Paclitaxel Treatment and Localized Cardiac Radiotherapy. Cancers (Basel) 2021; 13:cancers13163973. [PMID: 34439129 PMCID: PMC8394799 DOI: 10.3390/cancers13163973] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 01/10/2023] Open
Abstract
Simple Summary The objective of the present study was to investigate the impact of sex in the development of long-term toxicities affecting quality of life in cancer survivors after systemic paclitaxel treatment and cardiac irradiation. Sex-specific differences may affect tumor biology, drug pharmacokinetics and dynamics, and response to local treatment such as radiation therapy (RT). However, sex is rarely taken into consideration when administering cancer therapies. Interestingly, female mice are protected from paclitaxel-induced neurotoxicity as well as from radiotherapy-induced cardiotoxicity, and deficiency in the small GTPase RhoB reversed the protection in females but not in males. In conclusion, our results are the first to identify sex- and organ-specific responses to systemic paclitaxel administration and localized RT. These results may have important implications for the management of cancer patients and implementation of personalized medicine in oncology. Abstract The impact of sex in the development of long-term toxicities affecting the quality of life of cancer survivors has not been investigated experimentally. To address this issue, a series of neurologic and cardiologic endpoints were used to investigate sex-based differences triggered by paclitaxel treatment and radiotherapy exposure. Male and female wild-type (WT) mice were treated with paclitaxel (150 and 300 mg/kg) administered weekly over 6 weeks or exposed to 19 Gy cardiac irradiation. Cohorts were analyzed for behavioral and neurobiologic endpoints to assess systemic toxicity of paclitaxel or cardiovascular endpoints to assess radiotherapy toxicity. Interestingly, female WT mice exhibited enhanced tolerance compared to male WT mice regardless of the treatment regimen. To provide insight into the possible sex-specific protective mechanisms, rhoB-deficient animals and elderly mice (22 months) were used with a focus on the possible contribution of sex hormones, including estrogen. In females, RhoB deficiency and advanced age had no impact on neurocognitive impairment induced by paclitaxel but enhanced cardiac sensitivity to radiotherapy. Conversely, rhoB-deficiency protected males from radiation toxicity. In sum, RhoB was identified as a molecular determinant driving estrogen-dependent cardioprotection in female mice, whereas neuroprotection was not sex hormone dependent. To our knowledge, this study revealed for the first time sex- and organ-specific responses to paclitaxel and radiotherapy.
Collapse
Affiliation(s)
- Nicole Chmielewski-Stivers
- Department of Radiation Oncology, University of California at Irvine, Irvine, CA 92697, USA; (N.C.-S.); (X.L.)
| | - Benoit Petit
- Laboratory of Radiation Oncology, Radiation Oncology Service, Department of Oncology, CHUV, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland; (B.P.); (J.O.); (P.T.); (A.Q.P.)
| | - Jonathan Ollivier
- Laboratory of Radiation Oncology, Radiation Oncology Service, Department of Oncology, CHUV, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland; (B.P.); (J.O.); (P.T.); (A.Q.P.)
| | - Virginie Monceau
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), 92260 Fontenay aux Roses, France;
| | - Pelagia Tsoutsou
- Laboratory of Radiation Oncology, Radiation Oncology Service, Department of Oncology, CHUV, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland; (B.P.); (J.O.); (P.T.); (A.Q.P.)
- Department of Radiation Oncology, Hôpitaux Universitaires Genèvehug (HUG), 1205 Geneva, Switzerland
| | - Ana Quintela Pousa
- Laboratory of Radiation Oncology, Radiation Oncology Service, Department of Oncology, CHUV, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland; (B.P.); (J.O.); (P.T.); (A.Q.P.)
| | - Xiaomeng Lin
- Department of Radiation Oncology, University of California at Irvine, Irvine, CA 92697, USA; (N.C.-S.); (X.L.)
| | - Charles Limoli
- Department of Radiation Oncology, University of California at Irvine, Irvine, CA 92697, USA; (N.C.-S.); (X.L.)
- Correspondence: (C.L.); (M.-C.V.)
| | - Marie-Catherine Vozenin
- Laboratory of Radiation Oncology, Radiation Oncology Service, Department of Oncology, CHUV, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland; (B.P.); (J.O.); (P.T.); (A.Q.P.)
- Correspondence: (C.L.); (M.-C.V.)
| |
Collapse
|
19
|
Demaree JL, Ortiz RJ, Cai X, Aggarwal D, Senthilkumar I, Lawson C, Kulkarni P, Cushing BS, Ferris C. Exposure to methylphenidate during peri-adolescence decouples the prefrontal cortex: a multimodal MRI study. Am J Transl Res 2021; 13:8480-8495. [PMID: 34377346 PMCID: PMC8340152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/26/2021] [Indexed: 06/13/2023]
Abstract
This study was designed to assess the effects of daily psychostimulant exposure during juvenility and peri-adolescence on brain morphology and functional connectivity using multimodal magnetic resonance imaging. We hypothesized that long-term exposure to methylphenidate would enhance connectivity with the prefrontal cortex. Male rats were given daily injections of either methylphenidate (n=10), dextroamphetamine (n=10) or saline vehicle (n=10) from postnatal day 21 to 42. They were imaged between postnatal day 43 and 48. Voxel-based morphometry, diffusion weighted imaging, and resting state functional connectivity were used to quantify brain structure and function. Images from each modality were registered and analyzed, using a 3D MRI rat atlas providing site-specific data over 171 different brain areas. Following imaging, rats were tested for cognitive function using novel object preference. Long-lasting psychostimulant treatment was associated with only a few significant changes in brain volume and measures of anisotropy compared to vehicle. Resting state functional connectivity imaging revealed decreased coupling between the prefrontal cortex, basal ganglia and sensory motor cortices. There were no significant differences between experimental groups for cognitive behavior. In this exploratory study, we showed that chronic psychostimulant treatment throughout juvenility and preadolescence has a minimal effect on brain volume and gray matter microarchitecture, but significantly uncouples the connectivity in the cerebral/basal ganglia circuitry.
Collapse
Affiliation(s)
- Jack L Demaree
- Center for Translational NeuroImaging, Northeastern UniversityBoston, MA, USA
| | - Richard J Ortiz
- Department of Biological Sciences, University of Texas at El PasoEl Paso, TX 79968, USA
| | - Xuezhu Cai
- Center for Translational NeuroImaging, Northeastern UniversityBoston, MA, USA
| | - Dipak Aggarwal
- Center for Translational NeuroImaging, Northeastern UniversityBoston, MA, USA
| | - Ilakya Senthilkumar
- Center for Translational NeuroImaging, Northeastern UniversityBoston, MA, USA
| | - Christopher Lawson
- Center for Translational NeuroImaging, Northeastern UniversityBoston, MA, USA
| | - Praveen Kulkarni
- Center for Translational NeuroImaging, Northeastern UniversityBoston, MA, USA
| | - Bruce S Cushing
- Department of Biological Sciences, University of Texas at El PasoEl Paso, TX 79968, USA
| | - Craig Ferris
- Center for Translational NeuroImaging, Northeastern UniversityBoston, MA, USA
- Psychology and Pharmaceutical Sciences Northeastern UniversityBoston, MA, USA
| |
Collapse
|
20
|
Omran M, Belcher EK, Mohile NA, Kesler SR, Janelsins MC, Hohmann AG, Kleckner IR. Review of the Role of the Brain in Chemotherapy-Induced Peripheral Neuropathy. Front Mol Biosci 2021; 8:693133. [PMID: 34179101 PMCID: PMC8226121 DOI: 10.3389/fmolb.2021.693133] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/24/2021] [Indexed: 12/18/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a common, debilitating, and dose-limiting side effect of many chemotherapy regimens yet has limited treatments due to incomplete knowledge of its pathophysiology. Research on the pathophysiology of CIPN has focused on peripheral nerves because CIPN symptoms are felt in the hands and feet. However, better understanding the role of the brain in CIPN may accelerate understanding, diagnosing, and treating CIPN. The goals of this review are to (1) investigate the role of the brain in CIPN, and (2) use this knowledge to inform future research and treatment of CIPN. We identified 16 papers using brain interventions in animal models of CIPN and five papers using brain imaging in humans or monkeys with CIPN. These studies suggest that CIPN is partly caused by (1) brain hyperactivity, (2) reduced GABAergic inhibition, (3) neuroinflammation, and (4) overactivation of GPCR/MAPK pathways. These four features were observed in several brain regions including the thalamus, periaqueductal gray, anterior cingulate cortex, somatosensory cortex, and insula. We discuss how to leverage this knowledge for future preclinical research, clinical research, and brain-based treatments for CIPN.
Collapse
Affiliation(s)
- Maryam Omran
- University of Rochester Medical Center, Rochester, NY, United States
| | | | - Nimish A Mohile
- University of Rochester Medical Center, Rochester, NY, United States
| | - Shelli R Kesler
- The University of Texas at Austin, Austin, TX, United States
| | | | - Andrea G Hohmann
- Psychological and Brain Sciences, Program in Neuroscience and Gill Center for Biomolecular Science, Indiana University Bloomington, Bloomington, IN, United States
| | - Ian R Kleckner
- University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
21
|
Arezoumandan S, Cai X, Kalkarni P, Davis SA, Wilson K, Ferris CF, Cairns NJ, Gitcho MA. Hippocampal neurobiology and function in an aged mouse model of TDP-43 proteinopathy in an APP/PSEN1 background. Neurosci Lett 2021; 758:136010. [PMID: 34090937 DOI: 10.1016/j.neulet.2021.136010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/18/2021] [Accepted: 05/28/2021] [Indexed: 10/21/2022]
Abstract
Aging is a major risk factor for Alzheimer's disease (AD), the most common cause of dementia worldwide. TDP-43 proteinopathy is reported to be associated with AD pathology is almost 50% of cases. Our exploratory study examined near end-stage (28 months old) mice selectively driving expression of human TDP-43 in the hippocampus and cortex in an APP/PSEN1 background. We hypothesized that hippocampal neuropathology caused by β-amyloidosis with TDP-43 proteinopathy induced in this model, resembling the pathology seen in AD cases, manifest with changes in resting state functional connectivity. In vivo magnetic resonance imaging and post-mortem histology were performed on four genotypes: wild type, APP/PSEN1, Camk2a/TDP-43, and Camk2a/TDP-43/APP/PSEN1. Our results revealed loss of functional coupling in hippocampus and amygdala that was associated with severe neuronal loss in dentate gyrus of Camk2a/TDP-43/APP/PSEN1 mice compared to APP/PSEN1 and wild type mice. The loss of cells was accompanied by high background of β-amyloid plaques with sparse phosphorylated TDP-43 pathology. The survival rate was also reduced in Camk2a/TDP-43/APP/PSEN1 mice compared to other groups. This end-of-life study provides exploratory data to reach a better understanding of the role of TDP-43 hippocampal neuropathology in diseases with co-pathologies of TDP-43 proteinopathy and β-amyloidosis such as AD and limbic predominant age-related TDP-43 encephalopathy (LATE).
Collapse
Affiliation(s)
- Sanaz Arezoumandan
- Department of Biological Sciences, Delaware State University, Dover, DE, USA; Delaware Center for Neuroscience Research, Delaware State University, Dover, DE, USA
| | - Xuezhu Cai
- Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, MA, USA; Center for Translational NeuroImaging, Northeastern University, Boston, MA, USA
| | - Praveen Kalkarni
- Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, MA, USA; Center for Translational NeuroImaging, Northeastern University, Boston, MA, USA
| | - Stephani A Davis
- Department of Biological Sciences, Delaware State University, Dover, DE, USA; Delaware Center for Neuroscience Research, Delaware State University, Dover, DE, USA
| | - Katherine Wilson
- Department of Biological Sciences, Delaware State University, Dover, DE, USA; Delaware Center for Neuroscience Research, Delaware State University, Dover, DE, USA
| | - Craig F Ferris
- Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, MA, USA; Center for Translational NeuroImaging, Northeastern University, Boston, MA, USA
| | - Nigel J Cairns
- College of Medicine and Health, University of Exeter, Exeter, UK
| | - Michael A Gitcho
- Department of Biological Sciences, Delaware State University, Dover, DE, USA; Delaware Center for Neuroscience Research, Delaware State University, Dover, DE, USA.
| |
Collapse
|
22
|
Zhao YX, Yu XC, Gao JH, Yao MJ, Zhu B. Acupuncture for Paclitaxel-Induced Peripheral Neuropathy: A Review of Clinical and Basic Studies. J Pain Res 2021; 14:993-1005. [PMID: 33883931 PMCID: PMC8055287 DOI: 10.2147/jpr.s296150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/17/2021] [Indexed: 11/23/2022] Open
Abstract
Paclitaxel-induced peripheral neuropathy (PIPN) is a common and intractable side effect of the conventional chemotherapeutic agent paclitaxel. Acupuncture has been reported as an effective alternative therapy in treatment of PIPN in both basic studies and clinical trials. However, there is a lack of comprehensive surveys to summarize the action of acupuncture in management of PIPN. In this review, we briefly demonstrate the basic pathology of PIPN, which includes the activation of ion channels, mitochondrial dysfunction, disruption of axonal transport and also neuro-inflammatory involvement. Meanwhile, we review both the clinical and basic studies as an emphasis to give a general overview of the therapeutic effect of acupuncture against PIPN. Finally, we summarize the current known mechanisms underlying the action of acupuncture against PIPN mainly at peripheral and spinal levels, which include various neurotransmitters, multiple receptors, different types of enzymes and molecules. In conclusion, acupuncture could be considered as a potential alternative therapy in treatment of PIPN, and further clinical and experimental studies are called for in the future.
Collapse
Affiliation(s)
- Yu-Xue Zhao
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Xiao-Chun Yu
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Jun-Hong Gao
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Ming-Jiang Yao
- Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, People's Republic of China.,Key Laboratory of Pharmacology of Chinese Materia Medica, Beijing, 100091, People's Republic of China
| | - Bing Zhu
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| |
Collapse
|
23
|
Alkislar I, Miller AR, Hohmann AG, Sadaka AH, Cai X, Kulkarni P, Ferris CF. Inhaled Cannabis Suppresses Chemotherapy-Induced Neuropathic Nociception by Decoupling the Raphe Nucleus: A Functional Imaging Study in Rats. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2020; 6:479-489. [PMID: 33622657 DOI: 10.1016/j.bpsc.2020.11.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/30/2020] [Accepted: 11/22/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Efficacy of inhaled cannabis for treating pain is controversial. Effective treatment for chemotherapy-induced neuropathy represents an unmet medical need. We hypothesized that cannabis reduces neuropathic pain by reducing functional coupling in the raphe nuclei. METHODS We assessed the impact of inhalation of vaporized cannabis plant (containing 10.3% Δ9-tetrahydrocannabinol/0.05% cannabidiol) or placebo cannabis on brain resting-state blood oxygen level-dependent functional connectivity and pain behavior induced by paclitaxel in rats. Rats received paclitaxel to produce chemotherapy-induced peripheral neuropathy or its vehicle. Behavioral and imaging experiments were performed after neuropathy was established and stable. Images were registered to, and analyzed using, a 3D magnetic resonance imaging rat atlas providing site-specific data on more than 168 different brain areas. RESULTS Prior to vaporization, paclitaxel produced cold allodynia. Inhaled vaporized cannabis increased cold withdrawal latencies relative to prevaporization or placebo cannabis, consistent with Δ9-tetrahydrocannabinol-induced antinociception. In paclitaxel-treated rats, the midbrain serotonergic system, comprising the dorsal and median raphe, showed hyperconnectivity to cortical, brainstem, and hippocampal areas, consistent with nociceptive processing. Inhalation of vaporized cannabis uncoupled paclitaxel-induced hyperconnectivity patterns. No such changes in connectivity or cold responsiveness were observed following placebo cannabis vaporization. CONCLUSIONS Inhaled vaporized cannabis plant uncoupled brain resting-state connectivity in the raphe nuclei, normalizing paclitaxel-induced hyperconnectivity to levels observed in vehicle-treated rats. Inhaled vaporized cannabis produced antinociception in both paclitaxel- and vehicle-treated rats. Our study elucidates neural circuitry implicated in the therapeutic effects of Δ9-tetrahydrocannabinol and supports a role for functional imaging studies in animals in guiding indications for future clinical trials.
Collapse
Affiliation(s)
- Ilayda Alkislar
- Center for Translational Neuroimaging, Northeastern University, Boston, Massachusetts
| | - Alison R Miller
- Center for Translational Neuroimaging, Northeastern University, Boston, Massachusetts
| | - Andrea G Hohmann
- Psychological and Brain Sciences, Program in Neuroscience, and Gill Center for Biomolecular Science, Indiana University, Bloomington, Indiana
| | - Aymen H Sadaka
- Center for Translational Neuroimaging, Northeastern University, Boston, Massachusetts
| | - Xuezhu Cai
- Center for Translational Neuroimaging, Northeastern University, Boston, Massachusetts
| | - Praveen Kulkarni
- Center for Translational Neuroimaging, Northeastern University, Boston, Massachusetts
| | - Craig F Ferris
- Center for Translational Neuroimaging, Northeastern University, Boston, Massachusetts; Department of Psychology, Northeastern University, Boston, Massachusetts.
| |
Collapse
|
24
|
Glare P, Overton S, Aubrey K. Transition from acute to chronic pain: where cells, systems and society meet. Pain Manag 2020; 10:421-436. [PMID: 33111634 DOI: 10.2217/pmt-2019-0039] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Current treatments for chronic pain are often ineffective. At the same as searching for novel therapeutics, there is growing interest in preventing acute pain becoming chronic. While the field is still in its infancy, its knowledge base is increasingly expanding. Certainly, biomedical factors, for example, the type of tissue damage, are important but they are often not modifiable. Psychosocial risk factors (e.g., thoughts and beliefs about pain, mood, social support, workplace problems) are modifiable. There is an increasing body of research that cognitive behavioral therapy can prevent transition. Internet-based delivery of cognitive behavioral therapy improves access. Clinicians need to be aware that they may inadvertently promote pain chronification in their patients by what they say and do.
Collapse
Affiliation(s)
- Paul Glare
- Pain Management Research Institute, Faculty of Medicine & Health, University of Sydney, NSW, 2028, Australia
| | - Sarah Overton
- Pain Management Research Centre, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia
| | - Karin Aubrey
- Pain Management Research Institute, Faculty of Medicine & Health, University of Sydney, NSW, 2028, Australia.,Kolling Institute of Medical Research, University of Sydney and Northern Sydney Local Health District, NSW, 2065, Australia
| |
Collapse
|
25
|
Kulkarni P, Bhosle MR, Lu SF, Simon NS, Iriah S, Brownstein MJ, Ferris CF. Evidence of early vasogenic edema following minor head impact that can be reduced with a vasopressin V1a receptor antagonist. Brain Res Bull 2020; 165:218-227. [PMID: 33053434 DOI: 10.1016/j.brainresbull.2020.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/08/2020] [Accepted: 10/01/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Does minor head impact without signs of structural brain damage cause short-term changes in vasogenic edema as measured by an increase apparent diffusion coefficient (ADC) using diffusion weighted imaging? If so, could the increase in vasogenic edema be treated with a vasopressin V1a receptor antagonist? We hypothesized that SRX251, a highly selective V1a antagonist, would reduce vasogenic edema in response to a single minor head impact. METHODS Lightly anesthetized male rats were subjected to a sham procedure or a single hit to the forehead using a closed skull, momentum exchange model. Animals recovered in five min and were injected with saline vehicle (n = 8) or SRX251 (n = 8) at 15 min post head impact and again 7-8 hrs later. At 2 h, 6 h, and 24 h post injury, rats were anesthetized and scanned for increases in ADC, a neurological measure of vasogenic edema. Sham rats (n = 6) were exposed to anesthesia and scanned at all time points but were not hit or treated. Images were registered to and analyzed using a 3D MRI rat atlas providing site-specific data on 150 different brain areas. These brain areas were parsed into 11 major brain regions. RESULTS Untreated rats with brain injury showed a significant increase in global brain vasogenic edema as compared to sham and SRX251 treated rats. Edema peaked at 6 h in injured, untreated rats in three brain regions where changes in ADC were observed, but returned to sham levels by 24 h. There were regional variations in the time course of vasogenic edema and drug efficacy. Edema was significantly reduced in cerebellum and thalamus with SRX251 treatment while the basal ganglia did not show a response to treatment. CONCLUSION A single minor impact to the forehead causes regional increases in vasogenic edema that peak at 6 h but return to baseline within a day in a subset of brain regions. Treatment with a selective V1a receptor antagonist can reduce much of the edema.
Collapse
Affiliation(s)
- Praveen Kulkarni
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA, United States
| | - Mansi R Bhosle
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA, United States
| | - Shi-Fang Lu
- Azevan Pharmaceuticals, Bethlehem, PA, United States; Dept.of Biological Sciences, Lehigh University, Bethlehem, PA, United States
| | - Neal S Simon
- Azevan Pharmaceuticals, Bethlehem, PA, United States; Dept.of Biological Sciences, Lehigh University, Bethlehem, PA, United States
| | - Sade Iriah
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA, United States
| | | | - Craig F Ferris
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA, United States; Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, MA, United States.
| |
Collapse
|
26
|
Resonance energy transfer sensitises and monitors in situ switching of LOV2-based optogenetic actuators. Nat Commun 2020; 11:5107. [PMID: 33037199 PMCID: PMC7547724 DOI: 10.1038/s41467-020-18816-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 09/08/2020] [Indexed: 11/16/2022] Open
Abstract
Engineered light-dependent switches provide uniquely powerful opportunities to investigate and control cell regulatory mechanisms. Existing tools offer high spatiotemporal resolution, reversibility and repeatability. Cellular optogenetics applications remain limited with diffusible targets as the response of the actuator is difficult to independently validate. Blue light levels commonly needed for actuation can be cytotoxic, precluding long-term experiments. We describe a simple approach overcoming these obstacles. Resonance energy transfer can be used to constitutively or dynamically modulate actuation sensitivity. This simultaneously offers on-line monitoring of light-dependent switching and precise quantification of activation-relaxation properties in intact living cells. Applying this approach to different LOV2-based switches reveals that flanking sequences can lead to relaxation times up to 11-fold faster than anticipated. In situ–measured parameter values guide the design of target-inhibiting actuation trains with minimal blue-light exposure, and context-based optimisation can increase sensitivity and experimental throughput a further 10-fold without loss of temporal precision. Cellular optogenetics applications are limited by difficulties in quantification and blue light toxicity. Here the authors design LOV2-based switches that use resonance energy transfer to overcome these concerns.
Collapse
|
27
|
Ortiz R, Yee JR, Kulkarni PP, Solomon NG, Keane B, Cai X, Ferris CF, Cushing BS. Differences in Diffusion-Weighted Imaging and Resting-State Functional Connectivity Between Two Culturally Distinct Populations of Prairie Vole. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2020; 7:588-597. [PMID: 33239258 DOI: 10.1016/j.bpsc.2020.08.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/07/2020] [Accepted: 08/18/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND We used the highly prosocial prairie vole to test the hypothesis that higher-order brain structure-microarchitecture and functional connectivity (FC)-would differ between males from populations with distinctly different levels of prosocial behavior. Specifically, we studied males from Illinois (IL), which display high levels of prosocial behavior, and first generation males from Kansas dams and IL males (KI), which display the lowest level of prosocial behavior and higher aggression. Behavioral differences between these males are associated with overexpression of estrogen receptor alpha in the medial amygdala and bed nucleus of the stria terminalis and neuropeptide expression in the paraventricular nucleus. METHODS We compared apparent diffusion coefficient, fractional anisotropy, and blood oxygen level-dependent resting-state FC between males. RESULTS IL males displayed higher apparent diffusion coefficient in regions associated with prosocial behavior, including the bed nucleus of the stria terminalis, paraventricular nucleus, and anterior thalamic nuclei, while KI males showed higher apparent diffusion coefficient in the brainstem. KI males showed significantly higher fractional anisotropy than IL males in 26 brain regions, with the majority being in the brainstem reticular activating system. IL males showed more blood oxygen level-dependent resting-state FC between the bed nucleus of the stria terminalis, paraventricular nucleus, and medial amygdala along with other brain regions, including the hippocampus and areas associated with social and reward networks. CONCLUSIONS Our results suggest that gray matter microarchitecture and FC may play a role the expression of prosocial behavior and that differences in other brain regions, especially the brainstem, could be involved. The differences between males suggests that this system represents a potentially valuable model system for studying emotional differences and vulnerability to stress and addiction.
Collapse
Affiliation(s)
- Richard Ortiz
- Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas
| | - Jason R Yee
- Center for Translational NeuroImaging, Department of Psychology, Northeastern University, Boston, Massachusetts
| | - Praveen P Kulkarni
- Center for Translational NeuroImaging, Department of Psychology, Northeastern University, Boston, Massachusetts
| | | | - Brian Keane
- Department of Biological Sciences, Miami University, Hamilton, Ohio
| | - Xuezhu Cai
- Center for Translational NeuroImaging, Department of Psychology, Northeastern University, Boston, Massachusetts
| | - Craig F Ferris
- Center for Translational NeuroImaging, Department of Psychology, Northeastern University, Boston, Massachusetts
| | - Bruce S Cushing
- Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas.
| |
Collapse
|
28
|
Meade JA, Alkhlaif Y, Contreras KM, Obeng S, Toma W, Sim-Selley LJ, Selley DE, Damaj MI. Kappa opioid receptors mediate an initial aversive component of paclitaxel-induced neuropathy. Psychopharmacology (Berl) 2020; 237:2777-2793. [PMID: 32529265 DOI: 10.1007/s00213-020-05572-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 05/29/2020] [Indexed: 02/06/2023]
Abstract
RATIONALE Cancer patients receiving the antineoplastic drug paclitaxel report higher incidences and longer duration of treatment-resistant depression than patients receiving other classes of chemotherapeutics. Rodents treated with paclitaxel exhibit a suite of changes in affect-like behaviors. Further, paclitaxel causes chemotherapy-induced peripheral neuropathy (CIPN) in humans and rodents. Kappa opioid receptors (KOR) have a well-established role in depression and neuropathy. The contributions of KOR signaling to paclitaxel-induced aversive-like state and CIPN in rodents remain to be explored. OBJECTIVES We aimed to investigate whether dysregulation of the KOR/dynorphin system is associated with paclitaxel-mediated pain-like behavior and depression-like behavior. METHODS Cancer-free male C57BL/6J mice were treated with four injections of vehicle or paclitaxel (32 mg/kg cumulative). The effects of the selective KOR antagonist norbinaltorphimine (norBNI) on paclitaxel-induced sucrose preference deficits and mechanical hypersensitivity were measured. Prodynorphin mRNA and receptor-mediated G protein activation were measured at two time points following the last paclitaxel injection using quantitative real-time polymerase chain reaction and agonist-stimulated [35S]guanosine-5'-O'-(γ-thio)-triphosphate ([35S]GTPγS) binding, respectively, in the nucleus accumbens (NAc), caudate-putamen, amygdala, and spinal cord. RESULTS Paclitaxel produced a norBNI-reversible sucrose preference deficit, whereas mechanical hypersensitivity was not reversed by norBNI. Paclitaxel treatment increased the levels of mRNA for prodynorphin, a precursor for endogenous KOR agonists, in the NAc. Paclitaxel also had time-dependent effects on KOR-mediated G protein activation in the NAc. CONCLUSIONS These results suggest that KOR signaling mediates an initial aversive component of paclitaxel, but not necessarily paclitaxel-induced mechanical hypersensitivity.
Collapse
Affiliation(s)
- Julie A Meade
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia Campus, Box 980613, Richmond, VA, 23298-0613, USA.
| | - Y Alkhlaif
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia Campus, Box 980613, Richmond, VA, 23298-0613, USA
| | - K M Contreras
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia Campus, Box 980613, Richmond, VA, 23298-0613, USA
| | - S Obeng
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - W Toma
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia Campus, Box 980613, Richmond, VA, 23298-0613, USA
| | - L J Sim-Selley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia Campus, Box 980613, Richmond, VA, 23298-0613, USA
| | - D E Selley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia Campus, Box 980613, Richmond, VA, 23298-0613, USA
| | - M I Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Medical College of Virginia Campus, Box 980613, Richmond, VA, 23298-0613, USA.,Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, Richmond, VA, 23298, USA
| |
Collapse
|
29
|
Sałat K. Chemotherapy-induced peripheral neuropathy: part 1-current state of knowledge and perspectives for pharmacotherapy. Pharmacol Rep 2020; 72:486-507. [PMID: 32394362 PMCID: PMC7329796 DOI: 10.1007/s43440-020-00109-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/20/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022]
Abstract
Background Despite the increasing knowledge of the etiology of neuropathic pain, this type of chronic pain is resistant to available analgesics in approximately 50% of patients and therefore is continuously a subject of considerable interest for physiologists, neurologists, medicinal chemists, pharmacologists and others searching for more effective treatment options for this debilitating condition. Materials and methods The present review article is the first of the two articles focused on chemotherapy-induced peripheral neuropathy (CIPN). Results CIPN is regarded as one of the most common drug-induced neuropathies and is highly pharmacoresistant. The lack of efficacious pharmacological methods for treating CIPN and preventing its development makes CIPN-related neuropathic pain a serious therapeutic gap in current medicine and pharmacotherapy. In this paper, the most recent advances in the field of studies on CIPN caused by platinum compounds (namely oxaliplatin and cisplatin), taxanes, vinca alkaloids and bortezomib are summarized. Conclusions The prevalence of CIPN, potential causes, risk factors, symptoms and molecular mechanisms underlying this pharmacoresistant condition are discussed. Graphic abstract ![]()
Collapse
Affiliation(s)
- Kinga Sałat
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St., 30-688, Krakow, Poland.
| |
Collapse
|
30
|
Slivicki RA, Iyer V, Mali SS, Garai S, Thakur GA, Crystal JD, Hohmann AG. Positive Allosteric Modulation of CB 1 Cannabinoid Receptor Signaling Enhances Morphine Antinociception and Attenuates Morphine Tolerance Without Enhancing Morphine- Induced Dependence or Reward. Front Mol Neurosci 2020; 13:54. [PMID: 32410959 PMCID: PMC7199816 DOI: 10.3389/fnmol.2020.00054] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 03/17/2020] [Indexed: 01/09/2023] Open
Abstract
Opioid analgesics represent a critical treatment for chronic pain in the analgesic ladder of the World Health Organization. However, their use can result in a number of unwanted side-effects including incomplete efficacy, constipation, physical dependence, and overdose liability. Cannabinoids enhance the pain-relieving effects of opioids in preclinical studies and dampen unwanted side-effects resulting from excessive opioid intake. We recently reported that a CB1 positive allosteric modulator (PAM) exhibits antinociceptive efficacy in models of pathological pain and lacks the adverse side effects of direct CB1 receptor activation. In the present study, we evaluated whether a CB1 PAM would enhance morphine’s therapeutic efficacy in an animal model of chemotherapy-induced neuropathic pain and characterized its impact on unwanted side-effects associated with chronic opioid administration. In paclitaxel-treated mice, both the CB1 PAM GAT211 and the opioid analgesic morphine reduced paclitaxel-induced behavioral hypersensitivities to mechanical and cold stimulation in a dose-dependent manner. Isobolographic analysis revealed that combinations of GAT211 and morphine resulted in anti-allodynic synergism. In paclitaxel-treated mice, a sub-threshold dose of GAT211 prevented the development of tolerance to the anti-allodynic effects of morphine over 20 days of once daily dosing. However, GAT211 did not reliably alter somatic withdrawal signs (i.e., jumps, paw tremors) in morphine-dependent neuropathic mice challenged with naloxone. In otherwise naïve mice, GAT211 also prolonged antinociceptive efficacy of morphine in the tail-flick test and reduced the overall right-ward shift in the ED50 for morphine to produce antinociception in the tail-flick test, consistent with attenuation of morphine tolerance. Pretreatment with GAT211 did not alter somatic signs of μ opioid receptor dependence in mice rendered dependent upon morphine via subcutaneous implantation of a morphine pellet. Moreover, GAT211 did not reliably alter μ-opioid receptor-mediated reward as measured by conditioned place preference to morphine. Our results suggest that a CB1 PAM may be beneficial in enhancing and prolonging the therapeutic properties of opioids while potentially sparing unwanted side-effects (e.g., tolerance) that occur with repeated opioid treatment.
Collapse
Affiliation(s)
- Richard A Slivicki
- Program in Neuroscience, Indiana University, Bloomington, IN, United States.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Vishakh Iyer
- Program in Neuroscience, Indiana University, Bloomington, IN, United States.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Sonali S Mali
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Sumanta Garai
- Center for Drug Discovery, Bouve College of Health Sciences, Northeastern University, Boston, MA, United States
| | - Ganesh A Thakur
- Center for Drug Discovery, Bouve College of Health Sciences, Northeastern University, Boston, MA, United States
| | - Jonathon D Crystal
- Program in Neuroscience, Indiana University, Bloomington, IN, United States.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Andrea G Hohmann
- Program in Neuroscience, Indiana University, Bloomington, IN, United States.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States.,Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States
| |
Collapse
|
31
|
Costa-Pereira JT, Ribeiro J, Martins I, Tavares I. Role of Spinal Cord α 2-Adrenoreceptors in Noradrenergic Inhibition of Nociceptive Transmission During Chemotherapy-Induced Peripheral Neuropathy. Front Neurosci 2020; 13:1413. [PMID: 32009887 PMCID: PMC6974806 DOI: 10.3389/fnins.2019.01413] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/13/2019] [Indexed: 12/14/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a problem during cancer treatment and for cancer survivors but the central mechanisms underlying CIPN remain understudied. This study aims to determine if CIPN is associated with alterations of noradrenergic modulation of nociceptive transmission at the spinal cord. CIPN was induced in male Wistar rats by paclitaxel injections. One month after CIPN induction, the behavioral effects of the administration of reboxetine (noradrenaline reuptake inhibitor), clonidine (agonist of α2-adrenoreceptors; α2–AR) and atipamezole (antagonist of α2–AR) were evaluated using the von Frey and cold plate tests. Furthermore, we measured the expression of the noradrenaline biosynthetic enzyme dopamine-β-hydroxylase (DBH) and of α2–AR in the spinal dorsal horn. Reboxetine and clonidine reversed the behavioral signs of CIPN whereas the opposite occurred with atipamezole. In the 3 pharmacological approaches, a higher effect was detected in mechanical allodynia, the pain modality which is under descending noradrenergic control. DBH expression was increased at the spinal dorsal horn of paclitaxel-injected animals. The enhanced noradrenergic inhibition during CIPN may represent an adaptation of the descending noradrenergic pain control system to the increased arrival of peripheral nociceptive input. A potentiation of the α2–AR mediated antinociception at the spinal cord may represent a therapeutic opportunity to face CIPN.
Collapse
Affiliation(s)
- José Tiago Costa-Pereira
- Unit of Experimental Biology, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,Institute of Molecular and Cell Biology, University of Porto, Porto, Portugal.,I3S-Institute for Investigation and Innovation in Health, University of Porto, Porto, Portugal
| | - Joana Ribeiro
- Unit of Experimental Biology, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,Institute of Molecular and Cell Biology, University of Porto, Porto, Portugal.,I3S-Institute for Investigation and Innovation in Health, University of Porto, Porto, Portugal
| | - Isabel Martins
- Unit of Experimental Biology, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,Institute of Molecular and Cell Biology, University of Porto, Porto, Portugal.,I3S-Institute for Investigation and Innovation in Health, University of Porto, Porto, Portugal
| | - Isaura Tavares
- Unit of Experimental Biology, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,Institute of Molecular and Cell Biology, University of Porto, Porto, Portugal.,I3S-Institute for Investigation and Innovation in Health, University of Porto, Porto, Portugal
| |
Collapse
|
32
|
Costa‐Pereira JT, Serrão P, Martins I, Tavares I. Serotoninergic pain modulation from the rostral ventromedial medulla (RVM) in chemotherapy‐induced neuropathy: The role of spinal 5‐HT3 receptors. Eur J Neurosci 2019; 51:1756-1769. [DOI: 10.1111/ejn.14614] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 10/03/2019] [Accepted: 10/30/2019] [Indexed: 01/31/2023]
Affiliation(s)
- José Tiago Costa‐Pereira
- Department of Biomedicine Unit of Experimental Biology Faculty of Medicine University of Porto Porto Portugal
- IBMC‐Institute of Molecular and Cell Biology University of Porto Porto Portugal
- I3S‐ Institute of Investigation and Innovation in Health University of Porto Porto Portugal
| | - Paula Serrão
- Department of Biomedicine Unit of Pharmacology and Therapeutics Faculty of Medicine University of Porto Porto Portugal
- MedInUP ‐ Center for Drug Discovery and Innovative Medicines University of Porto Porto Portugal
| | - Isabel Martins
- Department of Biomedicine Unit of Experimental Biology Faculty of Medicine University of Porto Porto Portugal
- IBMC‐Institute of Molecular and Cell Biology University of Porto Porto Portugal
- I3S‐ Institute of Investigation and Innovation in Health University of Porto Porto Portugal
| | - Isaura Tavares
- Department of Biomedicine Unit of Experimental Biology Faculty of Medicine University of Porto Porto Portugal
- IBMC‐Institute of Molecular and Cell Biology University of Porto Porto Portugal
- I3S‐ Institute of Investigation and Innovation in Health University of Porto Porto Portugal
| |
Collapse
|
33
|
Slivicki RA, Mali SS, Hohmann AG. Voluntary exercise reduces both chemotherapy-induced neuropathic nociception and deficits in hippocampal cellular proliferation in a mouse model of paclitaxel-induced peripheral neuropathy. NEUROBIOLOGY OF PAIN 2019; 6:100035. [PMID: 31528755 PMCID: PMC6739464 DOI: 10.1016/j.ynpai.2019.100035] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/21/2019] [Accepted: 08/25/2019] [Indexed: 12/18/2022]
Abstract
Paclitaxel treatment did not alter voluntary running activity. Voluntary running reduced mechanical and cold allodynia induced by paclitaxel. Voluntary running reduced paclitaxel-induced deficits in hippocampal cellular proliferation.
Chemotherapy-induced peripheral neuropathy (CIPN) is a common dose-limiting side-effect of all major chemotherapeutic agents. Here, we explored efficacy of voluntary exercise as a nonpharmacological strategy for suppressing two distinct adverse side effects of chemotherapy treatment. We evaluated whether voluntary running would suppress both neuropathic pain and deficits in hippocampal cell proliferation in a mouse model of CIPN induced by the taxane chemotherapeutic agent paclitaxel. Mice were given free access to running wheels or were housed without running wheels during one of three different intervention phases: 1) during the onset (i.e. development phase) of paclitaxel-induced neuropathy, 2) prior to dosing with paclitaxel or its vehicle, or 3) following the establishment (i.e. maintenance phase) of paclitaxel-induced neuropathy. Paclitaxel treatment did not alter running wheel behavior relative to vehicle-treated animals in any study. Animals that engaged in voluntary running during the development phase of paclitaxel-induced neuropathy failed to display mechanical or cold hypersensitivities relative to sedentary control animals that did not have access to running wheels. A prior history of voluntary running delayed the onset of, but did not fully prevent, development of paclitaxel-induced neuropathic pain behavior. Voluntary running reduced already established mechanical and cold allodynia induced by paclitaxel. Importantly, voluntary running did not alter mechanical or cold responsivity in vehicle-treated animals, suggesting that the observed antinociceptive effect of exercise was dependent upon the presence of the pathological pain state. In the same animals evaluated for nociceptive responding, paclitaxel also reduced cellular proliferation but not cellular survival in the dentate gyrus of the hippocampus, as measured by immunohistochemistry for Ki67 and BrdU expression, respectively. Voluntary running abrogated paclitaxel-induced reductions in cellular proliferation to levels observed in vehicle-treated mice and also increased BrdU expression levels irrespective of chemotherapy treatment. Our studies support the hypothesis that voluntary exercise may be beneficial in suppressing both neuropathic pain and markers of hippocampal cellular function that are impacted by toxic challenge with chemotherapeutic agents.
Collapse
Affiliation(s)
- Richard A. Slivicki
- Program in Neuroscience, Indiana University, Bloomington, IN, United States
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Sonali S. Mali
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
| | - Andrea G. Hohmann
- Program in Neuroscience, Indiana University, Bloomington, IN, United States
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States
- Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, United States
- Corresponding author at: Department of Psychological and Brain Sciences, Indiana University, 1101 E 10th Street, Bloomington, IN 47405-7007, United States.
| |
Collapse
|