1
|
Coluzzi F, Scerpa MS, Alessandri E, Romualdi P, Rocco M. Role of TRP Channels in Cancer-Induced Bone Pain. Int J Mol Sci 2025; 26:1229. [PMID: 39940997 PMCID: PMC11818569 DOI: 10.3390/ijms26031229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
The burden of cancer is growing in almost every country. Bone metastases significantly affect the prognosis and lead to an increase in mortality and morbidity. The management of cancer-induced bone pain (CIBP) still shows various unmet needs. Opioid use is burdened by a number of possible side effects. Moreover, recent progresses in cancer treatment significantly increased the life expectancy of cancer patients, even those with metastatic disease. In this narrative review, we reported the main findings regarding TRP channel function in cancer pain models. TRP cation channels play a key role in different functions of cancer cells, including the regulation of their potential for metastasization, and are the main channels involved in the pathways of pain perception, through peripheral and central effects. Genetic deletion decreased pain sensitivity following tumour cell inoculation. Preclinical data suggest a potential role for modulators of some TRP channels, such as TRPV1, TRPA1, TRPM7 and TRPM8. Clinical results are still scarce; however, the physiological role in modulating bone remodelling and the involvement of TRP channels in preclinical models of bone cancer pain have garnered interest as areas of research in the last few years, as innovative analgesic strategies that may overcome the long-term side effects of opioids.
Collapse
Affiliation(s)
- Flaminia Coluzzi
- Department of Surgical and Medical Sciences and Translational Medicine, Sapienza University of Rome, 00189 Rome, Italy
- Unit Anesthesia, Intensive Care and Pain Therapy, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Maria Sole Scerpa
- Unit Anesthesia, Intensive Care and Pain Therapy, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Elisa Alessandri
- Unit Anesthesia, Intensive Care and Pain Therapy, Sant’Andrea University Hospital, 00189 Rome, Italy
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, 40126 Bologna, Italy
| | - Monica Rocco
- Department of Surgical and Medical Sciences and Translational Medicine, Sapienza University of Rome, 00189 Rome, Italy
- Unit Anesthesia, Intensive Care and Pain Therapy, Sant’Andrea University Hospital, 00189 Rome, Italy
| |
Collapse
|
2
|
Zhang Y, Xie J. Unveiling the role of ferroptosis-associated exosomal non-coding RNAs in cancer pathogenesis. Biomed Pharmacother 2024; 172:116235. [PMID: 38308967 DOI: 10.1016/j.biopha.2024.116235] [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: 11/22/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024] Open
Abstract
The pivotal regulatory role of non-coding RNAs (ncRNAs), especially exosomal ncRNAs, in ferroptosis significantly influences cancer cell fate. This review explores their involvement across various human cancers, focusing on microRNAs (miRNA), long non-coding RNAs (lncRNA), and circular RNAs (circRNA). These ncRNAs either stimulate or inhibit ferroptosis by targeting key components, impacting cancer susceptibility to this form of cell death. Specific studies in lung, gastric, liver, cervical, bladder, pancreatic, and osteosarcoma cancers underscore the crucial role of exosomal ncRNAs in modulating ferroptosis, influencing cancer progression, and therapeutic responses. Emphasizing the therapeutic potential of exosomal ncRNAs, we discuss their ability to deliver circRNA, miRNA, and lncRNA to target cells. Despite being in early stages with challenges in bioengineering for drug delivery, these studies hold promise for future clinical applications. Noteworthy findings include inhibiting exosome production to overcome ferroptosis resistance in lung adenocarcinoma and the potential of exosomal DACT3-AS1 to sensitize gastric cancer cells to ferroptosis. The review concludes by highlighting exosomal ncRNAs like miR-4443 and miR-660-5p as promising therapeutic targets, offering avenues for precise cancer interventions by modulating signaling pathways and sensitizing cells to ferroptosis. Overall, this review enhances our understanding of cancer pathogenesis and presents new horizons for targeted therapeutic interventions, revealing the intricate interplay between exosomal ncRNAs and ferroptosis.
Collapse
Affiliation(s)
- Yiping Zhang
- School of Life Sciences, Fudan University, Shanghai 200438, China; Wanchuanhui (Shanghai) Medical Technology Co., Ltd, Shanghai 201501, China.
| | - Jun Xie
- School of Life Sciences, Fudan University, Shanghai 200438, China; Wanchuanhui (Shanghai) Medical Technology Co., Ltd, Shanghai 201501, China.
| |
Collapse
|
3
|
Peterson IL, Thompson AD, Scholpa NE, Largent-Milnes T, Schnellmann RG. Isolation and monoculture of functional primary astrocytes from the adult mouse spinal cord. Front Neurosci 2024; 18:1367473. [PMID: 38435055 PMCID: PMC10906264 DOI: 10.3389/fnins.2024.1367473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/02/2024] [Indexed: 03/05/2024] Open
Abstract
Astrocytes are a widely heterogenic cell population that play major roles in central nervous system (CNS) homeostasis and neurotransmission, as well as in various neuropathologies, including spinal cord injury (SCI), traumatic brain injury, and neurodegenerative diseases, such as amyotrophic lateral sclerosis. Spinal cord astrocytes have distinct differences from those in the brain and accurate modeling of disease states is necessary for understanding disease progression and developing therapeutic interventions. Several limitations to modeling spinal cord astrocytes in vitro exist, including lack of commercially available adult-derived cells, lack of purchasable astrocytes with different genotypes, as well as time-consuming and costly in-house primary cell isolations that often result in low yield due to small tissue volume. To address these issues, we developed an efficient adult mouse spinal cord astrocyte isolation method that utilizes enzymatic digestion, debris filtration, and multiple ACSA-2 magnetic microbead purification cycles to achieve an astrocyte monoculture purity of ≅93-98%, based on all markers assessed. Importantly, the isolated cells contain active mitochondria and express key astrocyte markers including ACSA-1, ACSA-2, EAAT2, and GFAP. Furthermore, this isolation method can be applied to the spinal cord of male and female mice, mice subjected to SCI, and genetically modified mice. We present a primary adult mouse spinal cord astrocyte isolation protocol focused on purity, viability, and length of isolation that can be applied to a multitude of models and aid in targeted research on spinal-cord related CNS processes and pathologies.
Collapse
Affiliation(s)
- Ingrid L. Peterson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, United States
| | - Austin D. Thompson
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, United States
- Southern Arizona VA Health Care System, Tucson, AZ, United States
- Southwest Environmental Health Science Center, University of Arizona, Tucson, AZ, United States
| | - Natalie E. Scholpa
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, United States
- Southern Arizona VA Health Care System, Tucson, AZ, United States
| | - Tally Largent-Milnes
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Rick G. Schnellmann
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, United States
- Southern Arizona VA Health Care System, Tucson, AZ, United States
- Southwest Environmental Health Science Center, University of Arizona, Tucson, AZ, United States
- Department of Neuroscience, College of Medicine, University of Arizona, Tucson, AZ, United States
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
4
|
Jimenez-Andrade JM, Ramírez-Rosas MB, Hee Park S, Parker R, Eber MR, Cain R, Newland M, Hsu FC, Kittel CA, Martin TJ, Muñoz-Islas E, Shiozawa Y, Peters CM. Evaluation of pain related behaviors and disease related outcomes in an immunocompetent mouse model of prostate cancer induced bone pain. J Bone Oncol 2023; 43:100510. [PMID: 38075938 PMCID: PMC10701434 DOI: 10.1016/j.jbo.2023.100510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 02/12/2024] Open
Abstract
Cancer-induced bone pain (CIBP) is the most common and devastating symptom of bone metastatic cancer that substantially disrupts patients' quality of life. Currently, there are few effective analgesic treatments for CIBP other than opioids which come with severe side effects. In order to better understand the factors and mechanisms responsible for CIBP it is essential to have clinically relevant animal models that mirror pain-related symptoms and disease progression observed in patients with bone metastatic cancer. In the current study, we characterize a syngeneic mouse model of prostate cancer induced bone pain. We transfected a prostate cancer cell line (RM1) with green fluorescent protein (GFP) and luciferase reporters in order to visualize tumor growth longitudinally in vivo and to assess the relationship between sensory neurons and tumor cells within the bone microenvironment. Following intra-femoral injection of the RM1 prostate cancer cell line into male C57BL/6 mice, we observed a progressive increase in spontaneous guarding of the inoculated limb between 12 and 21 days post inoculation in tumor bearing compared to sham operated mice. Daily running wheel performance was evaluated as a measure of functional impairment and potentially movement evoked pain. We observed a progressive reduction in the distance traveled and percentage of time at optimal velocity between 12 and 21 days post inoculation in tumor bearing compared to sham operated mice. We utilized histological, radiographic and μCT analysis to examine tumor induced bone remodeling and observed osteolytic lesions as well as extra-periosteal aberrant bone formation in the tumor bearing femur, similar to clinical findings in patients with bone metastatic prostate cancer. Within the tumor bearing femur, we observed reorganization of blood vessels, macrophage and nerve fibers within the intramedullary space and periosteum adjacent to tumor cells. Tumor bearing mice displayed significant increases in the injury marker ATF3 and upregulation of the neuropeptides SP and CGRP in the ipsilateral DRG as well as increased measures of central sensitization and glial activation in the ipsilateral spinal cord. This immunocompetent mouse model will be useful when combined with cell type selective transgenic mice to examine tumor, immune cell and sensory neuron interactions in the bone microenvironment and their role in pain and disease progression associated with bone metastatic prostate cancer.
Collapse
Affiliation(s)
| | - Martha B. Ramírez-Rosas
- Universidad Autónoma de Tamaulipas, Campus Reynosa Aztlán, Reynosa, Tamaulipas, 88700 Mexico
| | - Sun Hee Park
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Renee Parker
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Matthew R. Eber
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Rebecca Cain
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Mary Newland
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Carol A. Kittel
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Thomas J. Martin
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Enriqueta Muñoz-Islas
- Universidad Autónoma de Tamaulipas, Campus Reynosa Aztlán, Reynosa, Tamaulipas, 88700 Mexico
| | - Yusuke Shiozawa
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Christopher M. Peters
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| |
Collapse
|
5
|
Thompson AL, Grenald SA, Ciccone HA, Mohty D, Smith AF, Coleman DL, Bahramnejad E, De Leon E, Kasper-Conella L, Uhrlab JL, Margolis DS, Salvemini D, Largent-Milnes TM, Vanderah TW. Morphine-induced osteolysis and hypersensitivity is mediated through toll-like receptor-4 in a murine model of metastatic breast cancer. Pain 2023; 164:2463-2476. [PMID: 37326644 PMCID: PMC10578422 DOI: 10.1097/j.pain.0000000000002953] [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] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/28/2023] [Accepted: 04/18/2023] [Indexed: 06/17/2023]
Abstract
ABSTRACT The propensity for breast cancer to metastasize to bone is coupled to the most common complaint among breast cancer patients: bone pain. Classically, this type of pain is treated using escalating doses of opioids, which lack long-term efficacy due to analgesic tolerance, opioid-induced hypersensitivity, and have recently been linked to enhanced bone loss. To date, the molecular mechanisms underlying these adverse effects have not been fully explored. Using an immunocompetent murine model of metastatic breast cancer, we demonstrated that sustained morphine infusion induced a significant increase in osteolysis and hypersensitivity within the ipsilateral femur through the activation of toll-like receptor-4 (TLR4). Pharmacological blockade with TAK242 (resatorvid) as well as the use of a TLR4 genetic knockout ameliorated the chronic morphine-induced osteolysis and hypersensitivity. Genetic MOR knockout did not mitigate chronic morphine hypersensitivity or bone loss. In vitro studies using RAW264.7 murine macrophages precursor cells demonstrated morphine-enhanced osteoclastogenesis that was inhibited by the TLR4 antagonist. Together, these data indicate that morphine induces osteolysis and hypersensitivity that are mediated, in part, through a TLR4 receptor mechanism.
Collapse
Affiliation(s)
- Austen L. Thompson
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Shaness A. Grenald
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Haley A. Ciccone
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Dieter Mohty
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Angela F. Smith
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Deziree L. Coleman
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Erfan Bahramnejad
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Erick De Leon
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, United States
| | - Logan Kasper-Conella
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, United States
| | | | - David S. Margolis
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, United States
- Orthopaedic Surgery, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Daniela Salvemini
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Tally M. Largent-Milnes
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
- Comprehensive Pain and Addiction Center, University of Arizona, Tucson, AZ, United States
| | - Todd W. Vanderah
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
- Comprehensive Pain and Addiction Center, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
6
|
Abstract
PURPOSE OF REVIEW Breast and prostate tumors frequently metastasize to the bone, but the underlying mechanisms for osteotropism remain elusive. An emerging feature of metastatic progression is metabolic adaptation of cancer cells to new environments. This review will summarize the recent advances on how cancer cells utilize amino acid metabolism during metastasis, from early dissemination to interactions with the bone microenvironment. RECENT FINDINGS Recent studies have suggested that certain metabolic preferences for amino acids may be associated with bone metastasis. Once in the bone microenvironment, cancer cells encounter a favorable microenvironment, where a changing nutrient composition of the tumor-bone microenvironment may alter metabolic interactions with bone-resident cells to further drive metastatic outgrowth. Enhanced amino acid metabolic programs are associated with bone metastatic disease and may be further augmented by the bone microenvironment. Additional studies are necessary to fully elucidate the role of amino acid metabolism on bone metastasis.
Collapse
Affiliation(s)
- Deanna N Edwards
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN, 37232, USA.
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA.
| |
Collapse
|
7
|
Brusco I, Becker G, Palma TV, Pillat MM, Scussel R, Steiner BT, Sampaio TB, Ardisson-Araújo DMP, de Andrade CM, Oliveira MS, Machado-De-Avila RA, Oliveira SM. Kinin B 1 and B 2 receptors mediate cancer pain associated with both the tumor and oncology therapy using aromatase inhibitors. Sci Rep 2023; 13:4418. [PMID: 36932156 PMCID: PMC10023805 DOI: 10.1038/s41598-023-31535-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
Pain caused by the tumor or aromatase inhibitors (AIs) is a disabling symptom in breast cancer survivors. Their mechanisms are unclear, but pro-algesic and inflammatory mediators seem to be involved. Kinins are endogenous algogenic mediators associated with various painful conditions via B1 and B2 receptor activation, including chemotherapy-induced pain and breast cancer proliferation. We investigate the involvement of the kinin B1 and B2 receptors in metastatic breast tumor (4T1 breast cancer cells)-caused pain and in aromatase inhibitors (anastrozole or letrozole) therapy-associated pain. A protocol associating the tumor and antineoplastic therapy was also performed. Kinin receptors' role was investigated via pharmacological antagonism, receptors protein expression, and kinin levels. Mechanical and cold allodynia and muscle strength were evaluated. AIs and breast tumor increased kinin receptors expression, and tumor also increased kinin levels. AIs caused mechanical allodynia and reduced the muscle strength of mice. Kinin B1 (DALBk) and B2 (Icatibant) receptor antagonists attenuated these effects and reduced breast tumor-induced mechanical and cold allodynia. AIs or paclitaxel enhanced breast tumor-induced mechanical hypersensitivity, while DALBk and Icatibant prevented this increase. Antagonists did not interfere with paclitaxel's cytotoxic action in vitro. Thus, kinin B1 or B2 receptors can be a potential target for treating the pain caused by metastatic breast tumor and their antineoplastic therapy.
Collapse
Affiliation(s)
- Indiara Brusco
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Av. Roraima 1000, Camobi, Santa Maria, RS, 97105-900, Brazil.
| | - Gabriela Becker
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Av. Roraima 1000, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - Tais Vidal Palma
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Av. Roraima 1000, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - Micheli Mainardi Pillat
- Department of Microbiology and Parasitology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Rahisa Scussel
- Graduate Program in Health Sciences, University of Extreme South Catarinense, Criciuma, SC, Brazil
| | - Bethina Trevisol Steiner
- Graduate Program in Health Sciences, University of Extreme South Catarinense, Criciuma, SC, Brazil
| | - Tuane Bazanella Sampaio
- Graduate Program in Pharmacology, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Daniel Mendes Pereira Ardisson-Araújo
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Av. Roraima 1000, Camobi, Santa Maria, RS, 97105-900, Brazil
- Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia, DF, Brazil
| | - Cinthia Melazzo de Andrade
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Av. Roraima 1000, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - Mauro Schneider Oliveira
- Graduate Program in Pharmacology, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | | | - Sara Marchesan Oliveira
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Av. Roraima 1000, Camobi, Santa Maria, RS, 97105-900, Brazil.
| |
Collapse
|
8
|
Hu JL, Zhang WJ. The role and pharmacological properties of P2Y12 receptor in cancer and cancer pain. Biomed Pharmacother 2023; 157:113927. [PMID: 36462316 DOI: 10.1016/j.biopha.2022.113927] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/22/2022] [Accepted: 10/24/2022] [Indexed: 12/05/2022] Open
Abstract
The G protein-coupled P2Y12 receptor (P2Y12R) was cloned in platelets and found to play a key role in maintaining platelet function in hemostasis and thrombosis, and these effects could be mediated by the P2Y12R. However, it has recently been found that P2Y12R-mediated the progression of tumor through interactions between platelets and tumor and stromal cells, as well as through products secreted by platelets. During tumor progression, tumor cells or other cells in the tumor microenvironment (such as immune cells) can secrete large amounts of ATP into the extracellular matrix, and extracellular ATP can be hydrolyzed into ADP. ADP is a P2Y12R activator and plays an important regulatory role in the proliferation and metastasis of tumor cells. P2Y12R is involved in platelet-cancer cell crosstalk and become a potential target for anticancer therapy. Moreover, tumor progression can induce pain, which seriously affects the quality of life of patients. P2Y12R is expressed in microglia and mediates the activities of microglial and participates in the occurrence of cancer pain. Conversely, inhibiting P2Y12R activation and down-regulating its expression has the effect of inhibiting tumor progression and pain. Therefore, P2Y12R can be a common therapeutic target for both. In this article, we explored the potential link between P2Y12R and cancer, discussed the intrinsic link of P2Y12R in cancer pain and the pharmacological properties of P2Y12R antagonists in the treatment of both.
Collapse
Affiliation(s)
- Jia-Ling Hu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province 343000, China
| | - Wen-Jun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province 343000, China.
| |
Collapse
|
9
|
Hang TD, Hung HM, Beckers P, Desmet N, Lamrani M, Massie A, Hermans E, Vanommeslaeghe K. Structural investigation of human cystine/glutamate antiporter system xc− (Sxc−) using homology modeling and molecular dynamics. Front Mol Biosci 2022; 9:1064199. [DOI: 10.3389/fmolb.2022.1064199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/15/2022] [Indexed: 12/04/2022] Open
Abstract
The cystine/glutamate antiporter system xc− (Sxc−) belongs to the SLC7 family of plasma membrane transporters. It exports intracellular glutamate along the latter’s concentration gradient as a driving force for cellular uptake of cystine. Once imported, cystine is mainly used for the production of glutathione, a tripeptide thiol crucial in maintenance of redox homeostasis and protection of cells against oxidative stress. Overexpression of Sxc− has been found in several cancer cells, where it is thought to counteract the increased oxidative stress. In addition, Sxc− is important in the central nervous system, playing a complex role in regulating glutamatergic neurotransmission and glutamate toxicity. Accordingly, this transporter is considered a potential target for the treatment of cancer as well as neurodegenerative diseases. Till now, no specific inhibitors are available. We herein present four conformations of Sxc− along its transport pathway, obtained using multi-template homology modeling and refined by means of Molecular Dynamics. Comparison with a very recently released cryo-EM structure revealed an excellent agreement with our inward-open conformation. Intriguingly, our models contain a structured N-terminal domain that is unresolved in the experimental structures and is thought to play a gating role in the transport mechanism of other SLC7 family members. In contrast to the inward-open model, there is no direct experimental counterpart for the other three conformations we obtained, although they are in fair agreement with the other stages of the transport mechanism seen in other SLC7 transporters. Therefore, our models open the prospect for targeting alternative Sxc− conformations in structure-based drug design efforts.
Collapse
|
10
|
Xu L, Wang S, Zhang L, Liu B, Zheng S, Yao M. Cobratoxin Alleviates Cancer-Induced Bone Pain in Rats via Inhibiting CaMKII Signaling Pathway after Acting on M4 Muscarinic Cholinergic Receptors. ACS Chem Neurosci 2022; 13:1422-1432. [PMID: 35420768 DOI: 10.1021/acschemneuro.2c00048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Cancer-induced bone pain (CIBP) is a common pain in clinics, which can reduce the quality of life and increase the mortality of patients, but the treatment of CIBP is limited. This study was designed to investigate the analgesic effect of α-cobratoxin on CIBP and further to explore the molecular target and potential signal pathway. As shown by the mechanical allodynia test in a CIBP rat model, administration of α-cobratoxin produced significant analgesia in a dose-dependent manner, and the analgesic effects were blocked by pretreatment with an intrathecal injection of M4 mAChR-siRNA or intraperitoneal injection of tropicamide, an antagonist of M4 muscarinic cholinergic receptor. Whole-cell patch-clamp recording showed that α-cobratoxin can decrease the spontaneous firing and spontaneous excitatory postsynaptic currents of SDH neurons in CIBP rats. In primary lumber SDH neurons, intracellular calcium measurement revealed that α-cobratoxin decreased intracellular calcium concentration, and immunofluorescence demonstrated that M4 muscarinic cholinergic receptor and CaMKII/CREB were co-expressed. In the CIBP model and primary SDH neurons, Western blot showed that the levels of p-CaMKII and p-CREB were increased by α-cobratoxin and the effect of α-cobratoxin was antagonized by M4 mAChR-siRNA. The quantitative polymerase chain reaction (qPCR) results showed that α-cobratoxin downregulated the expression of proinflammatory cytokines through M4 muscarinic cholinergic receptor in SDH. These results suggest that α-cobratoxin may activate M4 muscarinic cholinergic receptor, triggering the inhibition of SDH neuronal excitability via CaMKII signaling pathway, thereby resulting in antagonistic effects in the CIBP rat model.
Collapse
Affiliation(s)
- Longsheng Xu
- Department of Anesthesia and Pain Medicine, Affiliated Hospital of Jiaxing University, Jiaxing 314000, China
| | - Shizhen Wang
- Department of Basic Medicine, Jiangsu Vocational College of Nursing, Huaian 223001, China
| | - Ling Zhang
- Department of central laboratory, Affiliated Zhangjiagang Hospital of Suzhou University, Zhangjiagang 215600, China
| | - Beibei Liu
- Department of Anesthesia and Pain Medicine, Affiliated Hospital of Jiaxing University, Jiaxing 314000, China
| | - Shang Zheng
- Department of Anesthesia and Pain Medicine, Affiliated Hospital of Jiaxing University, Jiaxing 314000, China
| | - Ming Yao
- Department of Anesthesia and Pain Medicine, Affiliated Hospital of Jiaxing University, Jiaxing 314000, China
| |
Collapse
|
11
|
System Xc− inhibition blocks bone marrow-multiple myeloma exosomal crosstalk, thereby countering bortezomib resistance. Cancer Lett 2022; 535:215649. [DOI: 10.1016/j.canlet.2022.215649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 02/28/2022] [Accepted: 03/15/2022] [Indexed: 12/27/2022]
|
12
|
Squillace S, Salvemini D. Nitroxidative stress in pain and opioid-induced adverse effects: therapeutic opportunities. Pain 2022; 163:205-213. [PMID: 34145168 DOI: 10.1097/j.pain.0000000000002347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/17/2021] [Indexed: 11/25/2022]
Affiliation(s)
- Silvia Squillace
- Department of Pharmacology and Physiology, Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, MO, United States
| | | |
Collapse
|
13
|
Abstract
Recent decades have demonstrated significant strides in cancer screening, diagnostics and therapeutics. As such there have been dramatic changes in survival following a diagnosis of cancer.
Collapse
Affiliation(s)
- Matthew R D Brown
- The Royal Marsden NHS Foundation Trust, Fulham Road, London, SW3 6JJ, UK
| | | | - David J Magee
- The Royal Marsden Hospital, Fulham Road, London, SW3 6JJ, UK.
| |
Collapse
|
14
|
Abstract
PURPOSE OF REVIEW The treatment of cancer-induced bone pain (CIBP) has been proven ineffective and relies heavily on opioids, the target of highly visible criticism for their negative side effects. Alternative therapeutic agents are needed and the last few years have brought promising results, detailed in this review. RECENT FINDINGS Cysteine/glutamate antiporter system, xc, cannabinoids, kappa opioids, and a ceramide axis have all been shown to have potential as novel therapeutic targets without the negative effects of opioids. SUMMARY Review of the most recent and promising studies involving CIBP, specifically within murine models. Cancer pain has been reported by 30-50% of all cancer patients and even more in late stages, however the standard of care is not effective to treat CIBP. The complicated and chronic nature of this type of pain response renders over the counter analgesics and opioids largely ineffective as well as difficult to use due to unwanted side effects. Preclinical studies have been standardized and replicated while novel treatments have been explored utilizing various alternative receptor pathways: cysteine/glutamate antiporter system, xc, cannabinoid type 1 receptor, kappa opioids, and a ceramide axis sphingosine-1-phosphate/sphingosine-1-phosphate receptor 1.
Collapse
|
15
|
Doyle TM, Braden K, Harada CM, Mufti F, Schafer RM, Salvemini D. Novel Non-Opioid Based Therapeutics for Chronic Neuropathic Pain. MISSOURI MEDICINE 2021; 118:327-333. [PMID: 34373667 PMCID: PMC8343628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Chronic neuropathic pain is currently a major health issue in U.S. complicated by the lack of non-opioid analgesic alternatives. Our investigations led to the discovery of major signaling pathways involved in the transition of acute to chronic neuropathic pain and the identification of several targets for therapeutic intervention. Our translational approach has facilitated the advancement of novel medicines for chronic neuropathic pain that are in advanced clinical development and clinical trials.
Collapse
Affiliation(s)
- Timothy M Doyle
- Department of Pharmacology and Physiology and the Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Kathryn Braden
- Department of Pharmacology and Physiology and the Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Caron M Harada
- Department of Pharmacology and Physiology and the Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Fatma Mufti
- Department of Pharmacology and Physiology and the Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Rachel M Schafer
- Department of Pharmacology and Physiology and the Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Daniela Salvemini
- Department of Pharmacology and Physiology and the Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri
| |
Collapse
|
16
|
Pineda-Farias JB, Saloman JL, Scheff NN. Animal Models of Cancer-Related Pain: Current Perspectives in Translation. Front Pharmacol 2021; 11:610894. [PMID: 33381048 PMCID: PMC7768910 DOI: 10.3389/fphar.2020.610894] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 10/30/2020] [Indexed: 01/15/2023] Open
Abstract
The incidence of pain in cancer patients during diagnosis and treatment is exceedingly high. Although advances in cancer detection and therapy have improved patient prognosis, cancer and its treatment-associated pain have gained clinical prominence. The biological mechanisms involved in cancer-related pain are multifactorial; different processes for pain may be responsible depending on the type and anatomic location of cancer. Animal models of cancer-related pain have provided mechanistic insights into the development and process of pain under a dynamic molecular environment. However, while cancer-evoked nociceptive responses in animals reflect some of the patients’ symptoms, the current models have failed to address the complexity of interactions within the natural disease state. Although there has been a recent convergence of the investigation of carcinogenesis and pain neurobiology, identification of new targets for novel therapies to treat cancer-related pain requires standardization of methodologies within the cancer pain field as well as across disciplines. Limited success of translation from preclinical studies to the clinic may be due to our poor understanding of the crosstalk between cancer cells and their microenvironment (e.g., sensory neurons, infiltrating immune cells, stromal cells etc.). This relatively new line of inquiry also highlights the broader limitations in translatability and interpretation of basic cancer pain research. The goal of this review is to summarize recent findings in cancer pain based on preclinical animal models, discuss the translational benefit of these discoveries, and propose considerations for future translational models of cancer pain.
Collapse
Affiliation(s)
- Jorge B Pineda-Farias
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jami L Saloman
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Nicole N Scheff
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Hillman Cancer Center, University of Pittsburgh Medicine Center, Pittsburgh, PA, United States
| |
Collapse
|
17
|
Lin W, Wang C, Liu G, Bi C, Wang X, Zhou Q, Jin H. SLC7A11/xCT in cancer: biological functions and therapeutic implications. Am J Cancer Res 2020; 10:3106-3126. [PMID: 33163260 PMCID: PMC7642655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/01/2020] [Indexed: 06/11/2023] Open
Abstract
Amino acid transporters mediate substrates across cellular membranes and their fine-tuned regulations are critical to cellular metabolism, growth, and death. As the functional component of system Xc-, which imports extracellular cystine with intracellular glutamate release at a ratio of 1:1, SLC7A11 has diverse functional roles in regulating many pathophysiological processes such as cellular redox homeostasis, ferroptosis, and drug resistance in cancer. Notably, accumulated evidence demonstrated that SLC7A11 is overexpressed in many types of cancers and is associated with patients' poor prognosis. As a result, SLC7A11 becomes a new potential target for cancer therapy. In this review, we first briefly introduce the structure and function of SLC7A11, then discuss its pathological role in cancer. We next summarize current available data of how SLC7A11 is subjected to fine regulations at multiple levels. We further describe the potential inhibitors of the SLC7A11 and their roles in human cancer cells. Finally, we propose novel insights for future perspectives on the modulation of SLC7A11, as well as possible targeted strategies for SLC7A11-based anti-cancer therapies.
Collapse
Affiliation(s)
- Wenyu Lin
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Zhejiang University School of MedicineHangzhou 310016, Zhejiang, China
| | - Chaoqun Wang
- Department of Pathology, Affiliated Dongyang Hospital of Wenzhou Medical UniversityDongyang 322100, Zhejiang, China
| | - Guangping Liu
- College of Life Sciences, Yan’an UniversityYan’an 716000, Shaanxi, China
| | - Chao Bi
- Institute of Translational Medicine, Zhejiang University School of MedicineHangzhou 310029, Zhejiang, China
| | - Xian Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of MedicineHangzhou 310016, Zhejiang, China
| | - Qiyin Zhou
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of MedicineHangzhou 310016, Zhejiang, China
| | - Hongchuan Jin
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Zhejiang University School of MedicineHangzhou 310016, Zhejiang, China
| |
Collapse
|
18
|
Thompson AL, Grenald SA, Ciccone HA, BassiriRad N, Niphakis MJ, Cravatt BF, Largent-Milnes TM, Vanderah TW. The Endocannabinoid System Alleviates Pain in a Murine Model of Cancer-Induced Bone Pain. J Pharmacol Exp Ther 2020; 373:230-238. [PMID: 32054717 PMCID: PMC7160863 DOI: 10.1124/jpet.119.262337] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 02/10/2020] [Indexed: 12/22/2022] Open
Abstract
Metastatic breast cancer is prevalent worldwide, and one of the most common sites of metastasis is long bones. Of patients with disease, the major symptom is pain, yet current medications fail to adequately result in analgesic efficacy and present major undesirable adverse effects. In our study, we investigate the potential of a novel monoacylglycerol lipase (MAGL) inhibitor, MJN110, in a murine model of cancer-induced bone pain. Literature has previously demonstrated that MAGL inhibitors function to increase the endogenous concentrations of 2-arachydonylglycerol, which then activates CB1 and CB2 receptors to inhibit inflammation and pain. We demonstrate that administration of MJN110 significantly and dose dependently alleviates spontaneous pain behavior during acute administration compared with vehicle control. In addition, MJN110 maintains its efficacy in a chronic-dosing paradigm over the course of 7 days without signs of receptor sensitization. In vitro analysis of MJN110 demonstrated a dose-dependent and significant decrease in cell viability and proliferation of 66.1 breast adenocarcinoma cells to a greater extent than KML29, an alternate MAGL inhibitor, or the CB2 agonist JWH015. Chronic administration of the compound did not appear to affect tumor burden, as evidenced by radiograph or histologic analysis. Together, these data support the application for MJN110 as a novel therapeutic for cancer-induced bone pain. SIGNIFICANCE STATEMENT: Current standard of care for metastatic breast cancer pain is opioid-based therapies with adjunctive chemotherapy, which have highly addictive and other deleterious side effects. The need for effective, non-opioid-based therapies is essential, and harnessing the endogenous cannabinoid system is proving to be a new target to treat various types of pain conditions. We present a novel drug targeting the endogenous cannabinoid system that is effective at reducing pain in a mouse model of metastatic breast cancer to bone.
Collapse
Affiliation(s)
- A L Thompson
- Department of Medical Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona (A.L.T., S.A.G., H.A.C., N.B., T.M.L.-M., T.W.V); Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland (S.A.G.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - S A Grenald
- Department of Medical Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona (A.L.T., S.A.G., H.A.C., N.B., T.M.L.-M., T.W.V); Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland (S.A.G.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - H A Ciccone
- Department of Medical Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona (A.L.T., S.A.G., H.A.C., N.B., T.M.L.-M., T.W.V); Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland (S.A.G.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - N BassiriRad
- Department of Medical Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona (A.L.T., S.A.G., H.A.C., N.B., T.M.L.-M., T.W.V); Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland (S.A.G.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - M J Niphakis
- Department of Medical Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona (A.L.T., S.A.G., H.A.C., N.B., T.M.L.-M., T.W.V); Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland (S.A.G.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - B F Cravatt
- Department of Medical Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona (A.L.T., S.A.G., H.A.C., N.B., T.M.L.-M., T.W.V); Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland (S.A.G.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - T M Largent-Milnes
- Department of Medical Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona (A.L.T., S.A.G., H.A.C., N.B., T.M.L.-M., T.W.V); Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland (S.A.G.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| | - T W Vanderah
- Department of Medical Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona (A.L.T., S.A.G., H.A.C., N.B., T.M.L.-M., T.W.V); Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland (S.A.G.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California (M.J.N., B.F.C.)
| |
Collapse
|
19
|
de Almeida AS, Rigo FK, De Prá SDT, Milioli AM, Pereira GC, Lückemeyer DD, Antoniazzi CT, Kudsi SQ, Araújo DMPA, Oliveira SM, Ferreira J, Trevisan G. Role of transient receptor potential ankyrin 1 (TRPA1) on nociception caused by a murine model of breast carcinoma. Pharmacol Res 2020; 152:104576. [DOI: 10.1016/j.phrs.2019.104576] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/28/2022]
|
20
|
Zajączkowska R, Kocot-Kępska M, Leppert W, Wordliczek J. Bone Pain in Cancer Patients: Mechanisms and Current Treatment. Int J Mol Sci 2019; 20:E6047. [PMID: 31801267 PMCID: PMC6928918 DOI: 10.3390/ijms20236047] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/17/2019] [Accepted: 11/28/2019] [Indexed: 02/06/2023] Open
Abstract
The skeletal system is the third most common site for cancer metastases, surpassed only by the lungs and liver. Many tumors, especially those of the breast, prostate, lungs, and kidneys, have a strong predilection to metastasize to bone, which causes pain, hypercalcemia, pathological skeletal fractures, compression of the spinal cord or other nervous structures, decreased mobility, and increased mortality. Metastatic cancer-induced bone pain (CIBP) is a type of chronic pain with unique and complex pathophysiology characterized by nociceptive and neuropathic components. Its treatment should be multimodal (pharmacological and non-pharmacological), including causal anticancer and symptomatic analgesic treatment to improve quality of life (QoL). The aim of this paper is to discuss the mechanisms involved in the occurrence and persistence of cancer-associated bone pain and to review the treatment methods recommended by experts in clinical practice. The final part of the paper reviews experimental therapeutic methods that are currently being studied and that may improve the efficacy of bone pain treatment in cancer patients in the future.
Collapse
Affiliation(s)
- Renata Zajączkowska
- Department of Interdisciplinary Intensive Care, Jagiellonian University Medical College, 31-008 Krakow, Poland;
| | - Magdalena Kocot-Kępska
- Department of Pain Research and Treatment, Jagiellonian University Medical College, 31-008 Krakow, Poland
| | - Wojciech Leppert
- Laboratory of Quality of Life Research, Chair and Department of Palliative Medicine, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| | - Jerzy Wordliczek
- Department of Interdisciplinary Intensive Care, Jagiellonian University Medical College, 31-008 Krakow, Poland;
| |
Collapse
|
21
|
He Q, Wang T, Ni H, Liu Q, An K, Tao J, Chen Y, Xu L, Zhu C, Yao M. Endoplasmic reticulum stress promoting caspase signaling pathway-dependent apoptosis contributes to bone cancer pain in the spinal dorsal horn. Mol Pain 2019; 15:1744806919876150. [PMID: 31452457 PMCID: PMC6767730 DOI: 10.1177/1744806919876150] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Qiuli He
- Department of Anesthesiology, Bengbu Medical College, Bengbu, China.,Department of Anesthesiology and Pain Research Center, The First Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Tingting Wang
- Department of Anesthesiology and Pain Research Center, The First Affiliated Hospital of Jiaxing University, Jiaxing, China.,The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Huadong Ni
- Department of Anesthesiology and Pain Research Center, The First Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Qianying Liu
- Department of Anesthesiology, Bengbu Medical College, Bengbu, China
| | - Kang An
- Department of Anesthesiology, Affliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, China
| | - Jiachun Tao
- Department of Anesthesiology and Pain Research Center, The First Affiliated Hospital of Jiaxing University, Jiaxing, China.,The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yajing Chen
- Department of Anesthesiology, Bengbu Medical College, Bengbu, China
| | - Longsheng Xu
- Department of Anesthesiology and Pain Research Center, The First Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Chunyan Zhu
- Department of Anesthesiology and Pain Research Center, The First Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Ming Yao
- Department of Anesthesiology and Pain Research Center, The First Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
22
|
de Almeida AS, Rigo FK, De Prá SDT, Milioli AM, Dalenogare DP, Pereira GC, Ritter CDS, Peres DS, Antoniazzi CTDD, Stein C, Moresco RN, Oliveira SM, Trevisan G. Characterization of Cancer-Induced Nociception in a Murine Model of Breast Carcinoma. Cell Mol Neurobiol 2019; 39:605-617. [PMID: 30850915 PMCID: PMC11462836 DOI: 10.1007/s10571-019-00666-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 02/25/2019] [Indexed: 12/14/2022]
Abstract
Severe and poorly treated pain often accompanies breast cancer. Thus, novel mechanisms involved in breast cancer-induced pain should be investigated. Then, it is necessary to characterize animal models that are reliable with the symptoms and progression of the disease as observed in humans. Explaining cancer-induced nociception in a murine model of breast carcinoma was the aim of this study. 4T1 (104) lineage cells were inoculated in the right fourth mammary fat pad of female BALB/c mice; after this, mechanical and cold allodynia, or mouse grimace scale (MGS) were observed for 30 days. To determine the presence of bone metastasis, we performed the metastatic clonogenic test and measure calcium serum levels. At 20 days after tumor induction, the antinociceptive effect of analgesics used to relieve pain in cancer patients (acetaminophen, naproxen, codeine or morphine) or a cannabinoid agonist (WIN 55,212-2) was tested. Mice inoculated with 4T1 cells developed mechanical and cold allodynia and increased MGS. Bone metastasis was confirmed using the clonogenic assay, and hypercalcemia was observed 20 days after cells inoculation. All analgesic drugs reduced the mechanical and cold allodynia, while the MGS was decreased only by the administration of naproxen, codeine, or morphine. Also, WIN 55,212-2 improved all nociceptive measures. This pain model could be a reliable form to observe the mechanisms of breast cancer-induced pain or to observe the efficacy of novel analgesic compounds.
Collapse
Affiliation(s)
- Amanda Spring de Almeida
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Flávia Karine Rigo
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (Unesc), Criciúma, SC, 88006-000, Brazil
| | - Samira Dal-Toé De Prá
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (Unesc), Criciúma, SC, 88006-000, Brazil
| | - Alessandra Marcone Milioli
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (Unesc), Criciúma, SC, 88006-000, Brazil
| | - Diéssica Padilha Dalenogare
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Gabriele Cheiran Pereira
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Camila Dos Santos Ritter
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Diulle Spat Peres
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | | | - Carolina Stein
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Rafael Noal Moresco
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Sara Marchesan Oliveira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Gabriela Trevisan
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil.
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense (Unesc), Criciúma, SC, 88006-000, Brazil.
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Avenida Roraima, 1000, Building 21, Room 5207, Santa Maria, RS, 97105-900, Brazil.
| |
Collapse
|
23
|
Spinal microglia contribute to cancer-induced pain through system x C --mediated glutamate release. Pain Rep 2019; 4:e738. [PMID: 31583353 PMCID: PMC6749914 DOI: 10.1097/pr9.0000000000000738] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 02/26/2019] [Accepted: 02/27/2019] [Indexed: 12/13/2022] Open
Abstract
Introduction: Microglial cells, the resident macrophages of the central nervous system, are a key contributor to the generation and maintenance of cancer-induced pain (CIP). In healthy organisms, activated microglia promote recovery through the release of trophic and anti-inflammatory factors to clear toxins and pathogens and support neuronal survival. Chronically activated microglia, however, release toxic substances, including excess glutamate, causing cytotoxicity. Accordingly, rising attention is given to microglia for their role in abnormal physiology and in mediating neurotoxicity. Objectives: To examine the nociceptive relationship between peripherally-released glutamate and microglial xCT. Methods: A validated murine model of 4T1 carcinoma cell–induced nociception was used to assess the effect of peripheral tumour on spinal microglial activation and xCT expression. Coculture systems were then used to investigate the direct effect of glutamate released by wildtype and xCT knockdown MDA-MB-231 carcinoma cells on microglial activation, functional system xC− activity, and protein levels of interferon regulatory factor 8 (IRF8), a transcription factor implicated in microglia-mediated nociception. Results: Blockade of system xC− with sulfasalazine (SSZ) in vivo attenuated nociception in a 4T1 murine model of CIP and attenuates tumour-induced microglial activation in the dorsal horn of the spinal cord. Furthermore, knockdown of xCT in MDA-MB-231 cells mitigated tumour cell–induced microglial activation and functional system xC− activity in vitro. Conclusions: These data collectively demonstrate that the system xCT antiporter is functionally implicated in CIP and may be particularly relevant to pain progression through microglia. Upregulated xCT in chronically activated spinal microglia may be one pathway to central glutamate cytotoxicity. Microglial xCT may therefore be a valuable target for mitigating CIP.
Collapse
|
24
|
Ungard RG, Linher-Melville K, Nashed MG, Sharma M, Wen J, Singh G. xCT knockdown in human breast cancer cells delays onset of cancer-induced bone pain. Mol Pain 2019; 15:1744806918822185. [PMID: 30799686 PMCID: PMC6329019 DOI: 10.1177/1744806918822185] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cancers in the bone produce a number of severe symptoms including pain that compromises patient functional status, quality of life, and survival. The source of this pain is multifaceted and includes factors secreted from tumor cells. Malignant cells release the neurotransmitter and cell-signaling molecule glutamate via the oxidative stress-related cystine/glutamate antiporter, system xC-, which reciprocally imports cystine for synthesis of glutathione and the cystine/cysteine redox cycle. Pharmacological inhibition of system xC- has shown success in reducing and delaying the onset of cancer pain-related behavior in mouse models. This investigation describes the development of a stable siRNA-induced knockdown of the functional trans-membrane system xC- subunit xCT ( SLC7A11) in the human breast cancer cell line MDA-MB-231. Clones were verified for xCT knockdown at the transcript, protein, and functional levels. RNAseq was performed on a representative clone to comprehensively examine the transcriptional cellular signature in response to xCT knockdown, identifying multiple differentially regulated factors relevant to cancer pain including nerve growth factor, interleukin-1, and colony-stimulating factor-1. Mice were inoculated intrafemorally and recordings of pain-related behaviors including weight bearing, mechanical withdrawal, and limb use were performed. Animals implanted with xCT knockdown cancer cells displayed a delay until the onset of nociceptive behaviors relative to control cells. These results add to the body of evidence suggesting that a reduction in glutamate release from cancers in bone by inhibition of the system xC- transporter may decrease the severe and intractable pain associated with bone metastases.
Collapse
Affiliation(s)
- Robert G Ungard
- 1 Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada.,2 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Katja Linher-Melville
- 1 Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada.,2 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Mina G. Nashed
- 1 Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada.,2 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Manu Sharma
- 1 Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada.,2 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jianping Wen
- 2 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Gurmit Singh
- 1 Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada.,2 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
25
|
Linher-Melville K, Sharma M, Nakhla P, Kum E, Ungard R, Park J, Rosa D, Gunning P, Singh G. Inhibiting STAT3 in a murine model of human breast cancer-induced bone pain delays the onset of nociception. Mol Pain 2019; 15:1744806918823477. [PMID: 30799695 PMCID: PMC6329039 DOI: 10.1177/1744806918823477] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aggressive breast cancer subtypes utilize system xc-, a membrane antiporter, to import cystine for glutathione synthesis and maintenance of redox homeostasis, in turn releasing glutamate as a metabolic pro-nociceptive by-product. Metastatic breast cancers establish themselves at distal sites including bone, where changes in extracellular glutamate levels contribute to cancer-induced bone pain. We previously established that stearically blocking system xc- activity with sulfasalazine delays the onset of nociceptive behaviours and that xCT, the functional antiporter subunit, is positively regulated by signal transducer and activator of transcription 3 (STAT3). In the current investigation, a murine xenograft cancer-induced bone pain model was applied to examine whether pharmacological inhibition of phosphorylated STAT3 (pSTAT3) induces changes in nociception. A high glutamate-releasing, xCT/pSTAT3 over-expressing human breast cancer cell line was selected for injection into the distal epiphysis of the right femur of female nude mice. A 14-day regimen of intraperitoneal injections with either vehicle or the novel STAT3 inhibitor DR-1-55 commenced three weeks after initial intrafemoral bone injection. Nociceptive behaviours were temporally monitored by automated von Frey, dynamic weight bearing and open-field testing for the duration of the study, beginning at the baseline. Prior to sacrifice and at ethical end point, tumour-induced osteolytic lesions were radiographically assessed. Treatment with DR-1-55 significantly delayed the onset and severity of spontaneous and induced nociceptive behaviours, also decreasing human SLC7A11 ( xCT) mRNA levels in tumour-bearing limbs without altering osteolysis. In addition, two pro-inflammatory cytokines released by this cell line, interleukin 6 and interleukin 1β, were also down-regulated at the mRNA level in response to DR-1-55 treatment in vivo, with lower human interleukin 6 levels detected in the host circulation. This study demonstrates that targeting pSTAT3 may be a viable therapeutic means to manage cancer-induced bone pain, alone or in combination with stearic system xc- blockers.
Collapse
Affiliation(s)
- Katja Linher-Melville
- 1 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Manu Sharma
- 1 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Peter Nakhla
- 1 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Elena Kum
- 1 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Robert Ungard
- 1 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Ji Park
- 2 Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - David Rosa
- 2 Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Patrick Gunning
- 2 Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Gurmit Singh
- 1 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
26
|
Miladinovic T, Ungard RG, Linher-Melville K, Popovic S, Singh G. Functional effects of TrkA inhibition on system x C--mediated glutamate release and cancer-induced bone pain. Mol Pain 2018; 14:1744806918776467. [PMID: 29761734 PMCID: PMC5956640 DOI: 10.1177/1744806918776467] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Breast cancer cells release the signalling molecule glutamate via the system xC− antiporter, which is upregulated to exchange extracellular cystine for intracellular glutamate to protect against oxidative stress. Here, we demonstrate that this antiporter is functionally influenced by the actions of the neurotrophin nerve growth factor on its cognate receptor tyrosine kinase, TrkA, and that inhibiting this complex may reduce cancer-induced bone pain via its downstream actions on xCT, the functional subunit of system xC−. We have characterized the effects of the selective TrkA inhibitor AG879 on system xC− activity in murine 4T1 and human MDA-MB-231 mammary carcinoma cells, as well as its effects on nociception in our validated immunocompetent mouse model of cancer-induced bone pain, in which BALB/c mice are intrafemorally inoculated with 4T1 murine carcinoma cells. AG879 decreased functional system xC− activity, as measured by cystine uptake and glutamate release, and inhibited nociceptive and physiologically relevant responses in tumour-bearing animals. Cumulatively, these data suggest that the activation of TrkA by nerve growth factor may have functional implications on system xC−-mediated cancer pain. System xC−-mediated TrkA activation therefore presents a promising target for therapeutic intervention in cancer pain treatment.
Collapse
Affiliation(s)
- Tanya Miladinovic
- 1 Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada.,2 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Robert G Ungard
- 1 Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada.,2 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Katja Linher-Melville
- 1 Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada.,2 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Snezana Popovic
- 2 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Gurmit Singh
- 1 Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada.,2 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
27
|
Li X, Li C, Tang Y, Huang Y, Cheng Q, Huang X, Zhao F, Hao C, Feng D, Xu J, Han J, Tang S, Liu W, Yue S, Luo Z. NMDA receptor activation inhibits the antifibrotic effect of BM-MSCs on bleomycin-induced pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2018; 315:L404-L421. [PMID: 29722562 PMCID: PMC6172623 DOI: 10.1152/ajplung.00002.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Endogenous glutamate (Glu) release and N-methyl-d-aspartate (NMDA) receptor (NMDAR) activation are associated with lung injury in different animal models. However, the underlying mechanism is unclear. Bone marrow-derived mesenchymal stem cells (BM-MSCs), which show potential use for immunomodulation and tissue protection, play a protective role in pulmonary fibrosis (PF) process. Here, we found the increased Glu release from the BM cells of bleomycin (BLM)-induced PF mice in vivo. BLM stimulation also increased the extracellular Glu in BM-MSCs via the antiporter system xc− in vitro. The gene expression of each subunit of NMDAR was detected in BM-MSCs. NMDAR activation inhibited the proliferation, migration, and paracrine function of BM-MSCs in vitro. BM-MSCs were derived from male C57BL/6 mice, transfected with lentiviral vectors carrying the enhanced green fluorescence protein gene, pretreated with NMDA, and transplanted into the female recipient mice that were intratracheally injected with BLM to induce PF. Transplantation of NMDA-pretreated BM-MSCs significantly aggravated PF as compared with that in the normal BM-MSCs transplantation group. The sex determination gene Y chromosome and green fluorescence protein genes of BM-MSCs were detected to observe BM-MSCs homing in the fibrotic lungs. Moreover, NMDAR activation inhibited BM-MSC migration by downregulating the stromal cell-derived factor-1/C-X-C chemokine receptor type 4 signaling axis. NMDAR activation aggravated the transforming growth factor-β1-induced extracellular matrix production in alveolar epithelial cells and fibroblasts through the paracrine effects of BM-MSCs. In summary, these findings suggested that NMDAR activation-mediated Glu excitotoxicity induced by BLM in BM-MSCs abolished the therapeutic effects of normal BM-MSCs transplantation on BLM-induced PF.
Collapse
Affiliation(s)
- Xiaohong Li
- Department of Physiology, Xiangya School of Medicine, Central South University , Changsha, Hunan , China
| | - Chen Li
- Department of Physiology, Changzhi Medical College, Changzhi, Shanxi , China
| | - Yiting Tang
- Department of Physiology, Xiangya School of Medicine, Central South University , Changsha, Hunan , China
| | - Yanhong Huang
- Department of Physiology, Xiangya School of Medicine, Central South University , Changsha, Hunan , China
| | - Qingmei Cheng
- Department of Physiology, Xiangya School of Medicine, Central South University , Changsha, Hunan , China
| | - Xiaoting Huang
- Department of Physiology, Xiangya School of Medicine, Central South University , Changsha, Hunan , China
| | - Feiyan Zhao
- Department of Physiology, Xiangya School of Medicine, Central South University , Changsha, Hunan , China
| | - Caixia Hao
- Department of Physiology, Xiangya School of Medicine, Central South University , Changsha, Hunan , China
| | - Dandan Feng
- Department of Physiology, Xiangya School of Medicine, Central South University , Changsha, Hunan , China
| | - Jianping Xu
- Department of Physiology, Xiangya School of Medicine, Central South University , Changsha, Hunan , China
| | - Jianzhong Han
- Department of Physiology, Xiangya School of Medicine, Central South University , Changsha, Hunan , China
| | - Siyuan Tang
- Xiangya Nursing School, Central South University , Changsha, Hunan , China
| | - Wei Liu
- Xiangya Nursing School, Central South University , Changsha, Hunan , China
| | - Shaojie Yue
- Department of Pediatrics, Xiangya Hospital, Central South University , Changsha, Hunan , China
| | - Ziqiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University , Changsha, Hunan , China
| |
Collapse
|
28
|
Kong X, Wei J, Wang D, Zhu X, Zhou Y, Wang S, Xu GY, Jiang GQ. Upregulation of Spinal Voltage-Dependent Anion Channel 1 Contributes to Bone Cancer Pain Hypersensitivity in Rats. Neurosci Bull 2017; 33:711-721. [PMID: 29196874 DOI: 10.1007/s12264-017-0195-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 11/08/2017] [Indexed: 01/02/2023] Open
Abstract
Voltage-dependent anion channel 1 (VDAC1) is thought to contribute to the progression of tumor development. However, whether VDAC1 contributes to bone cancer pain remains unknown. In this study, we found that the expression of VDAC1 was upregulated in the L2-5 segments of the spinal dorsal horn at 2 and 3 weeks after injection of tumor cells into the tibial cavity. Intrathecal injection of a VDAC1 inhibitor significantly reversed the pain hypersensitivity and reduced the over-expression of Toll-like receptor 4 (TLR4). Intrathecal injection of minocycline, an inhibitor of microglia, also attenuated the pain hypersensitivity of rat models of bone cancer pain. These results suggest that VDAC1 plays a significant role in the development of complicated cancer pain, possibly by regulating the expression of TLR4.
Collapse
Affiliation(s)
- Xiangpeng Kong
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psychiatric Diseases and Institute of Neuroscience, The Second Affiliated Hospital, Soochow University, Suzhou, 215123, China
| | - Jinrong Wei
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psychiatric Diseases and Institute of Neuroscience, The Second Affiliated Hospital, Soochow University, Suzhou, 215123, China
| | - Diyu Wang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psychiatric Diseases and Institute of Neuroscience, The Second Affiliated Hospital, Soochow University, Suzhou, 215123, China
| | - Xiaoju Zhu
- Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, 215600, China
| | - Youlang Zhou
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psychiatric Diseases and Institute of Neuroscience, The Second Affiliated Hospital, Soochow University, Suzhou, 215123, China
| | - Shusheng Wang
- Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, 215600, China
| | - Guang-Yin Xu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psychiatric Diseases and Institute of Neuroscience, The Second Affiliated Hospital, Soochow University, Suzhou, 215123, China.
- Center for Translational Medicine, Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, 215600, China.
| | - Guo-Qin Jiang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psychiatric Diseases and Institute of Neuroscience, The Second Affiliated Hospital, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
29
|
Abstract
Many cancerous solid tumors metastasize to the bone and induce pain (cancer-induced bone pain [CIBP]). Cancer-induced bone pain is often severe because of enhanced inflammation, rapid bone degradation, and disease progression. Opioids are prescribed to manage this pain, but they may enhance bone loss and increase tumor proliferation, further compromising patient quality of life. Angiotensin-(1-7) (Ang-(1-7)) binds and activates the Mas receptor (MasR). Angiotensin-(1-7)/MasR activation modulates inflammatory signaling after acute tissue insult, yet no studies have investigated whether Ang-(1-7)/MasR play a role in CIBP. We hypothesized that Ang-(1-7) inhibits CIBP by targeting MasR in a murine model of breast CIBP. 66.1 breast cancer cells were implanted into the femur of BALB/cAnNHsd mice as a model of CIBP. Spontaneous and evoked pain behaviors were assessed before and after acute and chronic administration of Ang-(1-7). Tissues were collected from animals for ex vivo analyses of MasR expression, tumor burden, and bone integrity. Cancer inoculation increased spontaneous pain behaviors by day 7 that were significantly reduced after a single injection of Ang-(1-7) and after sustained administration. Preadministration of A-779 a selective MasR antagonist prevented this reduction, whereas pretreatment with the AT2 antagonist had no effect; an AT1 antagonist enhanced the antinociceptive activity of Ang-(1-7) in CIBP. Repeated Ang-(1-7) administration did not significantly change tumor burden or bone remodeling. Data here suggest that Ang-(1-7)/MasR activation significantly attenuates CIBP, while lacking many side effects seen with opioids. Thus, Ang-(1-7) may be an alternative therapeutic strategy for the nearly 90% of patients with advanced-stage cancer who experience excruciating pain.
Collapse
|
30
|
Brown M, Farquhar-Smith P. Pain in cancer survivors; filling in the gaps. Br J Anaesth 2017; 119:723-736. [DOI: 10.1093/bja/aex202] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
31
|
Fazzari J, Balenko MD, Zacal N, Singh G. Identification of capsazepine as a novel inhibitor of system x c- and cancer-induced bone pain. J Pain Res 2017; 10:915-925. [PMID: 28458574 PMCID: PMC5402992 DOI: 10.2147/jpr.s125045] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The cystine/glutamate antiporter has been implicated in a variety of cancers as a major mediator of redox homeostasis. The excess glutamate secreted by this transporter in aggressive cancer cells has been associated with cancer-induced bone pain (CIBP) from distal breast cancer metastases. High-throughput screening of small molecule inhibitors of glutamate release from breast cancer cells identified several potential compounds. One such compound, capsazepine (CPZ), was confirmed to inhibit the functional unit of system xc- (xCT) through its ability to block uptake of its radiolabeled substrate, cystine. Blockade of this antiporter induced production of reactive oxygen species (ROS) within 4 hours and induced cell death within 48 hours at concentrations exceeding 25 μM. Furthermore, cell death and ROS production were significantly reduced by co-treatment with N-acetylcysteine, suggesting that CPZ toxicity is associated with ROS-induced cell death. These data suggest that CPZ can modulate system xc- activity in vitro and this translates into antinociception in an in vivo model of CIBP where systemic administration of CPZ successfully delayed the onset and reversed CIBP-induced nociceptive behaviors resulting from intrafemoral MDA-MB-231 tumors.
Collapse
Affiliation(s)
- Jennifer Fazzari
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Matthew D Balenko
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Natalie Zacal
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Gurmit Singh
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
32
|
Yao P, Ding Y, Han Z, Mu Y, Hong T, Zhu Y, Li H. Suppression of asparaginyl endopeptidase attenuates breast cancer-induced bone pain through inhibition of neurotrophin receptors. Mol Pain 2017; 13:1744806917708127. [PMID: 28554249 PMCID: PMC5453632 DOI: 10.1177/1744806917708127] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 03/03/2017] [Accepted: 04/06/2017] [Indexed: 01/08/2023] Open
Abstract
Objective Cancer-induced bone pain is a common clinical problem in breast cancer patients with bone metastasis. However, the mechanisms driving cancer-induced bone pain are poorly known. Recent studies show that a novel protease, asparaginyl endopeptidase (AEP) plays crucial roles in breast cancer metastasis and progression. We aim to determine the functions and targeted suppress of AEP in a mouse model of breast cancer-induced bone pain. Methods Breast cancer cells with AEP knocked-down or overexpression were constructed and implanted into the intramedullary space of the femur to induce pain-like behavior in mice. AEP-specific inhibitors or purified AEP proteins were further used in animal model. The histological characters of femur and pain ethological changes were measured. The expressions of AEP and neurotrophin receptors (p75NTR and TrkA) in dorsal root ganglion and spinal cord were examined. Results Femur radiographs and histological analysis revealed that cells with AEP knocked-down reduced bone destruction and pain behaviors. However, cells with AEP overexpression elevated bone damage and pain behaviors. Further, Western blot results found that the expressions of p75NTR and TrkA in dorsal root ganglions and spinal cords were reduced in mice inoculated with AEP knocked-down cells. Targeted suppression of AEP with specific small compounds significantly reduced the bone pain while purified recombinant AEP proteins increased bone pain. Conclusions AEP aggravate the development of breast cancer bone metastasis and bone pain by increasing the expression of neurotrophin receptors. AEP might be an effective target for treatment of breast cancerinduced bone pain.
Collapse
Affiliation(s)
- Peng Yao
- Department of Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuanyuan Ding
- Department of Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhenkai Han
- Department of Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ying Mu
- Department of Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tao Hong
- Department of Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yongqiang Zhu
- Department of Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hongxi Li
- Department of Pain Management, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
33
|
Grace PM, Gaudet AD, Staikopoulos V, Maier SF, Hutchinson MR, Salvemini D, Watkins LR. Nitroxidative Signaling Mechanisms in Pathological Pain. Trends Neurosci 2016; 39:862-879. [PMID: 27842920 PMCID: PMC5148691 DOI: 10.1016/j.tins.2016.10.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 10/18/2016] [Accepted: 10/20/2016] [Indexed: 12/14/2022]
Abstract
Tissue injury can initiate bidirectional signaling between neurons, glia, and immune cells that creates and amplifies pain. While the ability for neurotransmitters, neuropeptides, and cytokines to initiate and maintain pain has been extensively studied, recent work has identified a key role for reactive oxygen and nitrogen species (ROS/RNS; nitroxidative species), including superoxide, peroxynitrite, and hydrogen peroxide. In this review we describe how nitroxidative species are generated after tissue injury and the mechanisms by which they enhance neuroexcitability in pain pathways. Finally, we discuss potential therapeutic strategies for normalizing nitroxidative signaling, which may also enhance opioid analgesia, to help to alleviate the enormous burden of pathological pain.
Collapse
Affiliation(s)
- Peter M Grace
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO, USA; Current address: Department of Critical Care Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Andrew D Gaudet
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Vasiliki Staikopoulos
- Discipline of Physiology, School of Medicine, and the Australian Research Council (ARC) Centre for Nanoscale BioPhotonics (CNBP), University of Adelaide, Adelaide, SA, Australia
| | - Steven F Maier
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Mark R Hutchinson
- Discipline of Physiology, School of Medicine, and the Australian Research Council (ARC) Centre for Nanoscale BioPhotonics (CNBP), University of Adelaide, Adelaide, SA, Australia
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Linda R Watkins
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO, USA
| |
Collapse
|