1
|
Ortiz YT, Shamir LG, McMahon LR, Wilkerson JL. Characterization of commercially available murine fibrosarcoma NCTC-2472 cells both in vitro and as a model of bone cancer pain in vivo. PLoS One 2024; 19:e0309398. [PMID: 39208033 PMCID: PMC11361427 DOI: 10.1371/journal.pone.0309398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
For many cancer patients tumor burden negatively impacts quality of life due to associated pain onset. Neuropathic pain is commonly associated with late cancer stages, and is resultant of tumor metastasis to bone, herein referred to as cancer-induced bone pain. Given the severe impact on quality of life and clinical treatment strategies focusing on symptom management, novel therapeutics are needed to alleviate cancer-induced bone pain and/or reduce cancer burden. In the current study we characterized a commercially available murine fibrosarcoma cell line, NCTC-2472 in vitro, which can be used to assess the capacity of novel compounds to impact cellular viability. We found that dimethyl sulfoxide, a known cytotoxic agent and common drug preparation compound, significantly decreased cell viability in a dose-related manner. We then characterized the in vivo tumor development and associated pain behavior characteristics following implantation of NCTC-2472 fibrosarcoma into male and female C3H/HeJ mice. The C3H/HeJ strain was utilized as these mice are syngeneic with NCTC-2472 fibrosarcoma and their use reduces potential implantation failure. We found that tumor development in mice resulted in the development of mechanical allodynia but not thermal hyperalgesia. Gabapentin, a clinically relevant analgesic, produced dose-related mechanical allodynia reversal. These studies provide further characterization of a cancer-induced bone pain model that can be used to examine novel compounds as anti-cancer and analgesic therapeutics.
Collapse
Affiliation(s)
- Yuma T. Ortiz
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, United States of America
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
| | - Leila G. Shamir
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
| | - Lance R. McMahon
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
| | - Jenny L. Wilkerson
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
| |
Collapse
|
2
|
Yang Y, Yang W, Zhang R, Wang Y. Peripheral Mechanism of Cancer-Induced Bone Pain. Neurosci Bull 2024; 40:815-830. [PMID: 37798428 PMCID: PMC11178734 DOI: 10.1007/s12264-023-01126-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/28/2023] [Indexed: 10/07/2023] Open
Abstract
Cancer-induced bone pain (CIBP) is a type of ongoing or breakthrough pain caused by a primary bone tumor or bone metastasis. CIBP constitutes a specific pain state with distinct characteristics; however, it shares similarities with inflammatory and neuropathic pain. At present, although various therapies have been developed for this condition, complete relief from CIBP in patients with cancer is yet to be achieved. Hence, it is urgent to study the mechanism underlying CIBP to develop efficient analgesic drugs. Herein, we focused on the peripheral mechanism associated with the initiation of CIBP, which involves tissue injury in the bone and changes in the tumor microenvironment (TME) and dorsal root ganglion. The nerve-cancer and cancer-immunocyte cross-talk in the TME creates circumstances that promote tumor growth and metastasis, ultimately leading to CIBP. The peripheral mechanism of CIBP and current treatments as well as potential therapeutic targets are discussed in this review.
Collapse
Affiliation(s)
- Yachen Yang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Wei Yang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Ruofan Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Yanqing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
- Zhongshan-Fudan Joint Innovation Center, Zhongshan, 528437, China.
| |
Collapse
|
3
|
Zhao YX, Yao MJ, Shen JW, Zhang WX, Zhou YX. Electroacupuncture attenuates nociceptive behaviors in a mouse model of cancer pain. Mol Pain 2024; 20:17448069241240692. [PMID: 38443317 PMCID: PMC11010748 DOI: 10.1177/17448069241240692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/31/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024] Open
Abstract
Pain is a major symptom in cancer patients, and cancer-induced bone pain (CIBP) is the most common type of moderate and severe cancer-related pain. The current available analgesic treatments for CIBP have adverse effects as well as limited therapeutic effects. Acupuncture is proved effective in pain management as a safe alternative therapy. We evaluated the analgesic effect of acupuncture in treatment of cancer pain and try to explore the underlying analgesic mechanisms. Nude mice were inoculated with cancer cells into the left distal femur to establish cancer pain model. Electroacupuncture (EA) treatment was applied for the xenograft animals. Pain behaviors of mice were evaluated, followed by the detections of neuropeptide-related and inflammation-related indicators in peripheral and central levels. EA treatment alleviated cancer-induced pain behaviors covering mechanical allodynia, thermal hyperalgesia and spontaneous pain, and also down-regulated immunofluorescence expressions of neuropeptide CGRP and p75 in the skin of affected plantar area in xenograft mice, and inhibited expressions of overexpressed neuropeptide-related and inflammation-related protein in the lumbar spinal cord of xenograft mice. Overall, our findings suggest that EA treatment ameliorated cancer-induced pain behaviors in the mouse xenograft model of cancer pain, possibly through inhibiting the expressions of neuropeptide-related and inflammation-related protein in central level following tumor cell xenografts.
Collapse
Affiliation(s)
- Yu-Xue Zhao
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences , Beijing, China
| | - Ming-Jiang Yao
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences , Beijing, China
- Institute of Basic Medical Sciences, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Key Laboratory of Pharmacology of Chinese Materia Medica, Beijing, China
| | - Jian-Wu Shen
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences , Beijing, China
- Urology Department of Xiyuan Hospital, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Wen-Xi Zhang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences , Beijing, China
| | - Yuan-Xi Zhou
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences , Beijing, China
| |
Collapse
|
4
|
Edwards DN. Amino Acid Metabolism in Bone Metastatic Disease. Curr Osteoporos Rep 2023; 21:344-353. [PMID: 37277592 DOI: 10.1007/s11914-023-00797-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/26/2023] [Indexed: 06/07/2023]
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
|
5
|
Kreple CJ, Searles Nielsen S, Schoch KM, Shen T, Shabsovich M, Song Y, Racette BA, Miller TM. Protective Effects of Lovastatin in a Population-Based ALS Study and Mouse Model. Ann Neurol 2023; 93:881-892. [PMID: 36627836 DOI: 10.1002/ana.26600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 12/12/2022] [Accepted: 01/08/2023] [Indexed: 01/12/2023]
Abstract
OBJECTIVE The objective of this study was to use a novel combined pharmacoepidemiologic and amyotrophic lateral sclerosis (ALS) mouse model approach to identify potential motor neuron protective medications. METHODS We constructed a large, population-based case-control study to investigate motor neuron disease (MND) among US Medicare beneficiaries aged 66 to 90 in 2009. We included 1,128 incident MND cases and 56,400 age, sex, race, and ethnicity matched controls. We calculated MND relative risk for >1,000 active ingredients represented in Part D (pharmacy) claims in 2006 to 2007 (>1 year before diagnosis/reference). We then applied a comprehensive screening approach to select medications for testing in SOD1G93A mice: sulfasalazine, telmisartan, and lovastatin. We treated mice with the human dose equivalent of the medication or vehicle via subcutaneous osmotic pump before onset of weakness. We then assessed weight, gait, and survival. In additional mice, we conducted histological studies. RESULTS We observed previously established medical associations for MND and an inverse dose-response association between lovastatin and MND, with 28% reduced risk at 40 mg/day. In SOD1G93A mouse studies, sulfasalazine and telmisartan conferred no benefit, whereas lovastatin treatment delayed onset and prolonged survival. Lovastatin treated mice also had less microgliosis, misfolded SOD1, and spinal motor neuron loss in the ventral horn. INTERPRETATION Lovastatin reduced the risk of ALS in humans, which was confirmed in an ALS mouse model by delayed symptom onset, prolonged survival, and preservation of motor neurons. Although further studies to understand the mechanism are required, lovastatin may represent a potential neuroprotective therapy for patients with ALS. These data demonstrate the utility of a combined pharmacoepidemiologic and mouse model approach. ANN NEUROL 2023.
Collapse
Affiliation(s)
- Collin J Kreple
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | | | - Kathleen M Schoch
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Tao Shen
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Mark Shabsovich
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Yizhe Song
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Brad A Racette
- Department of Neurology, Washington University School of Medicine, St. Louis, MO.,Barrow Neurological Institute, Phoenix, AZ.,School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Parktown, South Africa
| | - Timothy M Miller
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
6
|
Is depression the missing link between inflammatory mediators and cancer? Pharmacol Ther 2022; 240:108293. [PMID: 36216210 DOI: 10.1016/j.pharmthera.2022.108293] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/14/2022] [Accepted: 09/26/2022] [Indexed: 11/05/2022]
Abstract
Patients with cancer are at greater risk of developing depression in comparison to the general population and this is associated with serious adverse effects, such as poorer quality of life, worse prognosis and higher mortality. Although the relationship between depression and cancer is now well established, a common underlying pathophysiological mechanism between the two conditions is yet to be elucidated. Existing theories of depression, based on monoamine neurotransmitter system dysfunction, are insufficient as explanations of the disorder. Recent advances have implicated neuroinflammatory mechanisms in the etiology of depression and it has been demonstrated that inflammation at a peripheral level may be mirrored centrally in astrocytes and microglia serving to promote chronic levels of inflammation in the brain. Three major routes to depression in cancer in which proinflammatory mediators are implicated, seem likely. Activation of the kynurenine pathway involving cytokines, increases tryptophan catabolism, resulting in diminished levels of serotonin which is widely acknowledged as being the hallmark of depression. It also results in neurotoxic effects on brain regions thought to be involved in the evolution of major depression. Proinflammatory mediators also play a crucial role in impairing regulatory glucocorticoid mediated feedback of the hypothalamic-pituitary-adrenal axis, which is activated by stress and considered to be involved in both depression and cancer. The third route is via the glutamatergic pathway, whereby glutamate excitotoxicity may lead to depression associated with cancer. A better understanding of the mechanisms underlying these dysregulated and other newly emerging pathways may provide a rationale for therapeutic targeting, serving to improve the care of cancer patients.
Collapse
|
7
|
Temmermand R, Barrett JE, Fontana ACK. Glutamatergic systems in neuropathic pain and emerging non-opioid therapies. Pharmacol Res 2022; 185:106492. [PMID: 36228868 PMCID: PMC10413816 DOI: 10.1016/j.phrs.2022.106492] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/05/2022] [Accepted: 10/06/2022] [Indexed: 01/14/2023]
Abstract
Neuropathic pain, a disease of the somatosensory nervous system, afflicts many individuals and adequate management with current pharmacotherapies remains elusive. The glutamatergic system of neurons, receptors and transporters are intimately involved in pain but, to date, there have been few drugs developed that therapeutically modulate this system. Glutamate transporters, or excitatory amino acid transporters (EAATs), remove excess glutamate around pain transmitting neurons to decrease nociception suggesting that the modulation of glutamate transporters may represent a novel approach to the treatment of pain. This review highlights and summarizes (1) the physiology of the glutamatergic system in neuropathic pain, (2) the preclinical evidence for dysregulation of glutamate transport in animal pain models, and (3) emerging novel therapies that modulate glutamate transporters. Successful drug discovery requires continuous focus on basic and translational methods to fully elucidate the etiologies of this disease to enable the development of targeted therapies. Increasing the efficacy of astrocytic EAATs may serve as a new way to successfully treat those suffering from this devastating disease.
Collapse
Affiliation(s)
- Rhea Temmermand
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - James E Barrett
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Andréia C K Fontana
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA.
| |
Collapse
|
8
|
Zhu L, Meng D, Wang X, Chen X. Ferroptosis-Driven Nanotherapeutics to Reverse Drug Resistance in Tumor Microenvironment. ACS APPLIED BIO MATERIALS 2022; 5:2481-2506. [PMID: 35614872 DOI: 10.1021/acsabm.2c00199] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Ferroptosis, characterized by iron-dependent lipid reactive oxygen species (ROS) accumulation, is non-apoptotic programmed cell death highly relevant to tumor development. It was found to manipulate oncogenes and resistant mutations of cancer cells via lipid metabolism pathways converging on phospholipid glutathione peroxidase (GPX4) that squanders lipid peroxides (L-OOH) to block the iron-mediated reactions of peroxides, thus rendering resistant cancer cells vulnerable to ferroptotic cell death. By accumulating ROS and lipid peroxidation (LPO) products to lethal levels in tumor microenvironment (TME), ferroptosis-driven nanotherapeutics show a superior ability of eradicating aggressive malignancies than traditional therapeutic modalities, especially for the drug-resistant tumors with high metastasis tendency. Moreover, Fenton reaction, inhibition of GPX-4, and exogenous regulation of LPO are three major therapeutic strategies to induce ferroptosis in cancer cells, which were generally applied in ferroptosis-driven nanotherapeutics. In this review, we elaborate current trends of ferroptosis-driven nanotherapeutics to reverse drug resistance of tumors in anticancer fields at the intersection of cancer biology, materials science, and chemistry. Finally, their challenges and perspectives toward feasible translational studies are spotlighted, which would ignite the hope of anti-resistant cancer treatment.
Collapse
Affiliation(s)
- Liyun Zhu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Danni Meng
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Xu Wang
- Hangzhou Medical College, Binjiang Higher Education Park, Binwen Road 481, Hangzhou 310053, China
| | - Xuerui Chen
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| |
Collapse
|
9
|
Rao R, Shah S, Bhattacharya D, Toukam DK, Cáceres R, Pomeranz Krummel DA, Sengupta S. Ligand-Gated Ion Channels as Targets for Treatment and Management of Cancers. Front Physiol 2022; 13:839437. [PMID: 35350689 PMCID: PMC8957973 DOI: 10.3389/fphys.2022.839437] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/07/2022] [Indexed: 12/24/2022] Open
Abstract
Ligand-gated ion channels are an ionotropic receptor subtype characterized by the binding of an extracellular ligand, followed by the transient passage of ions through a transmembrane pore. Ligand-gated ion channels are commonly subcategorized into three superfamilies: purinoreceptors, glutamate receptors, and Cys-loop receptors. This classification is based on the differing topographical morphology of the receptors, which in turn confers functional differences. Ligand-gated ion channels have a diverse spatial and temporal expression which implicate them in key cellular processes. Given that the transcellular electrochemical gradient is finely tuned in eukaryotic cells, any disruption in this homeostasis can contribute to aberrancies, including altering the activity of pro-tumorigenic molecular pathways, such as the MAPK/ERK, RAS, and mTOR pathways. Ligand-gated ion channels therefore serve as a potential targetable system for cancer therapeutics. In this review, we analyze the role that each of the three ligand-gated ion channel superfamilies has concerning tumor proliferation and as a target for the treatment of cancer symptomatology.
Collapse
Affiliation(s)
| | | | | | | | | | - Daniel A. Pomeranz Krummel
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Soma Sengupta
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
10
|
Verbruggen L, Sprimont L, Bentea E, Janssen P, Gharib A, Deneyer L, De Pauw L, Lara O, Sato H, Nicaise C, Massie A. Chronic Sulfasalazine Treatment in Mice Induces System x c - - Independent Adverse Effects. Front Pharmacol 2021; 12:625699. [PMID: 34084129 PMCID: PMC8167035 DOI: 10.3389/fphar.2021.625699] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 04/26/2021] [Indexed: 01/17/2023] Open
Abstract
Despite ample evidence for the therapeutic potential of inhibition of the cystine/glutamate antiporter system xc− in neurological disorders and in cancer, none of the proposed inhibitors is selective. In this context, a lot of research has been performed using the EMA- and FDA-approved drug sulfasalazine (SAS). Even though this molecule is already on the market for decades as an anti-inflammatory drug, serious side effects due to its use have been reported. Whereas for the treatment of the main indications, SAS needs to be cleaved in the intestine into the anti-inflammatory compound mesalazine, it needs to reach the systemic circulation in its intact form to allow inhibition of system xc−. The higher plasma levels of intact SAS (or its metabolites) might induce adverse effects, independent of its action on system xc−. Some of these effects have however been attributed to system xc− inhibition, calling into question the safety of targeting system xc−. In this study we chronically treated system xc− - deficient mice and their wildtype littermates with two different doses of SAS (160 mg/kg twice daily or 320 mg/kg once daily, i.p.) and studied some of the adverse effects that were previously reported. SAS had a negative impact on the survival rate, the body weight, the thermoregulation and/or stress reaction of mice of both genotypes, and thus independent of its inhibitory action on system xc−. While SAS decreased the total distance travelled in the open-field test the first time the mice encountered the test, it did not influence this parameter on the long-term and it did not induce other behavioral changes such as anxiety- or depressive-like behavior. Finally, no major histological abnormalities were observed in the spinal cord. To conclude, we were unable to identify any undesirable system xc−-dependent effect of chronic administration of SAS.
Collapse
Affiliation(s)
- Lise Verbruggen
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lindsay Sprimont
- Laboratory Neurodegeneration and Regeneration, URPHyM-NARILIS, Université de Namur, Namur, Belgium
| | - Eduard Bentea
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Pauline Janssen
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Azzedine Gharib
- Laboratory Neurodegeneration and Regeneration, URPHyM-NARILIS, Université de Namur, Namur, Belgium
| | - Lauren Deneyer
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Laura De Pauw
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Olaya Lara
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hideyo Sato
- Department of Medical Technology, Niigata University, Niigata, Japan
| | - Charles Nicaise
- Laboratory Neurodegeneration and Regeneration, URPHyM-NARILIS, Université de Namur, Namur, Belgium
| | - Ann Massie
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
11
|
Zhu YF, Linher-Melville K, Wu J, Fazzari J, Miladinovic T, Ungard R, Zhu KL, Singh G. Bone cancer-induced pain is associated with glutamate signalling in peripheral sensory neurons. Mol Pain 2021; 16:1744806920911536. [PMID: 32133928 PMCID: PMC7059229 DOI: 10.1177/1744806920911536] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We previously identified that several cancer cell lines known to induce
nociception in mouse models release glutamate in vitro. Although the mechanisms
of glutamatergic signalling have been characterized primarily in the central
nervous system, its importance in the peripheral nervous system has been
recognized in various pathologies, including cancer pain. We therefore
investigated the effect of glutamate on intracellular electrophysiological
characteristics of peripheral sensory neurons in an immunocompetent rat model of
cancer-induced pain based on surgical implantation of mammary rat metastasis
tumour-1 cells into the distal epiphysis of the right femur. Behavioural
evidence of nociception was detected using von Frey tactile assessment. Activity
of sensory neurons was measured by intracellular electrophysiological recordings
in vivo. Glutamate receptor expression at the mRNA level in relevant dorsal root
ganglia was determined by reverse transcription polymerase chain reaction using
rat-specific primers. Nociceptive and non-nociceptive mechanoreceptor neurons
exhibiting changes in neural firing patterns associated with increased
nociception due to the presence of a bone tumour rapidly responded to
sulphasalazine injection, an agent that pharmacologically blocks non-vesicular
glutamate release by inhibiting the activity of the system
xC− antiporter. In addition, both types of
mechanoreceptor neurons demonstrated excitation in response to intramuscular
glutamate injection near the femoral head, which corresponds to the location of
cancer cell injection to induce the bone cancer-induced pain model. Therefore,
glutamatergic signalling contributes to cancer pain and may be a factor in
peripheral sensitization and induced tactile hypersensitivity associated with
bone cancer-induced pain.
Collapse
Affiliation(s)
- Yong Fang Zhu
- Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Katja Linher-Melville
- Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Jianhan Wu
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Jennifer Fazzari
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Tanya Miladinovic
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Robert Ungard
- Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Kan Lun Zhu
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Gurmit Singh
- Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
12
|
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: 2.3] [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
|
13
|
Long H, Zheng H, Ai L, Osman K, Liu Z. Down-Regulation of NOX4 Expression in Dorsal Horn of Spinal Cord Could Alleviate Cancer-Induced Bone Pain in Rats by Reducing Oxidative Stress Response. Cancer Manag Res 2020; 12:10929-10938. [PMID: 33154672 PMCID: PMC7608490 DOI: 10.2147/cmar.s263177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/02/2020] [Indexed: 01/06/2023] Open
Abstract
Introduction Cancer-induced bone pain (CIBP) is very common in patients with advanced cancer. Recent studies have shown that reactive oxygen species (ROS) can sense and regulate pain response process through spinal cord mechanism, and play a role in CIBP. NADPH oxidase (NOX) is a group of protease complexes that produce ROS. In the current study, we investigated the expression of NOX4 in the spinal dorsal horn of rats with CIBP and its related role and molecular mechanism. Materials and Methods A rat CIBP model was established by injecting Walker-256 cells into the tibia medullary cavity, and the expression of NOX4 in spinal dorsal horn was down-regulated by injecting lentivirus into spinal cord. RT-PCR, Western blot and immunofluorescence staining were used to detect the expression of NOX4 in CIBP rats, cell localization and its effect on CIBP rats, and the effect of down-regulating the expression of NOX4 on the expression of H2O2, nitric oxide synthase nNO, antioxidant enzyme SOD, and the activity of neuro-receptor in spinal dorsal horn of rats. Results In rats with CIBP, the expression of NOX4 was significantly increased, and immunofluorescence showed that NOX4 was mainly expressed in microglia in the dorsal horn of spinal cord. Down-regulating the expression of NOX4 in rats can reduce the level of H2O2 and nNO in dorsal horn tissue, and increase the expression of SOD to reduce the oxidative stress response. At the same time, down-regulating NOX4 can reduce the sensitivity of spinal cord and relieve the pain of bone cancer by inhibiting the expression of NMDAR and GABAA-γ2 in dorsal horn tissue. Conclusion NOX4 is a promising therapeutic target in CIBP, and down-regulation of NOX4 expression can alleviate CIBP in rats by reducing oxidative stress and weakening spinal cord sensitization.
Collapse
Affiliation(s)
- Hao Long
- Department of Pain Management, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Hui Zheng
- Orthopedics Department, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Long Ai
- Department of Pain Management, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Kamil Osman
- Department of Pain Management, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Zhigang Liu
- Department of Pain Management, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| |
Collapse
|
14
|
The impact of statins on physical activity and exercise capacity: an overview of the evidence, mechanisms, and recommendations. Eur J Appl Physiol 2020; 120:1205-1225. [PMID: 32248287 DOI: 10.1007/s00421-020-04360-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 03/24/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE Statins are among the most widely prescribed medications worldwide. Considered the 'gold-standard' treatment for cardiovascular disease (CVD), statins inhibit HMG-CoA reductase to ultimately reduce serum LDL-cholesterol levels. Unfortunately, the main adverse event of statin use is the development of muscle-associated problems, referred to as SAMS (statin-associated muscle symptoms). While regular moderate physical activity also decreases CVD risk, there is apprehension that physical activity may induce and/or exacerbate SAMS. While much work has gone into identifying the epidemiology of SAMS, only recent research has focused on the extent to which these muscle symptoms are accompanied by functional declines. The purpose of this review is to provide an overview of possible mechanisms underlying SAMS and summarize current evidence regarding the relationship between statin treatment, physical activity, exercise capacity, and SAMS development. METHODS PubMed and Google Scholar databases were used to search the most relevant and up-to-date peer-reviewed research on the topic. RESULTS The mechanism(s) behind SAMS, including altered mitochondrial metabolism, reduced coenzyme Q10 levels, reduced vitamin D levels, impaired calcium homeostasis, elevated extracellular glutamate, and genetic polymorphisms, still lack consensus and remain up for debate. Our summation of the evidence leads us to suggest that the etiology of SAMS development is likely multifactorial. Our review also demonstrates that there is limited evidence for statins impairing exercise adaptations or reducing exercise capacity for the majority of the investigated populations. CONCLUSION The available evidence indicates that the benefits of engaging in physical activity while on statin medication largely outweigh the risks.
Collapse
|
15
|
Ungard RG, Zhu YF, Yang S, Nakhla P, Parzei N, Zhu KL, Singh G. Response to pregabalin and progesterone differs in male and female rat models of neuropathic and cancer pain. CANADIAN JOURNAL OF PAIN-REVUE CANADIENNE DE LA DOULEUR 2020; 4:39-58. [PMID: 33987485 PMCID: PMC7951160 DOI: 10.1080/24740527.2020.1724776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Background: Cancer pain involves nervous system damage and pathological neurogenesis. Neuropathic pain arises from damage to the nervous system and is driven by ectopic signaling. Both progesterone and pregabalin are neuroprotective in animal models, and there is evidence that both drugs bind to and inhibit voltage-gated calcium channels. Aims: This study was designed to characterize the effects of progesterone and pregabalin in preclinical models of cancer and neuropathic pain in both sexes. Methods: We measured peripheral sensory signaling by intracellular in vivo electrophysiology and behavioral indicators of pain in rat models of cancer-induced bone pain and neuropathic pain. Results: Female but not male models of cancer pain showed a behavioral response to treatment and pregabalin reduced excitability in C and A high-threshold but not low-threshold sensory neurons of both sexes. Male models of neuropathic pain treated with pregabalin demonstrated higher signaling thresholds only in A high-threshold neurons, and behavioral data indicated a clear recovery to baseline mechanical withdrawal thresholds in all treatment groups. Female rat treatment groups did not show excitability changes in sensory neurons, but all demonstrated higher mechanical withdrawal thresholds than vehicle-treated females, although not to baseline levels. Athymic female rat models of neuropathic pain showed no behavioral or electrophysiological responses to treatment. Conclusions: Both pregabalin and progesterone showed evidence of efficacy in male models of neuropathic pain. These results add to the evidence demonstrating differential effects of treatments for pain in male and female animals and widely differing responses in models of cancer and neuropathic pain.
Collapse
Affiliation(s)
- Robert G Ungard
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Yong Fang Zhu
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Sarah Yang
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Peter Nakhla
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Natalka Parzei
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Kan Lun Zhu
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Gurmit Singh
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
16
|
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: 107] [Impact Index Per Article: 21.4] [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
|
17
|
Rebalka IA, Cao AW, May LL, Tarnopolsky MA, Hawke TJ. Statin administration activates system xC - in skeletal muscle: a potential mechanism explaining statin-induced muscle pain. Am J Physiol Cell Physiol 2019; 317:C894-C899. [PMID: 31509447 DOI: 10.1152/ajpcell.00308.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Statins are a cholesterol-lowering drug class that significantly reduce cardiovascular disease risk. Despite their safety and effectiveness, musculoskeletal side-effects, particularly myalgia, are prominent and the most common reason for discontinuance. The cause of statin-induced myalgia is unknown, so defining the underlying mechanism(s) and potential therapeutic strategies is of clinical importance. Here we tested the hypothesis that statin administration activates skeletal muscle system xC-, a cystine/glutamate antiporter, to increase intracellular cysteine and therefore glutathione synthesis to attenuate statin-induced oxidative stress. Increased system xC- activity would increase interstitial glutamate; an amino acid associated with peripheral nociception. Consistent with our hypothesis, atorvastatin treatment significantly increased mitochondrial reactive oxygen species (ROS; 41%) and glutamate efflux (up to 122%) in C2C12 mouse skeletal muscle myotubes. Statin-induced glutamate efflux was confirmed to be the result of system xC- activation, as cotreatment with sulfasalazine (system xC- inhibitor) negated this rise in extracellular glutamate. These findings were reproduced in primary human myotubes but, consistent with being muscle-specific, were not observed in primary human dermal fibroblasts. To further demonstrate that statin-induced increases in ROS triggered glutamate efflux, C2C12 myotubes were cotreated with atorvastatin and various antioxidants. α-Tocopherol and cysteamine bitartrate reversed the increase in statin-induced glutamate efflux, bringing glutamate levels between 50 and 92% of control-treated levels. N-acetylcysteine (a system xC- substrate) increased glutamate efflux above statin treatment alone: up to 732% greater than control treatment. Taken together, we provide a mechanistic foundation for statin-induced myalgia and offer therapeutic insights to alleviate this particular statin-associated side-effect.
Collapse
Affiliation(s)
- Irena A Rebalka
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Andrew W Cao
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Linda L May
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Mark A Tarnopolsky
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
18
|
Fazzari J, Singh G. Effect of glutaminase inhibition on cancer-induced bone pain. BREAST CANCER-TARGETS AND THERAPY 2019; 11:273-282. [PMID: 31571981 PMCID: PMC6750878 DOI: 10.2147/bctt.s215655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/23/2019] [Indexed: 12/15/2022]
Abstract
Purpose The complex nature of cancer-induced bone pain (CIBP) has led to investigation into cancer-targeted therapies. This has involved targeting glutamate release from the tumor, secreted as a byproduct of antioxidant responses and metabolic disruption. Cancer cells undergo many metabolic changes that result in increased glutamine metabolism and subsequently the production of glutamate. Glutaminase (GLS) is the enzyme that mediates the conversion of glutamine to glutamate and has been shown to be upregulated in many cancer types including malignancies of the breast. This enzyme, therefore, represents another potential therapeutic target for CIBP, one that lies upstream of glutamate secretion. Methods A recently developed inhibitor of GLS, CB-839, was tested in an animal model of CIBP induced by intrafemoral MDA-MB-231 xenografts. CIBP behaviors were assessed using Dynamic Weight Bearing and Dynamic Plantar Aesthesiometer readings of mechanical hyperalgesia and allodynia. Results CB-839 failed to modulate any of the associated nociceptive behaviors induced by intrafemoral MDA-MB-231 tumor growth. Further investigation in vitro revealed the sensitivity of the drug is dependent on the metabolic flexibility of the cell line being tested which can be modulated by cell culture environment. Conclusion Adaptation to metabolic disturbances may explain the failure of CB-839 to exhibit any significant effects in vivo and the metabolic flexibility of the cell line tested should be considered for future investigations studying the metabolic effects of glutaminase inhibition.
Collapse
Affiliation(s)
- Jennifer Fazzari
- 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
|
19
|
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: 0.8] [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
|
20
|
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: 2.4] [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
|
21
|
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: 0.8] [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
|
22
|
Miladinovic T, Sharma M, Phan A, Geres H, Ungard RG, Linher-Melville K, Singh G. Activation of hippocampal microglia in a murine model of cancer-induced pain. J Pain Res 2019; 12:1003-1016. [PMID: 30936739 PMCID: PMC6430067 DOI: 10.2147/jpr.s191860] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Introduction Pain is a common and debilitating comorbidity of metastatic breast cancer. The hippocampus has been implicated in nociceptive processing, particularly relating to the subjective aspect of pain. Here, a syngeneic mouse model was used to characterize the effects of peripheral tumors on hippocampal microglial activation in relation to cancer-induced pain (CIP). Materials and methods Mice were systemically treated with the colony-stimulating factor 1 receptor inhibitor Pexidartinib prior to intrafemoral (IF) or subcutaneous 4T1 carcinoma cell inoculation. Spontaneous and evoked nociceptive responses were quantitated throughout tumor development, and contralateral hippocampi were collected via endpoint microdissection for RNA analysis. Additionally, IF tumor-bearing animals were sacrificed on days 5, 10, 15, and 20 post 4T1 cell inoculation, and brain sections were immunofluorescently stained for Iba1, a marker of activated microglia. Results Ablation of these neuroimmune cells with the CSF1R inhibitor Pexidartinib delayed the onset and severity of cancer-induced nociceptive behaviors in IF tumor-bearing animals, adding to the body of literature that demonstrates microglial contribution to the development and maintenance of CIP. Furthermore, in untreated IF tumor-bearing mice, nociceptive behaviors appeared to progress in parallel with microglial activation in hippocampal regions. Immunofluorescent Iba1+ microglia increased in the dentate gyrus and cornu ammonis 1 hippocampal regions in IF tumor-bearing animals over time, which was confirmed at the mRNA level using relevant microglial markers. Conclusion This is the first experimental evidence to demonstrate the effects of peripheral tumor-induced nociception on hippocampal microglial activation. The increase in hippocampal microglia observed in the present study may reflect the emotional and cognitive deficits reported by patients with CIP.
Collapse
Affiliation(s)
- Tanya Miladinovic
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, ON L8S 4M1, Canada, .,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4M1, Canada,
| | - Manu Sharma
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, ON L8S 4M1, Canada, .,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4M1, Canada,
| | - Andy Phan
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, ON L8S 4M1, Canada, .,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4M1, Canada,
| | - Hana Geres
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, ON L8S 4M1, Canada, .,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4M1, Canada,
| | - Robert G Ungard
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, ON L8S 4M1, Canada, .,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4M1, Canada,
| | - Katja Linher-Melville
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, ON L8S 4M1, Canada, .,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4M1, Canada,
| | - Gurmit Singh
- Michael G. DeGroote Institute for Pain Research and Care, Medicine, McMaster University, Hamilton, ON L8S 4M1, Canada, .,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4M1, Canada,
| |
Collapse
|
23
|
Lux S, Lobos N, Lespay-Rebolledo C, Salas-Huenuleo E, Kogan MJ, Flores C, Pinto M, Hernandez A, Pelissier T, Constandil L. The antinociceptive effect of resveratrol in bone cancer pain is inhibited by the Silent Information Regulator 1 inhibitor selisistat. J Pharm Pharmacol 2018; 71:816-825. [DOI: 10.1111/jphp.13064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 12/02/2018] [Indexed: 01/10/2023]
Abstract
Abstract
Objectives
To study the antinociceptive effect of single and repeated doses of resveratrol in a bone cancer pain model, and whether this effect is prevented by the Silent Information Regulator 1 (SIRT1) inhibitor selisistat.
Methods
The femoral intercondylar bone of BALB/c mice was injected with 1 000 000 BJ3Z cancer cells. Bone resorption and tumour mass growth (measured by in vivo X-ray and fluorescence imaging), as well as mechanical nociceptive thresholds (von Frey device) and dynamic functionality (rotarod machine), were evaluated during the following 4 weeks. Acute resveratrol (100 mg/kg i.p.) and/or selisistat (10 mg/kg s.c.) were administered on day 14. Chronic resveratrol (100 mg/kg i.p., daily) and/or selisistat (0.5 μg/h s.c., Alzet pump) were administered between days 14 and 20.
Key findings
Tumour growth gradually incremented until day 31, while mechanical hyperalgesia started on day 3 after cancer cell injection. Acute resveratrol increased the mechanical threshold of pain (peaking at 1.5 h), while the dynamic functionality decreased. Chronic resveratrol produced a sustained antinociceptive effect on mechanical hyperalgesia and improved the loss of dynamic functionality induced by the bone cancer tumour. Selisistat prevented all the effects of resveratrol.
Conclusions
Acute and chronic resveratrol induces antinociceptive effect in the model of metastatic osseous oncological pain, an effect that would be mediated by SIRT1 molecular signalling.
Collapse
Affiliation(s)
- Sebastian Lux
- Department of Biology, Laboratory of Neurobiology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Nicolas Lobos
- Department of Biology, Laboratory of Neurobiology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Carolyne Lespay-Rebolledo
- Program of Molecular and Clinical Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Edison Salas-Huenuleo
- Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
| | - Marcelo J Kogan
- Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
| | - Christian Flores
- Department of Biology, Laboratory of Neurobiology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Mauricio Pinto
- Department of Biology, Laboratory of Immunology of Reproduction, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Alejandro Hernandez
- Department of Biology, Laboratory of Neurobiology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Teresa Pelissier
- Department of Biology, Laboratory of Neurobiology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
- Program of Molecular and Clinical Pharmacology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Luis Constandil
- Department of Biology, Laboratory of Neurobiology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Santiago, Chile
| |
Collapse
|
24
|
Kawahara B, Ramadoss S, Chaudhuri G, Janzen C, Sen S, Mascharak PK. Carbon monoxide sensitizes cisplatin-resistant ovarian cancer cell lines toward cisplatin via attenuation of levels of glutathione and nuclear metallothionein. J Inorg Biochem 2018; 191:29-39. [PMID: 30458366 DOI: 10.1016/j.jinorgbio.2018.11.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 10/31/2018] [Accepted: 11/04/2018] [Indexed: 02/06/2023]
Abstract
Cisplatin resistance remains a major impediment to effective treatment of ovarian cancer. Despite initial platinum responsiveness, thiol-containing peptides and proteins, glutathione (GSH) and metallothionein (MT), bind and inactivate cisplatin in cancer cells. Indeed, high levels of GSH and MT in ovarian cancers impart cisplatin resistance and are predictive of poor prognosis. Cystathionine β-synthase (CBS), an enzyme involved in sulfur metabolism, is overexpressed in ovarian cancer tissues and is itself associated with cisplatin resistance. Treatment with exogenous carbon monoxide (CO), a known inhibitor of CBS, may mitigate cisplatin resistance in ovarian cancer cells by attenuation of GSH and MT levels. Using a photo-activated CO-releasing molecule (photoCORM), [Mn(CO)3(phen)(PTA)]CF3SO3 (phen = 1,10-phenanthroline, PTA = 1,3,5-triza-7-phosphaadamantane) we assessed the ability of CO to sensitize established cisplatin-resistant ovarian cancer cell lines to cisplatin. Cisplatin-resistant cells, treated with both cisplatin and CO, exhibited significantly lower cell viability and increased poly (ADP-ribose) polymerase (PARP) cleavage versus those treated with cisplatin alone. These cisplatin-resistant cell lines overexpressed CBS and had increased steady state levels of GSH and expression of nuclear MT. Both CO treatment and lentiviral-mediated silencing of CBS attenuated GSH and nuclear MT expression in cisplatin resistant cells. We have demonstrated that CO, delivered from a photoCORM, sensitizes established cisplatin-resistant cell lines to cisplatin. Furthermore, we have presented strong evidence that the effects of CO in circumventing chemotherapeutic drug resistance is at least in part mediated by the inactivation of endogenous CBS.
Collapse
Affiliation(s)
- Brian Kawahara
- Contribution from Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064, United States of America
| | - Sivakumar Ramadoss
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA 90095, United States of America
| | - Gautam Chaudhuri
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA 90095, United States of America
| | - Carla Janzen
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA 90095, United States of America
| | - Suvajit Sen
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA 90095, United States of America.
| | - Pradip K Mascharak
- Contribution from Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064, United States of America.
| |
Collapse
|
25
|
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: 2.3] [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
|
26
|
Yamamoto M, Motomura E, Yanagisawa R, Hoang VAT, Mogi M, Mori T, Nakamura M, Takeya M, Eto K. Evaluation of neurobehavioral impairment in methylmercury-treated KK-Ay mice by dynamic weight-bearing test. J Appl Toxicol 2018; 39:221-230. [PMID: 30175511 DOI: 10.1002/jat.3710] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/21/2018] [Accepted: 07/11/2018] [Indexed: 12/22/2022]
Abstract
Methylmercury (MeHg) is known to cause neurobehavioral impairment in human and experimental animals. We previously reported that MeHg (5 mg Hg/kg) induced severe neurobehavioral dysfunction in 4-week-old KK-Ay mice, although it is difficult to evaluate quantitatively the neurobehavioral impairment in MeHg-treated KK-Ay mice because of their obesity. The aim of this study was to evaluate MeHg-induced neurobehavioral dysfunction in KK-Ay mice using the dynamic weight-bearing test, which analyzes the animal's weight distribution between the four limbs. Male 12-week-old KK-Ay mice were treated with MeHg (5 mg Hg/kg) three times per week for 5 weeks. Body weight loss began after approximately 2 weeks of MeHg treatment, and decreased significantly at 4 weeks. Seven of the nine MeHg-treated mice exhibited overt neurological symptoms such as ataxia and gait disturbance. The weight-bearing load was lower for the forelimb than for the hindlimb at baseline and until 1 week after MeHg treatment was initiated. In weeks 2-4, the dynamic weight-bearing loads on the forelimb and hindlimb were similar. The load on the forelimb exceeded the load on the hindlimb after 5 weeks of treatment. This finding indicates that the dynamic weight-bearing test is useful for semi-quantitative evaluation of neurobehavioral impairment in MeHg-treated rodents, and is less stressful for the animals. Infiltration of CD204-positive macrophages was observed in the sciatic nerve of MeHg-treated mice, suggesting that CD204 can serve as a useful marker of tissue injury in peripheral nerves and a possible target in regenerating peripheral nerves and controlling neuropathies.
Collapse
Affiliation(s)
- Megumi Yamamoto
- Integrated Physiology Section, Department of Basic Medical Science, National Institute for Minamata Disease, 4058-18 Hama, Minamata, Kumamoto, 867-0008, Japan
| | - Eriko Motomura
- Integrated Physiology Section, Department of Basic Medical Science, National Institute for Minamata Disease, 4058-18 Hama, Minamata, Kumamoto, 867-0008, Japan
| | - Rie Yanagisawa
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, Ibaraki, 305-8506, Japan
| | - Van Anh Thi Hoang
- Integrated Physiology Section, Department of Basic Medical Science, National Institute for Minamata Disease, 4058-18 Hama, Minamata, Kumamoto, 867-0008, Japan.,Graduate School of Environmental and Symbiotic Science, Prefectural University of Kumamoto, 3-1-100 Tsukide, Higashi-ku, Kumamoto, 862-8502, Japan
| | - Masaki Mogi
- Department of Pharmacology, Ehime University Graduate School of Medicine, Ehime, 791-0295, Japan
| | - Tomohisa Mori
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, 142-8501, Japan
| | - Masaaki Nakamura
- Department of Clinical Medicine, National Institute for Minamata Disease, 4058-18 Hama, Minamata, Kumamoto, 867-0008, Japan
| | - Motohiro Takeya
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Komyo Eto
- Health and Nursing Facilities for the Aged, Jushindai, Shinwakai, 272 Ikurakitakata, Tamana, Kumamoto, 865-0041, Japan
| |
Collapse
|
27
|
Dalley CB, Wroblewska B, Wolfe BB, Wroblewski JT. The Role of Metabotropic Glutamate Receptor 1 Dependent Signaling in Glioma Viability. J Pharmacol Exp Ther 2018; 367:59-70. [PMID: 30054311 DOI: 10.1124/jpet.118.250159] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/18/2018] [Indexed: 12/31/2022] Open
Abstract
Glioma refers to malignant central nervous system tumors that have histologic characteristics in common with glial cells. The most prevalent type, glioblastoma multiforme, is associated with a poor prognosis and few treatment options. On the basis of reports of aberrant expression of mGluR1 mRNA in glioma, evidence that melanoma growth is directly influenced by glutamate metabotropic receptor 1 (mGluR1), and characterization of β-arrestin-dependent prosurvival signaling by this receptor, this study investigated the hypothesis that glioma cell lines aberrantly express mGluR1 and depend on mGluR1-mediated signaling to maintain viability and proliferation. Three glioma cell lines (Hs683, A172, and U87) were tested to confirm mGluR1 mRNA expression and the dependence of glioma cell viability on glutamate. Pharmacologic and genetic evidence is presented that suggests mGluR1 signaling specifically supports glioma proliferation and viability. For example, selective noncompetitive antagonists of mGluR1, CPCCOEt and JNJ16259685, decreased the viability of these cells in a dose-dependent manner, and glutamate metabotropic receptor 1 gene silencing significantly reduced glioma cell proliferation. Also, results of an anchorage-independent growth assay suggested that noncompetitive antagonism of mGluR1 may decrease the tumorigenic potential of Hs683 glioma cells. Finally, data are provided that support the hypothesis that a β-arrestin-dependent signaling cascade may be involved in glutamate-stimulated viability in glioma cells and that ligand bias may exist at mGluR1 expressed in these cells. Taken together, the results strongly suggest that mGluR1 may act as a proto-oncogene in glioma and be a viable drug target in glioma treatment.
Collapse
Affiliation(s)
- Carrie Bowman Dalley
- The Wroblewski Laboratory, Department of Pharmacology and Physiology (C.B.D., B.W., B.B.W., J.T.W.) and School of Nursing and Health Studies (C.B.D.), Georgetown University Medical Center, Washington, District of Columbia
| | - Barbara Wroblewska
- The Wroblewski Laboratory, Department of Pharmacology and Physiology (C.B.D., B.W., B.B.W., J.T.W.) and School of Nursing and Health Studies (C.B.D.), Georgetown University Medical Center, Washington, District of Columbia
| | - Barry B Wolfe
- The Wroblewski Laboratory, Department of Pharmacology and Physiology (C.B.D., B.W., B.B.W., J.T.W.) and School of Nursing and Health Studies (C.B.D.), Georgetown University Medical Center, Washington, District of Columbia
| | - Jarda T Wroblewski
- The Wroblewski Laboratory, Department of Pharmacology and Physiology (C.B.D., B.W., B.B.W., J.T.W.) and School of Nursing and Health Studies (C.B.D.), Georgetown University Medical Center, Washington, District of Columbia
| |
Collapse
|
28
|
Fazzari J, Linher-Melville K, Singh G. Tumour-Derived Glutamate: Linking Aberrant Cancer Cell Metabolism to Peripheral Sensory Pain Pathways. Curr Neuropharmacol 2018; 15:620-636. [PMID: 27157265 PMCID: PMC5543678 DOI: 10.2174/1570159x14666160509123042] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/16/2016] [Accepted: 04/17/2016] [Indexed: 01/22/2023] Open
Abstract
Background Chronic pain is a major symptom that develops in cancer patients, most commonly emerging during advanced stages of the disease. The nature of cancer-induced pain is complex, and the efficacy of current therapeutic interventions is restricted by the dose-limiting side-effects that accompany common centrally targeted analgesics. Methods This review focuses on how up-regulated glutamate production and export by the tumour converge at peripheral afferent nerve terminals to transmit nociceptive signals through the transient receptor cation channel, TRPV1, thereby initiating central sensitization in response to peripheral disease-mediated stimuli. Results Cancer cells undergo numerous metabolic changes that include increased glutamine catabolism and over-expression of enzymes involved in glutaminolysis, including glutaminase. This mitochondrial enzyme mediates glutaminolysis, producing large pools of intracellular glutamate. Up-regulation of the plasma membrane cystine/glutamate antiporter, system xc-, promotes aberrant glutamate release from cancer cells. Increased levels of extracellular glutamate have been associated with the progression of cancer-induced pain and we discuss how this can be mediated by activation of TRPV1. Conclusion With a growing population of patients receiving inadequate treatment for intractable pain, new targets need to be considered to better address this largely unmet clinical need for improving their quality of life. A better understanding of the mechanisms that underlie the unique qualities of cancer pain will help to identify novel targets that are able to limit the initiation of pain from a peripheral source–the tumour.
Collapse
Affiliation(s)
| | | | - Gurmit Singh
- Department of Pathology and Molecular Medicine; Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON. Canada
| |
Collapse
|
29
|
The cystine/glutamate antiporter system xc- drives breast tumor cell glutamate release and cancer-induced bone pain. Pain 2017; 157:2605-2616. [PMID: 27482630 PMCID: PMC5065056 DOI: 10.1097/j.pain.0000000000000681] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Supplemental Digital Content is Available in the Text. Tumor-derived glutamate may significantly contribute to cancer-induced bone pain. The glutamate transporter system xc− is a promising therapeutic target in this pain state. Bone is one of the leading sites of metastasis for frequently diagnosed malignancies, including those arising in the breast, prostate and lung. Although these cancers develop unnoticed and are painless in their primary sites, bone metastases result in debilitating pain. Deeper investigation of this pain may reveal etiology and lead to early cancer detection. Cancer-induced bone pain (CIBP) is inadequately managed with current standard-of-care analgesics and dramatically diminishes patient quality of life. While CIBP etiology is multifaceted, elevated levels of glutamate, an excitatory neurotransmitter, in the bone-tumor microenvironment may drive maladaptive nociceptive signaling. Here, we establish a relationship between the reactive nitrogen species peroxynitrite, tumor-derived glutamate, and CIBP. In vitro and in a syngeneic in vivo model of breast CIBP, murine mammary adenocarcinoma cells significantly elevated glutamate via the cystine/glutamate antiporter system xc−. The well-known system xc− inhibitor sulfasalazine significantly reduced levels of glutamate and attenuated CIBP-associated flinching and guarding behaviors. Peroxynitrite, a highly reactive species produced in tumors, significantly increased system xc− functional expression and tumor cell glutamate release. Scavenging peroxynitrite with the iron and mangano-based porphyrins, FeTMPyP and SRI10, significantly diminished tumor cell system xc− functional expression, reduced femur glutamate levels and mitigated CIBP. In sum, we demonstrate how breast cancer bone metastases upregulate a cystine/glutamate co-transporter to elevate extracellular glutamate. Pharmacological manipulation of peroxynitrite or system xc− attenuates CIBP, supporting a role for tumor-derived glutamate in CIBP and validating the targeting of system xc− as a novel therapeutic strategy for the management of metastatic bone pain.
Collapse
|
30
|
The Src family kinase inhibitor dasatinib delays pain-related behaviour and conserves bone in a rat model of cancer-induced bone pain. Sci Rep 2017; 7:4792. [PMID: 28684771 PMCID: PMC5500481 DOI: 10.1038/s41598-017-05029-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/23/2017] [Indexed: 12/18/2022] Open
Abstract
Pain is a severe and debilitating complication of metastatic bone cancer. Current analgesics do not provide sufficient pain relief for all patients, creating a great need for new treatment options. The Src kinase, a non-receptor protein tyrosine kinase, is implicated in processes involved in cancer-induced bone pain, including cancer growth, osteoclastic bone degradation and nociceptive signalling. Here we investigate the role of dasatinib, an oral Src kinase family and Bcr-Abl tyrosine kinase inhibitor, in an animal model of cancer-induced bone pain. Daily administration of dasatinib (15 mg/kg, p.o.) from day 7 after inoculation of MRMT-1 mammary carcinoma cells significantly attenuated movement-evoked and non-evoked pain behaviour in cancer-bearing rats. Radiographic - and microcomputed tomographic analyses showed significantly higher relative bone density and considerably preserved bone micro-architecture in the dasatinib treated groups, suggesting a bone-preserving effect. This was supported by a significant reduction of serum TRACP 5b levels in cancer-bearing rats treated with 15 mg/kg dasatinib. Furthermore, immunoblotting of lumbar spinal segments showed an increased activation of Src but not the NMDA receptor subunit 2B. These findings support a role of dasatinib as a disease modifying drug in pain pathologies characterized by increased osteoclast activity, such as bone metastases.
Collapse
|
31
|
Balza E, Castellani P, Moreno PS, Piccioli P, Medraño-Fernandez I, Semino C, Rubartelli A. Restoring microenvironmental redox and pH homeostasis inhibits neoplastic cell growth and migration: therapeutic efficacy of esomeprazole plus sulfasalazine on 3-MCA-induced sarcoma. Oncotarget 2017; 8:67482-67496. [PMID: 28978047 PMCID: PMC5620187 DOI: 10.18632/oncotarget.18713] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/22/2017] [Indexed: 01/25/2023] Open
Abstract
Neoplastic cells live in a stressful context and survive thanks to their ability to overcome stress. Thus, tumor cell responses to stress are potential therapeutic targets. We selected two such responses in melanoma and sarcoma cells: the xc- antioxidant system, that opposes oxidative stress, and surface v-ATPases that counteract the low pHi by extruding protons, and targeted them with the xc- blocker sulfasalazine and the proton pump inhibitor esomeprazole. Sulfasalazine inhibited the cystine/cysteine redox cycle and esomeprazole decreased pHi while increasing pHe in tumor cell lines. Although the single treatment with either drug slightly inhibited cell proliferation and motility, the association of sulfasalazine and esomeprazole powerfully decreased sarcoma and melanoma growth and migration. In the 3-methylcholanthrene (3-MCA)-induced sarcoma model, the combined therapy strongly reduced the tumor burden and increased the survival time: notably, 22 % of double-treated mice recovered and survived off therapy. Tumor-associated macrophages (TAM) displaying M2 markers, that abundantly infiltrate sarcoma and melanoma, overexpress xc- and membrane v-ATPases and were drastically decreased in tumors from mice undergone the combined therapy. Thus, the double targeting of tumor cells and macrophages by sulfasalazine and esomeprazole has a double therapeutic effect, as decreasing TAM infiltration deprives tumor cells of a crucial allied. Sulfasalazine and esomeprazole may therefore display unexpected therapeutic values, especially in case of hard-to-treat cancers.
Collapse
Affiliation(s)
- Enrica Balza
- Cell Biology Unit, IRCCS AOU San Martino - IST, 16132 Genoa, Italy
| | | | - Paola Sanchez Moreno
- Cell Biology Unit, IRCCS AOU San Martino - IST, 16132 Genoa, Italy.,Present address: Nanobiointeractions and Nanodiagnostics, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | | | - Iria Medraño-Fernandez
- Protein Transport Unit, Division of Cell and Molecular Biology, San Raffaele Institute, 20132 Milan, Italy
| | - Claudia Semino
- Protein Transport Unit, Division of Cell and Molecular Biology, San Raffaele Institute, 20132 Milan, Italy
| | - Anna Rubartelli
- Cell Biology Unit, IRCCS AOU San Martino - IST, 16132 Genoa, Italy
| |
Collapse
|
32
|
Rat model of cancer-induced bone pain: changes in nonnociceptive sensory neurons in vivo. Pain Rep 2017; 2:e603. [PMID: 29392218 PMCID: PMC5741358 DOI: 10.1097/pr9.0000000000000603] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/15/2017] [Accepted: 04/25/2017] [Indexed: 01/31/2023] Open
Abstract
Nonnociceptive sensory neurons relate to transient episodes of intense pain that characterize neuropathic pain. They are involved in the peripheral sensitization and tactile hypersensitivity. Introduction: Clinical data on cancer-induced bone pain (CIBP) suggest extensive changes in sensory function. In a previous investigation of an animal model of CIBP, we have observed that changes in intrinsic membrane properties and excitability of dorsal root ganglion (DRG) nociceptive neurons correspond to mechanical allodynia and hyperalgesia. Objectives: To investigate the mechanisms underlying changes in nonnociceptive sensory neurons in this model, we have compared the electrophysiological properties of primary nonnociceptive sensory neurons at <1 and >2 weeks after CIBP model induction with properties in sham control animals. Methods: Copenhagen rats were injected with 106 MAT-LyLu rat prostate cancer cells into the distal femur epiphysis to generate a model of CIBP. After von Frey tactile measurement of mechanical withdrawal thresholds, the animals were prepared for acute electrophysiological recordings of mechanically sensitive neurons in the DRG in vivo. Results: The mechanical withdrawal threshold progressively decreased in CIBP model rats. At <1 week after model induction, there were no changes observed in nonnociceptive Aβ-fiber DRG neurons between CIBP model rats and sham rats. However, at >2 weeks, the Aβ-fiber low-threshold mechanoreceptors (LTMs) in CIBP model rats exhibited a slowing of the dynamics of action potential (AP) genesis, including wider AP duration and lower AP amplitude compared with sham rats. Furthermore, enhanced excitability of Aβ-fiber LTM neurons was observed as an excitatory discharge in response to intracellular injection of depolarizing current into the soma. Conclusion: After induction of the CIBP model, Aβ-fiber LTMs at >2 weeks but not <1 week had undergone changes in electrophysiological properties. Importantly, changes observed are consistent with observations in models of peripheral neuropathy. Thus, Aβ-fiber nonnociceptive primary sensory neurons might be involved in the peripheral sensitization and tumor-induced tactile hypersensitivity in CIBP.
Collapse
|
33
|
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: 2.4] [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
|
34
|
Nashed MG, Ungard RG, Young K, Zacal NJ, Seidlitz EP, Fazzari J, Frey BN, Singh G. Behavioural Effects of Using Sulfasalazine to Inhibit Glutamate Released by Cancer Cells: A Novel target for Cancer-Induced Depression. Sci Rep 2017; 7:41382. [PMID: 28120908 PMCID: PMC5264609 DOI: 10.1038/srep41382] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 12/19/2016] [Indexed: 12/11/2022] Open
Abstract
Despite the lack of robust evidence of effectiveness, current treatment options for cancer-induced depression (CID) are limited to those developed for non-cancer related depression. Here, anhedonia-like and coping behaviours were assessed in female BALB/c mice inoculated with 4T1 mammary carcinoma cells. The behavioural effects of orally administered sulfasalazine (SSZ), a system xc− inhibitor, were compared with fluoxetine (FLX). FLX and SSZ prevented the development of anhedonia-like behaviour on the sucrose preference test (SPT) and passive coping behaviour on the forced swim test (FST). The SSZ metabolites 5-aminosalicylic acid (5-ASA) and sulfapyridine (SP) exerted an effect on the SPT but not on the FST. Although 5-ASA is a known anti-inflammatory agent, neither treatment with SSZ nor 5-ASA/SP prevented tumour-induced increases in serum levels of interleukin-1β (IL-1β) and IL-6, which are indicated in depressive disorders. Thus, the observed antidepressant-like effect of SSZ may primarily be attributable to the intact form of the drug, which inhibits system xc−. This study represents the first attempt at targeting cancer cells as a therapeutic strategy for CID, rather than targeting downstream effects of tumour burden on the central nervous system. In doing so, we have also begun to characterize the molecular pathways of CID.
Collapse
Affiliation(s)
- Mina G Nashed
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| | - Robert G Ungard
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| | - Kimberly Young
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| | - Natalie J Zacal
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| | - Eric P Seidlitz
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| | - Jennifer Fazzari
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| | - Benicio N Frey
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, L8N 3K7, Canada.,Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare Hamilton, ON, L8P 3K7, Canada
| | - Gurmit Singh
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
35
|
Yazdani A, Janzen N, Czorny S, Ungard RG, Miladinovic T, Singh G, Valliant JF. Preparation of tetrazine-containing [2 + 1] complexes of 99mTc and in vivo targeting using bioorthogonal inverse electron demand Diels–Alder chemistry. Dalton Trans 2017. [DOI: 10.1039/c7dt01497j] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A new 99mTc-labelled tetrazine for targeted imaging using bioorthogonal chemistry was developed and evaluated in vivo using a trans-cyclooctene derived bisphosphonate targeting regions of high bone turnover and bone lesions.
Collapse
Affiliation(s)
- Abdolreza Yazdani
- Department of Chemistry and Chemical Biology
- McMaster University
- Hamilton
- Canada
| | - Nancy Janzen
- Department of Chemistry and Chemical Biology
- McMaster University
- Hamilton
- Canada
| | - Shannon Czorny
- Centre for Probe Development and Commercialization
- Hamilton
- Canada
| | - Robert G. Ungard
- Department of Pathology and Molecular Medicine
- McMaster University
- Hamilton
- Canada
| | - Tanya Miladinovic
- Department of Pathology and Molecular Medicine
- McMaster University
- Hamilton
- Canada
| | - Gurmit Singh
- Department of Pathology and Molecular Medicine
- McMaster University
- Hamilton
- Canada
| | - John F. Valliant
- Department of Chemistry and Chemical Biology
- McMaster University
- Hamilton
- Canada
- Centre for Probe Development and Commercialization
| |
Collapse
|
36
|
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: 1.4] [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
|
37
|
Glutamate Transport System as a Novel Therapeutic Target in Chronic Pain: Molecular Mechanisms and Pharmacology. ADVANCES IN NEUROBIOLOGY 2017; 16:225-253. [PMID: 28828613 DOI: 10.1007/978-3-319-55769-4_11] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The vast majority of peripheral neurons sensing noxious stimuli and conducting pain signals to the dorsal horn of the spinal cord utilize glutamate as a chemical transmitter of excitation. High-affinity glutamate transporter subtypes GLAST/EAAT1, GLT1/EAAT2, EAAC1/EAAT3, and EAAT4, differentially expressed on sensory neurons, postsynaptic spinal interneurons, and neighboring glia, ensure fine modulation of glutamate neurotransmission in the spinal cord. The glutamate transport system seems to play important roles in molecular mechanisms underlying chronic pain and analgesia. Downregulation of glutamate transporters (GluTs) often precedes or occurs simultaneously with development of hypersensitivity to thermal or tactile stimuli in various models of chronic pain. Moreover, antisense knockdown or pharmacological inhibition of these membrane proteins can induce or aggravate pain. In contrast, upregulation of GluTs by positive pharmacological modulators or by viral gene transfer to the spinal cord can reverse the development of such pathological hypersensitivity. Furthermore, some multi-target drugs displaying analgesic properties (e.g., tricyclic antidepressant amitriptyline, riluzole, anticonvulsant valproate, tetracycline antibiotic minocycline, β-lactam antibiotic ceftriaxone and its structural analog devoid of antibacterial activity, clavulanic acid) can significantly increase the spinal glutamate uptake. Thus, mounting evidence points at GluTs as prospective therapeutic target for chronic pain treatment. However, design and development of new analgesics based on the modulation of glutamate uptake will require more precise knowledge of molecular mechanisms underlying physiological or aberrant functioning of this transport system in the spinal cord.
Collapse
|
38
|
Linher-Melville K, Nashed MG, Ungard RG, Haftchenary S, Rosa DA, Gunning PT, Singh G. Chronic Inhibition of STAT3/STAT5 in Treatment-Resistant Human Breast Cancer Cell Subtypes: Convergence on the ROS/SUMO Pathway and Its Effects on xCT Expression and System xc- Activity. PLoS One 2016; 11:e0161202. [PMID: 27513743 PMCID: PMC4981357 DOI: 10.1371/journal.pone.0161202] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 08/01/2016] [Indexed: 12/15/2022] Open
Abstract
Pharmacologically targeting activated STAT3 and/or STAT5 has been an active area of cancer research. The cystine/glutamate antiporter, system xc-, contributes to redox balance and export of intracellularly produced glutamate in response to up-regulated glutaminolysis in cancer cells. We have previously shown that blocking STAT3/5 using the small molecule inhibitor, SH-4-54, which targets the SH2 domains of both proteins, increases xCT expression, thereby increasing system xc- activity in human breast cancer cells. The current investigation demonstrates that chronic SH-4-54 administration, followed by clonal selection of treatment-resistant MDA-MB-231 and T47D breast cancer cells, elicits distinct subtype-dependent effects. xCT mRNA and protein levels, glutamate release, and cystine uptake are decreased relative to untreated passage-matched controls in triple-negative MDA-MB-231 cells, with the inverse occurring in estrogen-responsive T47D cells. This “ying-yang” effect is linked with a shifted balance between the phosphorylation status of STAT3 and STAT5, intracellular ROS levels, and STAT5 SUMOylation/de-SUMOylation. STAT5 emerged as a definitive negative regulator of xCT at the transcriptional level, while STAT3 activation is coupled with increased system xc- activity. We propose that careful classification of a patient’s breast cancer subtype is central to effectively targeting STAT3/5 as a therapeutic means of treating breast cancer, particularly given that xCT is emerging as an important biomarker of aggressive cancers.
Collapse
Affiliation(s)
- Katja Linher-Melville
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Mina G. Nashed
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Robert G. Ungard
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
| | - Sina Haftchenary
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, L5L 1C6, Canada
| | - David A. Rosa
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, L5L 1C6, Canada
| | - Patrick T. Gunning
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, L5L 1C6, Canada
| | - Gurmit Singh
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, L8S 4L8, Canada
- * E-mail:
| |
Collapse
|
39
|
Abstract
Bone metastatic disease remains a significant and frequent problem for cancer patients that can lead to increased morbidity and mortality. Unfortunately, despite decades of research, bone metastases remain incurable. Current studies have demonstrated that many properties and cell types within the bone and bone marrow microenvironment contribute to tumor-induced bone disease. Furthermore, they have pointed to the importance of understanding how tumor cells interact with their microenvironment in order to help improve both the development of new therapeutics and the prediction of response to therapy.
Collapse
Affiliation(s)
- Denise Buenrostro
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA
- Center for Bone Biology, Vanderbilt University, 2215B Garland Avenue, 1235 MRBIV, Nashville, TN 37232, USA
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Patrick L. Mulcrone
- Center for Bone Biology, Vanderbilt University, 2215B Garland Avenue, 1235 MRBIV, Nashville, TN 37232, USA
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Philip Owens
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Julie A. Sterling
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA
- Center for Bone Biology, Vanderbilt University, 2215B Garland Avenue, 1235 MRBIV, Nashville, TN 37232, USA
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, 2215B Garland Avenue, 1235 MRBIV, Nashville, TN 37232, USA
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
40
|
Zhu YF, Ungard R, Seidlitz E, Zacal N, Huizinga J, Henry JL, Singh G. Differences in electrophysiological properties of functionally identified nociceptive sensory neurons in an animal model of cancer-induced bone pain. Mol Pain 2016; 12:12/0/1744806916628778. [PMID: 27030711 PMCID: PMC4994860 DOI: 10.1177/1744806916628778] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 12/04/2015] [Indexed: 12/31/2022] Open
Abstract
Background Bone cancer pain is often severe, yet little is known about mechanisms generating this type of chronic pain. While previous studies have identified functional alterations in peripheral sensory neurons that correlate with bone tumours, none has provided direct evidence correlating behavioural nociceptive responses with properties of sensory neurons in an intact bone cancer model. Results In a rat model of prostate cancer-induced bone pain, we confirmed tactile hypersensitivity using the von Frey test. Subsequently, we recorded intracellularly from dorsal root ganglion neurons in vivo in anesthetized animals. Neurons remained connected to their peripheral receptive terminals and were classified on the basis of action potential properties, responses to dorsal root stimulation, and to mechanical stimulation of the respective peripheral receptive fields. Neurons included C-, Aδ-, and Aβ-fibre nociceptors, identified by their expression of substance P. We suggest that bone tumour may induce phenotypic changes in peripheral nociceptors and that these could contribute to bone cancer pain. Conclusions This work represents a significant technical and conceptual advance in the study of peripheral nociceptor functions in the development of cancer-induced bone pain. This is the first study to report that changes in sensitivity and excitability of dorsal root ganglion primary afferents directly correspond to mechanical allodynia and hyperalgesia behaviours following prostate cancer cell injection into the femur of rats. Furthermore, our unique combination of techniques has allowed us to follow, in a single neuron, mechanical pain-related behaviours, electrophysiological changes in action potential properties, and dorsal root substance P expression. These data provide a more complete understanding of this unique pain state at the cellular level that may allow for future development of mechanism-based treatments for cancer-induced bone pain.
Collapse
Affiliation(s)
- Yong Fang Zhu
- Michael G. DeGroote Institute for Pain Research and Care, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Robert Ungard
- Michael G. DeGroote Institute for Pain Research and Care, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Eric Seidlitz
- Michael G. DeGroote Institute for Pain Research and Care, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Natalie Zacal
- Michael G. DeGroote Institute for Pain Research and Care, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jan Huizinga
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - James L Henry
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Gurmit Singh
- Michael G. DeGroote Institute for Pain Research and Care, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
41
|
Laux-Biehlmann A, Boyken J, Dahllöf H, Schmidt N, Zollner TM, Nagel J. Dynamic weight bearing as a non-reflexive method for the measurement of abdominal pain in mice. Eur J Pain 2015; 20:742-52. [PMID: 26684879 DOI: 10.1002/ejp.800] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Chronic pelvic pain (CPP) is a high burden for patients and society. It affects 15-24% of women in reproductive age and is an area of high unmet medical need. CPP can be caused by a wide range of visceral diseases such as abdominal infections, gastrointestinal or gynaecological diseases like endometriosis. Despite the high medical need for this condition, pharmacological approaches are hampered by the limited number of available methods for the behavioural evaluation of pain in inflammation-driven animal models of pelvic pain. METHODS The dynamic weight bearing (DWB) system was used for the evaluation of spontaneous behaviour changes in the zymosan-induced peritonitis mouse model. Inflammatory mediator levels were evaluated in peritoneal lavage and their correlation with the behavioural endpoints was assessed. We evaluated the effect on behavioural endpoints of the selective cyclooxygenase-2 (COX-2) inhibitor celecoxib and the Nav 1.8 blocker A-803467. RESULTS The presence of a relief posture, characterized by a significantly increased weight distribution towards the front paws, was observed following intraperitoneal injection of zymosan. A positive correlation was detected between PGE2 levels in the peritoneal lavage and DWB endpoints. In addition, zymosan-induced weight bearing changes were reverted by celecoxib and A-803467. CONCLUSIONS This study described for the first time the use of DWB as a non-subjective and non-reflexive method for the evaluation of inflammatory-driven abdominal pain in a mouse model.
Collapse
Affiliation(s)
- A Laux-Biehlmann
- Global Drug Discovery, Global Therapeutic Research Groups, Gynecological Therapies, Bayer Healthcare, Berlin, Germany
| | - J Boyken
- Global Drug Discovery, Global Therapeutic Research Groups, Gynecological Therapies, Bayer Healthcare, Berlin, Germany
| | - H Dahllöf
- Global Drug Discovery, Global Therapeutic Research Groups, Gynecological Therapies, Bayer Healthcare, Berlin, Germany
| | - N Schmidt
- Global Drug Discovery, Global Therapeutic Research Groups, Gynecological Therapies, Bayer Healthcare, Berlin, Germany
| | - T M Zollner
- Global Drug Discovery, Global Therapeutic Research Groups, Gynecological Therapies, Bayer Healthcare, Berlin, Germany
| | - J Nagel
- Global Drug Discovery, Global Therapeutic Research Groups, Gynecological Therapies, Bayer Healthcare, Berlin, Germany
| |
Collapse
|
42
|
Miladinovic T, Nashed MG, Singh G. Overview of Glutamatergic Dysregulation in Central Pathologies. Biomolecules 2015; 5:3112-41. [PMID: 26569330 PMCID: PMC4693272 DOI: 10.3390/biom5043112] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 11/03/2015] [Accepted: 11/05/2015] [Indexed: 12/27/2022] Open
Abstract
As the major excitatory neurotransmitter in the mammalian central nervous system, glutamate plays a key role in many central pathologies, including gliomas, psychiatric, neurodevelopmental, and neurodegenerative disorders. Post-mortem and serological studies have implicated glutamatergic dysregulation in these pathologies, and pharmacological modulation of glutamate receptors and transporters has provided further validation for the involvement of glutamate. Furthermore, efforts from genetic, in vitro, and animal studies are actively elucidating the specific glutamatergic mechanisms that contribute to the aetiology of central pathologies. However, details regarding specific mechanisms remain sparse and progress in effectively modulating glutamate to alleviate symptoms or inhibit disease states has been relatively slow. In this report, we review what is currently known about glutamate signalling in central pathologies. We also discuss glutamate's mediating role in comorbidities, specifically cancer-induced bone pain and depression.
Collapse
Affiliation(s)
- Tanya Miladinovic
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada.
| | - Mina G Nashed
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada.
| | - Gurmit Singh
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada.
| |
Collapse
|
43
|
Slosky LM, Largent-Milnes TM, Vanderah TW. Use of Animal Models in Understanding Cancer-induced Bone Pain. CANCER GROWTH AND METASTASIS 2015; 8:47-62. [PMID: 26339191 PMCID: PMC4552039 DOI: 10.4137/cgm.s21215] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/14/2015] [Accepted: 06/16/2015] [Indexed: 12/13/2022]
Abstract
Many common cancers have a propensity to metastasize to bone. Although malignancies often go undetected in their native tissues, bone metastases produce excruciating pain that severely compromises patient quality of life. Cancer-induced bone pain (CIBP) is poorly managed with existing medications, and its multifaceted etiology remains to be fully elucidated. Novel analgesic targets arise as more is learned about this complex and distinct pain state. Over the past two decades, multiple animal models have been developed to study CIBP’s unique pathology and identify therapeutic targets. Here, we review animal models of CIBP and the mechanistic insights gained as these models evolve. Findings from immunocompromised and immunocompetent host systems are discussed separately to highlight the effect of model choice on outcome. Gaining an understanding of the unique neuromolecular profile of cancer pain through the use of appropriate animal models will aid in the development of more effective therapeutics for CIBP.
Collapse
Affiliation(s)
- Lauren M Slosky
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Tally M Largent-Milnes
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Todd W Vanderah
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, USA
| |
Collapse
|
44
|
Habib E, Linher-Melville K, Lin HX, Singh G. Expression of xCT and activity of system xc(-) are regulated by NRF2 in human breast cancer cells in response to oxidative stress. Redox Biol 2015; 5:33-42. [PMID: 25827424 PMCID: PMC4392061 DOI: 10.1016/j.redox.2015.03.003] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 03/13/2015] [Accepted: 03/16/2015] [Indexed: 12/20/2022] Open
Abstract
Cancer cells adapt to high levels of oxidative stress in order to survive and proliferate by activating key transcription factors. One such master regulator, the redox sensitive transcription factor NF E2 Related Factor 2 (NRF2), controls the expression of cellular defense genes including those encoding intracellular redox-balancing proteins involved in glutathione (GSH) synthesis. Under basal conditions, Kelch-like ECH-associated protein 1 (KEAP1) targets NRF2 for ubiquitination. In response to oxidative stress, NRF2 dissociates from KEAP1, entering the nucleus and binding to the antioxidant response element (ARE) in the promoter of its target genes. Elevated reactive oxygen species (ROS) production may deplete GSH levels within cancer cells. System xc(-), an antiporter that exports glutamate while importing cystine to be converted into cysteine for GSH synthesis, is upregulated in cancer cells in response to oxidative stress. Here, we provided evidence that the expression of xCT, the light chain subunit of system xc(-), is regulated by NRF2 in representative human breast cancer cells. Hydrogen peroxide (H2O2) treatment increased nuclear translocation of NRF2, also increasing levels of xCT mRNA and protein and extracellular glutamate release. Overexpression of NRF2 up-regulated the activity of the xCT promoter, which contains a proximal ARE. In contrast, overexpression of KEAP1 repressed promoter activity and decreased xCT protein levels, while siRNA knockdown of KEAP1 up-regulated xCT protein levels and transporter activity. These results demonstrate the importance of the KEAP1/NRF2 pathway in balancing oxidative stress in breast cancer cells through system xc(-). We have previously shown that xCT is upregulated in various cancer cell lines under oxidative stress. In the current investigation, we focused on MCF-7 cells as a model for mechanistic studies.
Collapse
Affiliation(s)
- Eric Habib
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada L8S 4L8
| | - Katja Linher-Melville
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada L8S 4L8
| | - Han-Xin Lin
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada L8S 4L8
| | - Gurmit Singh
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada L8S 4L8.
| |
Collapse
|
45
|
Inhibitors of glutamate release from breast cancer cells; new targets for cancer-induced bone-pain. Sci Rep 2015; 5:8380. [PMID: 25670024 PMCID: PMC4323637 DOI: 10.1038/srep08380] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 01/19/2015] [Indexed: 01/20/2023] Open
Abstract
Glutamate is an important signaling molecule in a wide variety of tissues. Aberrant glutamatergic signaling disrupts normal tissue homeostasis and induces several disruptive pathological conditions including pain. Breast cancer cells secrete high levels of glutamate and often metastasize to bone. Exogenous glutamate can disrupt normal bone turnover and may be responsible for cancer-induced bone pain (CIBP). CIBP is a significant co-morbidity that affects quality of life for many advanced-stage breast cancer patients. Current treatment options are commonly accompanied by serious side-effects that negatively impact patient care. Identifying small molecule inhibitors of glutamate release from aggressive breast cancer cells advances a novel, mechanistic approach to targeting CIBP that could advance treatment for several pathological conditions. Using high-throughput screening, we investigated the ability of approximately 30,000 compounds from the Canadian Compound Collection to reduce glutamate release from MDA-MB-231 breast cancer cells. This line is known to secrete high levels of glutamate and has been demonstrated to induce CIBP by this mechanism. Positive chemical hits were based on the potency of each molecule relative to a known pharmacological inhibitor of glutamate release, sulfasalazine. Efficacy was confirmed and drug-like molecules were identified as potent inhibitors of glutamate secretion from MDA-MB-231, MCF-7 and Mat-Ly-Lu cells.
Collapse
|
46
|
Gegelashvili G, Bjerrum OJ. High-affinity glutamate transporters in chronic pain: an emerging therapeutic target. J Neurochem 2014; 131:712-30. [DOI: 10.1111/jnc.12957] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 09/18/2014] [Accepted: 09/25/2014] [Indexed: 01/13/2023]
Affiliation(s)
- Georgi Gegelashvili
- Department of Drug Design and Pharmacology; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
- Institute of Chemical Biology; Ilia State University; Tbilisi Georgia
| | - Ole J. Bjerrum
- Department of Drug Design and Pharmacology; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| |
Collapse
|
47
|
Gordh T. GTP-cyclohydrolase 1 genetics and tetrahydrobiopterin—Modulators of pain hypersensitivity? Scand J Pain 2014; 5:119-120. [DOI: 10.1016/j.sjpain.2014.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Torsten Gordh
- Multidisciplinary Pain Center , Uppsala University Hospital , Uppsala , Sweden
| |
Collapse
|
48
|
|