1
|
Scerba MT, Tweedie D, Greig NH. 2-(Piperidin-3-yl)phthalimides reduce classical markers of cellular inflammation in LPS-challenged RAW 264.7 cells and also demonstrate potentially relevant sigma and serotonin receptor affinity in membrane preparations. Bioorg Med Chem Lett 2024; 110:129885. [PMID: 38996940 PMCID: PMC11479674 DOI: 10.1016/j.bmcl.2024.129885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024]
Abstract
Herein, we report the synthesis of new 4-amino-2-(piperidin-3-yl)isoindoline-1,3-diones and their biological evaluation in a series of in vitro experiments. The synthetic production of these materials was initiated upon the condensation of appropriate nitrophthalic acid derivatives with various 3-aminopiperidines; subsequent reduction provided the final products in moderate to good yields. Readily available chiral pool reagents facilitated entry into optically enriched samples, while the piperidine scaffold furnished a variety of amide and alkylated entries. In total, 16 candidates were produced, and their ensuing treatment in LPS-challenged RAW cells effected slight reductions in secreted TNF-α but provided more robust and dose-dependent declines in nitrite and IL-6 levels relative to basal amounts, all concurrent with maintenance of cellular viability across the concentration ranges screened. The secondary amine cohort including rac-6, (R)-7, and (S)-8 rendered the most pronounced dose-dependent reductions in nitrite and IL-6. When dosed at 30 μM, (R)-7 demonstrated the most compelling effects, with decreases of 32 % and 40 % for nitrite and IL-6, respectively. Notable reductions in the inflammatory markers were also observed for 19 which effected declines in TNF-α (14 %), nitrite (19 %), and IL-6 (11 %) when treated at 30 μM. Additionally, four representative compounds were further evaluated against numerous CNS receptors, channels, and transporters, with 6, 9, and 19 demonstrating varying degrees of nanomolar-to-low-micromolar binding to the σ-1 and σ-2 receptors and also to serotonin receptors 5HT2A, 5HT2B and 5HT3. In this regard, 6 displayed perhaps the most noteworthy affinities, with binding at σ-2 (Ki = 2.2uM), 5HT2B (Ki = 561 nM) and 5HT3 (Ki = 536 nM). Furthermore, no pronounced or dose-dependent Cereblon/DDB1 binding was observed for the screened representative compounds 6, 9, 18 and 19.
Collapse
Affiliation(s)
- Michael T Scerba
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA.
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| |
Collapse
|
2
|
Li J, Shen H, Guo LW. Transmembrane protein TMEM97 and epigenetic reader BAHCC1 constitute an axis that supports pro-inflammatory cytokine expression. Cell Signal 2024; 116:111069. [PMID: 38290642 PMCID: PMC10997414 DOI: 10.1016/j.cellsig.2024.111069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/15/2024] [Accepted: 01/26/2024] [Indexed: 02/01/2024]
Abstract
Pro-inflammatory cytokine production by the retinal pigment epithelium (RPE) is a key etiology in retinal degenerative diseases, yet the underlying mechanisms are not well understood. TMEM97 is a scarcely studied transmembrane protein recently implicated in retinal degeneration. BAH domain coiled coil 1 (BAHCC1) is a newly discovered histone code reader involved in oncogenesis. A role for TMEM97 and BAHCC1 in RPE inflammation was not known. Here we found that they constitute a novel axis regulating pro-inflammatory cytokine expression in RPE cells. Transcriptomic analysis using a TMEM97-/- ARPE19 human cell line and the validation via TMEM97 loss- and gain-of-function revealed a profound role of TMEM97 in promoting the expression of pro-inflammatory cytokines, notably IL1β and CCL2, and unexpectedly BAHCC1 as well. Moreover, co-immunoprecipitation indicated an association between the TMEM97 and BAHCC1 proteins. While TMEM97 ablation decreased and its overexpression increased NFκB (p50, p52, p65), the master transcription factor for pro-inflammatory cytokines, silencing BAHCC1 down-regulated NFκB and downstream pro-inflammatory cytokines. Furthermore, in an RPE-damage retinal degeneration mouse model, immunofluorescence illustrated down-regulation of IL1β and CCL2 total proteins and suppression of glial activation in the retina of Tmem97-/- mice compared to Tmem97+/+ mice. Thus, TMEM97 is a novel determinant of pro-inflammatory cytokine expression acting via a previously unknown TMEM97- > BAHCC1- > NFκB cascade. SYNOPSIS: Retinal pigment epithelium (RPE) inflammation can lead to blindness. We identify here a previously uncharacterized cascade that underlies RPE cell production of pro-inflammatory cytokines. Specifically, transmembrane protein TMEM97 positively regulates the recently discovered histone code reader BAHCC1, which in turn enhances pro-inflammatory cytokine expression via the transcription factor NFκB.
Collapse
Affiliation(s)
- Jing Li
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Hongtao Shen
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Lian-Wang Guo
- Division of Surgical Sciences, Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA; Department of Ophthalmology, University of Virginia, Charlottesville, VA 22908, USA; Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
3
|
Mackiewicz J, Lisek M, Boczek T. Targeting CaN/NFAT in Alzheimer's brain degeneration. Front Immunol 2023; 14:1281882. [PMID: 38077352 PMCID: PMC10701682 DOI: 10.3389/fimmu.2023.1281882] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a progressive loss of cognitive functions. While the exact causes of this debilitating disorder remain elusive, numerous investigations have characterized its two core pathologies: the presence of β-amyloid plaques and tau tangles. Additionally, multiple studies of postmortem brain tissue, as well as results from AD preclinical models, have consistently demonstrated the presence of a sustained inflammatory response. As the persistent immune response is associated with neurodegeneration, it became clear that it may also exacerbate other AD pathologies, providing a link between the initial deposition of β-amyloid plaques and the later development of neurofibrillary tangles. Initially discovered in T cells, the nuclear factor of activated T-cells (NFAT) is one of the main transcription factors driving the expression of inflammatory genes and thus regulating immune responses. NFAT-dependent production of inflammatory mediators is controlled by Ca2+-dependent protein phosphatase calcineurin (CaN), which dephosphorylates NFAT and promotes its transcriptional activity. A substantial body of evidence has demonstrated that aberrant CaN/NFAT signaling is linked to several pathologies observed in AD, including neuronal apoptosis, synaptic deficits, and glia activation. In view of this, the role of NFAT isoforms in AD has been linked to disease progression at different stages, some of which are paralleled to diminished cognitive status. The use of classical inhibitors of CaN/NFAT signaling, such as tacrolimus or cyclosporine, or adeno-associated viruses to specifically inhibit astrocytic NFAT activation, has alleviated some symptoms of AD by diminishing β-amyloid neurotoxicity and neuroinflammation. In this article, we discuss the recent findings related to the contribution of CaN/NFAT signaling to the progression of AD and highlight the possible benefits of targeting this pathway in AD treatment.
Collapse
Affiliation(s)
| | | | - Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
4
|
Dalwadi DA, Kim S, Schetz J, Schreihofer DA, Kim S. Brain-derived neurotrophic factor for high-throughput evaluation of selective Sigma-1 receptor ligands. J Pharmacol Toxicol Methods 2022; 113:107129. [PMID: 34678430 PMCID: PMC9358981 DOI: 10.1016/j.vascn.2021.107129] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/13/2021] [Accepted: 10/13/2021] [Indexed: 01/03/2023]
Abstract
The Sigma-1 receptor (S1R) is an endoplasmic reticulum (ER) chaperone protein that has been implicated in attenuating inflammatory stress-mediated brain injuries. Selective S1R agonists represent a new class of therapeutic agent for treating neuropsychiatric and neurodegenerative disorders, however, to date, no S1R ligand has been approved for therapeutic purposes. We used three potential methods on known and potential S1R ligands to develop an unambiguous high-throughput cell screen for S1R activity. We screened known and potential S1R ligands using radioligand binding and previously reported markers of S1R activity including BDNF release, modulation of IP3 mediated calcium release, and modulation of NGF-induced neurite sprouting. Here, we present results several prototypical S1R compounds and some compounds with the potential for drug repurposing. Using an in-situ ELISA approach we demonstrated that these compounds could stimulate S1R-mediated BDNF release, which is a valuable therapeutic property since BDNF plays a critical role in neuronal support. These compounds were classified as S1R agonists because the BDNF response was comparable to the prototypical agonist 4-PPBP and because it could be reversed by a S1R selective concentration of the antagonist BD1063. When modulation of IP3 mediated calcium response and NGF-induced neurite sprouting were used as a measure of S1R activation, we were unable to reproduce the published results and determined that they are not reliable measures for evaluating functional properties of S1R ligands.
Collapse
Affiliation(s)
- Dhwanil A Dalwadi
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; Department of Medicine, Division of Gastroenterology, Oregon Health Sciences University, Portland, OR 97239, USA
| | - Stephanie Kim
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; University of Texas Medical Branch at Galveston, School of Medicine, Galveston, TX 77555, USA
| | - John Schetz
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Derek A Schreihofer
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Seongcheol Kim
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; Department of Cellular and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153, USA.
| |
Collapse
|
5
|
Váczi S, Barna L, Harazin A, Mészáros M, Porkoláb G, Zvara Á, Ónody R, Földesi I, Veszelka S, Penke B, Fülöp L, Deli MA, Mezei Z. S1R agonist modulates rat platelet eicosanoid synthesis and aggregation. Platelets 2021; 33:709-718. [PMID: 34697991 DOI: 10.1080/09537104.2021.1981843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Sigma-1 receptor (S1R) is detected in different cell types and can regulate intracellular signaling pathways. S1R plays a role in the pathomechanism of diseases and the regulation of neurotransmitters. Fluvoxamine can bind to S1R and reduce the serotonin uptake of neurons and platelets. We therefore hypothesized that platelets express S1R, which can modify platelet function. The expression of the SIGMAR1 gene in rat platelets was examined with a reverse transcription polymerase chain reaction and a quantitative polymerase chain reaction. The receptor was also visualized by immunostaining and confocal laser scanning microscopy. The effect of S1R agonist PRE-084 on the eicosanoid synthesis of isolated rat platelets and ADP- and AA-induced platelet aggregation was examined. S1R was detected in rat platelets both at gene and protein levels. Pretreatment with PRE-084 of resting platelets induced elevation of eicosanoid synthesis. The rate of elevation in thromboxane B2 and prostaglandin D2 synthesis was similar, but the production of prostaglandin E2 was higher. The concentration-response curve showed a sigmoidal form. The most effective concentration of the agonist was 2 µM. PRE-084 increased the quantity of cyclooxygenase-1 as detected by ELISA. PRE-084 also elevated the ADP- and AA-induced platelet aggregation. S1R of platelets might regulate physiological or pathological functions.
Collapse
Affiliation(s)
- Sándor Váczi
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary.,Doctoral School of Theoretical Medicine, University of Szeged, Szeged, Hungary.,Gedeon Richter Talentum Foundation Scholarship, Budapest, Hungary
| | - L Barna
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary.,Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - A Harazin
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - M Mészáros
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - G Porkoláb
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary.,Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Á Zvara
- Institute of Genetics, Biological Research Centre, Szeged, Hungary
| | - R Ónody
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - I Földesi
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - S Veszelka
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - B Penke
- Department of Medical Chemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - L Fülöp
- Department of Medical Chemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - M A Deli
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - Z Mezei
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary.,Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| |
Collapse
|
6
|
Behensky AA, Katnik C, Yin H, Cuevas J. Activation of Sigma Receptors With Afobazole Modulates Microglial, but Not Neuronal, Apoptotic Gene Expression in Response to Long-Term Ischemia Exposure. Front Neurosci 2019; 13:414. [PMID: 31156357 PMCID: PMC6529844 DOI: 10.3389/fnins.2019.00414] [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: 02/26/2019] [Accepted: 04/11/2019] [Indexed: 12/14/2022] Open
Abstract
Stroke continues to be a leading cause of death and serious long-term disability. The lack of therapeutic options for treating stroke at delayed time points (≥6 h post-stroke) remains a challenge. The sigma receptor agonist, afobazole, an anxiolytic used clinically in Russia, has been shown to reduce neuronal and glial cell injury following ischemia and acidosis; both of which have been shown to play important roles following an ischemic stroke. However, the mechanism(s) responsible for this cytoprotection remain unknown. Experiments were carried out on isolated microglia from neonatal rats and cortical neurons from embryonic rats to gain further insight into these mechanisms. Prolonged exposure to in vitro ischemia resulted in microglial cell death, which was associated with increased expression of the pro-apoptotic protein, Bax, the death protease, caspase-3, and reduced expression in the anti-apoptotic protein Bcl-2. Incubation of cells with afobazole during ischemia decreased the number of microglia expressing both Bax and caspase-3, and increased cells expressing Bcl-2, which resulted in a concomitant enhancement in cell survival. In similar experiments, incubation of neurons under in vitro ischemic conditions resulted in higher expression of Bax and caspase-3, while at the same time expression of Bcl-2 was decreased. However, unlike observations made in microglial cells, afobazole was unable to modulate the expression of these apoptotic proteins, but a reduction in neuronal death was still noted. The functional state of surviving neurons was assessed by measuring metabolic activity, resting membrane potential, and responses to membrane depolarizations. Results showed that these neurons maintained membrane potential but had low metabolic activity and were unresponsive to membrane depolarizations. However, while these neurons were not fully functional, there was significant protection by afobazole against long-term ischemia-induced cell death. Thus, the effects of sigma receptor activation on microglial and neuronal responses to ischemia differ significantly.
Collapse
Affiliation(s)
- Adam A Behensky
- Department of Molecular Pharmacology and Physiology, College of Medicine, University of South Florida, Tampa, FL, United States
| | - Christopher Katnik
- Department of Molecular Pharmacology and Physiology, College of Medicine, University of South Florida, Tampa, FL, United States
| | - Huquan Yin
- Department of Molecular Pharmacology and Physiology, College of Medicine, University of South Florida, Tampa, FL, United States
| | - Javier Cuevas
- Department of Molecular Pharmacology and Physiology, College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
7
|
Intlekofer KA, Clements K, Woods H, Adams H, Suvorov A, Petersen SL. Progesterone receptor membrane component 1 inhibits tumor necrosis factor alpha induction of gene expression in neural cells. PLoS One 2019; 14:e0215389. [PMID: 31026287 PMCID: PMC6485904 DOI: 10.1371/journal.pone.0215389] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 04/01/2019] [Indexed: 12/31/2022] Open
Abstract
Progesterone membrane receptor component 1 (Pgrmc1) is a cytochrome b5-related protein with wide-ranging functions studied most extensively in non-neural tissues. We previously demonstrated that Pgrmc1 is widely distributed in the brain with highest expression in the limbic system. To determine Pgrmc1 functions in cells of these regions, we compared transcriptomes of control siRNA-treated and Pgrmc1 siRNA-treated N42 hypothalamic cells using whole genome microarrays. Our bioinformatics analyses suggested that Pgrmc1 plays a role in immune functions and likely regulates proinflammatory cytokine signaling. In follow-up studies, we showed that one of these cytokines, TNFα, increased expression of rtp4, ifit3 and gbp4, genes found on microarrays to be among the most highly upregulated by Pgrmc1 depletion. Moreover, either Pgrmc1 depletion or treatment with the Pgrmc1 antagonist, AG-205, increased both basal and TNFα-induced expression of these genes in N42 cells. TNFα had no effect on levels of Rtp4, Ifit3 or Gbp4 mRNAs in mHippoE-18 hippocampal control cells, but Pgrmc1 knock-down dramatically increased basal and TNFα-stimulated expression of these genes. P4 had no effect on gbp4, ifit3 or rtp4 expression or on the ability of Pgrmc1 to inhibit TNFα induction of these genes. However, a majority of the top upstream regulators of Pgrmc1 target genes were related to synthesis or activity of steroids, including P4, that exert neuroprotective effects. In addition, one of the identified Pgrmc1 targets was Nr4a1, an orphan receptor important for the synthesis of most steroidogenic molecules. Our findings indicate that Pgrmc1 may exert neuroprotective effects by suppressing TNFα-induced neuroinflammation and by regulating neurosteroid synthesis.
Collapse
Affiliation(s)
- Karlie A. Intlekofer
- Department of Veterinary and Animal Sciences, Institute of Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Kelsey Clements
- Department of Veterinary and Animal Sciences, Institute of Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Haley Woods
- Department of Veterinary and Animal Sciences, Institute of Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Hillary Adams
- Department of Veterinary and Animal Sciences, Institute of Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Alexander Suvorov
- Department of Environmental Health Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Sandra L. Petersen
- Department of Veterinary and Animal Sciences, Institute of Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| |
Collapse
|
8
|
Song YF, Liu DZ, Cheng Y, Teng ZH, Cui H, Liu M, Zhang BL, Mei QB, Zhou SY. Charge Reversible and Mitochondria/Nucleus Dual Target Lipid Hybrid Nanoparticles To Enhance Antitumor Activity of Doxorubicin. Mol Pharm 2018; 15:1296-1308. [PMID: 29432025 DOI: 10.1021/acs.molpharmaceut.7b01109] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The experiment aims to increase antitumor activity while decreasing the systemic toxicity of doxorubicin (DOX). Charge reversible and mitochondria/nucleus dual target lipid hybrid nanoparticles (LNPs) was prepared. The in vitro experimental results indicated that LNPs released more amount of DOX in acidic environment and delivered more amount of DOX to the mitochondria and nucleus of tumor cells than did free DOX, which resulted in the reduction of mitochondrial membrane potential and the enhancement of cytotoxicity of LNPs on tumor cells. Furthermore, the in vivo experimental results indicated that LNPs delivered more DOX to tumor tissue and significantly prolonged the retention time of DOX in tumor tissue as compared with free DOX, which consequently resulted in the high antitumor activity and low systemic toxicity of LNPs on tumor-bearing nude mice. The above results indicated that charge reversible mitochondria/nucleus dual targeted lipid hybrid nanoparticles greatly enhanced therapeutic efficacy of DOX for treating lung cancer.
Collapse
|
9
|
The Evolution of the Sigma-2 (σ 2) Receptor from Obscure Binding Site to Bona Fide Therapeutic Target. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:49-61. [PMID: 28315264 DOI: 10.1007/978-3-319-50174-1_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The sigma-2 (σ2) receptor represents one of the most poorly understood proteins in cell biology. Although this receptor was identified through in vitro binding studies over 25 years ago, the molecular identity of this protein is currently not unambiguously known, and the results from recent attempts to identify the σ2 receptor through protein purification and mass spectral analysis have been the subject of debate in the literature. However, there is overwhelming data demonstrating that the σ2 receptor is an important biomarker of tumor cell proliferation . The observation that σ2 receptor agonists are potent anticancer agents whereas σ2 antagonists block Aβ1-42 oligomer synaptic dysfunction in transgenic mouse models of Alzheimer's disease have clearly identified this protein as an important therapeutic target for the treatment of a variety of pathological conditions.
Collapse
|
10
|
Katnik C, Garcia A, Behensky AA, Yasny IE, Shuster AM, Seredenin SB, Petrov AV, Cuevas J. Activation of σ1 and σ2 receptors by afobazole increases glial cell survival and prevents glial cell activation and nitrosative stress after ischemic stroke. J Neurochem 2016; 139:497-509. [PMID: 27488244 DOI: 10.1111/jnc.13756] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 07/22/2016] [Accepted: 07/26/2016] [Indexed: 11/30/2022]
Abstract
Activation of sigma receptors at delayed time points has been shown to decrease injury following ischemic stroke. The mixed σ1/σ2 receptor agonist, 5-ethoxy-2-[2-(morpholino)-ethylthio]benzimidazole (afobazole), provides superior long-term outcomes compared to other σ ligands in the rat middle cerebral artery occlusion (MCAO) stroke model. Experiments using the MCAO model were carried out to determine the molecular mechanism involved in the beneficial effects of afobazole. Administration of afobazole (3 mg/kg) at delayed time points post-stroke significantly increased the number of microglia and astrocytes detected in the ipsilateral hemisphere at 96 h post-surgery. Morphological analysis of the microglia indicated that a greater number of these cells were found in the ramified resting state in MCAO animals treated with afobazole relative to MCAO vehicle controls. Similarly, fewer reactive astrocytes were detected in the injured hemisphere of afobazole-treated animals. Both the enhanced survival and reduced activation of glial cells were abolished by co-application of either a σ1 (BD-1063) or a σ2 (SM-21) receptor antagonist with afobazole. To gain further insight into the mechanisms by which afobazole lessens stroke injury, we probed the brain sections for markers of neuroinflammation (tumor necrosis factor α) and nitrosative stress (S-nitrosocysteine). Data show that afobazole significantly reduces S-nitrosocysteine levels, but does not alter tumor necrosis factor α expression 96 h after an ischemic stroke. Taken together our data indicate that afobazole acting via both σ1 and σ2 receptors decreases stroke injury by enhancing glial cell survival, blocking ischemia-induced glial cell activation, and decreasing nitrosative stress.
Collapse
Affiliation(s)
- Christopher Katnik
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Angela Garcia
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Adam A Behensky
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | | | | | | | | | - Javier Cuevas
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA.
| |
Collapse
|
11
|
Di Rosso ME, Palumbo ML, Genaro AM. Response to Letter to the Editor "On the targets of fluoxetine". Pharmacol Res 2016; 113:705-706. [PMID: 27297787 DOI: 10.1016/j.phrs.2016.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 06/03/2016] [Indexed: 11/28/2022]
Affiliation(s)
- María Emilia Di Rosso
- Instituto de Investigaciones Biomédicas (BIOMED), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad Católica Argentina (UCA), Av. Alicia Moreau de Justo 1600, Piso 3, 1107 Buenos Aires, Argentina
| | - María Laura Palumbo
- Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires (CIT NOBA)-CONICET-Universidad Nacional del Noroeste de la Provincia de Buenos Aires (UNNOBA), Monteagudo 2772, Pergamino, 2700 Buenos Aires, Argentina
| | - Ana María Genaro
- Instituto de Investigaciones Biomédicas (BIOMED), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad Católica Argentina (UCA), Av. Alicia Moreau de Justo 1600 , Piso 3, 1107 Buenos Aires, Argentina; Departamento de Farmacología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Paraguay 2155, Piso 15, 1121 Buenos Aires, Argentina.
| |
Collapse
|
12
|
Abstract
This review compares the biological and physiological function of Sigma receptors [σRs] and their potential therapeutic roles. Sigma receptors are widespread in the central nervous system and across multiple peripheral tissues. σRs consist of sigma receptor one (σ1R) and sigma receptor two (σ2R) and are expressed in numerous regions of the brain. The sigma receptor was originally proposed as a subtype of opioid receptors and was suggested to contribute to the delusions and psychoses induced by benzomorphans such as SKF-10047 and pentazocine. Later studies confirmed that σRs are non-opioid receptors (not an µ opioid receptor) and play a more diverse role in intracellular signaling, apoptosis and metabolic regulation. σ1Rs are intracellular receptors acting as chaperone proteins that modulate Ca2+ signaling through the IP3 receptor. They dynamically translocate inside cells, hence are transmembrane proteins. The σ1R receptor, at the mitochondrial-associated endoplasmic reticulum membrane, is responsible for mitochondrial metabolic regulation and promotes mitochondrial energy depletion and apoptosis. Studies have demonstrated that they play a role as a modulator of ion channels (K+ channels; N-methyl-d-aspartate receptors [NMDAR]; inositol 1,3,5 triphosphate receptors) and regulate lipid transport and metabolism, neuritogenesis, cellular differentiation and myelination in the brain. σ1R modulation of Ca2+ release, modulation of cardiac myocyte contractility and may have links to G-proteins. It has been proposed that σ1Rs are intracellular signal transduction amplifiers. This review of the literature examines the mechanism of action of the σRs, their interaction with neurotransmitters, pharmacology, location and adverse effects mediated through them.
Collapse
Affiliation(s)
- Colin G Rousseaux
- a Department of Pathology and Laboratory Medicine , University of Ottawa , Ottawa , ON , Canada and
| | | |
Collapse
|
13
|
Behensky AA, Yasny IE, Shuster AM, Seredenin SB, Petrov AV, Cuevas J. Stimulation of sigma receptors with afobazole blocks activation of microglia and reduces toxicity caused by amyloid-β25-35. J Pharmacol Exp Ther 2013; 347:458-67. [PMID: 24006337 DOI: 10.1124/jpet.113.208348] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease and the leading cause of senile dementia in the United States. Accumulation of amyloid-β (Aβ) and the effects of this peptide on microglial cells contribute greatly to the etiology of AD. Experiments were carried out to determine whether the pan-selective σ-receptor agonist afobazole can modulate microglial response to the cytotoxic Aβ fragment, Aβ25-35. Treatment with afobazole decreased microglial activation in response to Aβ, as indicated by reduced membrane ruffling and cell migration. The effects of afobazole on Aβ25-35-evoked migration were concentration dependent and consistent with σ-receptor activation. When afobazole was coapplied with either BD-1047 [N-[2-(3,4-dichlorophenyl)ethyl]-N-methyl-2-(dimethylamino)ethylamine dihydrobromide] or rimcazole, which are σ-1- and σ-2-selective antagonists, respectively, the inhibition of Aβ25-35-induced migration by afobazole was reduced. Prolonged exposure of microglia to Aβ25-35 resulted in glial cell death that was associated with increased expression of the proapoptotic protein Bax and the death protease caspase-3. Coapplication of afobazole with Aβ25-35 decreased the number of cells expressing both Bax and caspase-3 and resulted in a concomitant enhancement in cell survival. Although afobazole inhibited activation of microglia cells by Aβ25-35, it preserved normal functional responses in these cells after exposure to the amyloid peptide. Intracellular calcium increases induced by ATP were depressed in microglia after 24-hour exposure to Aβ25-35. However, coincubation in afobazole returned these responses to near control levels. Therefore, stimulation of σ-1 and σ-2 receptors by afobazole prevents Aβ25-35 activation of microglia and inhibits Aβ25-35-associated cytotoxicity, suggesting that afobazole may be useful for AD therapeutics.
Collapse
Affiliation(s)
- Adam A Behensky
- Department of Molecular Pharmacology and Physiology, University of South Florida, College of Medicine, Tampa, Florida (A.A.B., J.C.); IBC Generium, Volginsky, Russian Federation (I.E.Y., A.M.S., S.B.S.); and Zakusov Institute of Pharmacology, Russian Academy of Medical Sciences, Moscow, Russian Federation (S.B.S.)
| | | | | | | | | | | |
Collapse
|
14
|
Huang YS, Lu HL, Zhang LJ, Wu Z. Sigma-2 receptor ligands and their perspectives in cancer diagnosis and therapy. Med Res Rev 2013; 34:532-66. [PMID: 23922215 DOI: 10.1002/med.21297] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The sigma-2 receptor is highly expressed in various rapidly proliferating cancer cells and regarded as a cancer cell biomarker. Selective sigma-2 ligands have been shown to specifically label the tumor sites, induce cancer cells to undergo apoptosis, and inhibit tumor growth. Sigma-2 ligands are potentially useful as cancer diagnostics, anticancer therapeutics, or adjuvant anticancer treatment agents. However, both the cloning of this receptor and the identification of its endogenous ligand have not been successful, and the lack of structural information has severely hindered the understanding of its physiological roles, its signaling pathways, and the development of more selective sigma-2 ligands. Recent data have implicated that sigma-2 binding sites are within the lipid rafts and that PGRMC1 (progesterone receptor membrane component 1) complex and sigma-2 receptor may be coupled with EGFR (epidermal growth factor receptor), mTOR (mammalian target of rapamycin), caspases, and ion channels. Due to its promising applications in cancer management, there are rapidly increasing research efforts that are being directed into this field. This review article updates the current understanding of sigma-2 receptor and its potential physiological roles, applications, interaction with other effectors, with special focuses on the development of sigma-2 ligands, their chemical structures, pharmacological profiles, applications in imaging and anticancer therapy.
Collapse
Affiliation(s)
- Yun-Sheng Huang
- School of Pharmacy, Guangdong Medical College, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong, 523808, China
| | | | | | | |
Collapse
|