1
|
Wang T, Sun N, Ma Y, Zhang S. Recent Advances in the Development of Sigma Receptor (Radio)Ligands and Their Application in Tumors. ACS Pharmacol Transl Sci 2025; 8:951-977. [PMID: 40242588 PMCID: PMC11997895 DOI: 10.1021/acsptsci.4c00711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/19/2025] [Accepted: 02/26/2025] [Indexed: 04/18/2025]
Abstract
Cancer ranks among the top triumvirate leading causes of human deaths worldwide. The pathological mechanisms are notably intricate, demonstrating proliferative and metastatic capabilities, which complicate therapeutic interventions. The sigma-1 receptor (σ1R) plays a crucial role in tumor survival and migration, while the sigma-2 receptor (σ2R) is intimately associated with tumor proliferation. This review encapsulated the investigation concerning σ1R and σ2R in neoplasms and rigorously summarized the ligands and radio-ligands development and their tumor applications, such as antitumor cell proliferation and PET/SPECT imaging in tumors. A comprehensive classification discussion was undertaken regarding the chemical structures and emphasized the possibility of dual/multitargeted ligands. Ultimately, we discussed the effects of chiral structures and the pharmacological characteristics of ligands on affinity and pharmacokinetic features in vivo, particularly concerning radiopharmaceuticals. This review functions as a beneficial resource, fostering ligand deployment and stimulating the generation of innovative ideas for developing innovative radiopharmaceuticals.
Collapse
Affiliation(s)
- Tao Wang
- Department
of Nuclear Medicine, Xinqiao Hospital, Army
Medical University, Chongqing 400037, China
- School
of Medical Imaging, North Sichuan Medical
College, NanChong 637100, China
- Department
of Nuclear Medicine, Affiliated Hospital
of North Sichuan Medical College, North Sichuan Medical College, NanChong 637000, China
| | - Na Sun
- Department
of Nuclear Medicine, Xinqiao Hospital, Army
Medical University, Chongqing 400037, China
| | - Yanxi Ma
- Department
of Nuclear Medicine, Xinqiao Hospital, Army
Medical University, Chongqing 400037, China
| | - Song Zhang
- Department
of Nuclear Medicine, Xinqiao Hospital, Army
Medical University, Chongqing 400037, China
| |
Collapse
|
2
|
Yıldız B, Demirel R, Havadar HB, Yıldız G, Öziç C, Kamiloğlu NN, Özden Ö. Blocking SIG1R Along with Low Cadmium Exposure Display Anti-cancer Qualities in Both MCF7 and MDA-MB-231 Cells. Biol Trace Elem Res 2024; 202:3588-3600. [PMID: 37940833 DOI: 10.1007/s12011-023-03947-y] [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] [Received: 08/01/2023] [Accepted: 10/28/2023] [Indexed: 11/10/2023]
Abstract
Sigma-1 receptor (SIG1R) is a chaperone that modulates inositol 1,4,5-trisphosphate receptor type1 (IP3R1) calcium (Ca2+) channels on the endoplasmic reticulum. Therefore, SIG1R functions as an indirect regulator of Ca2+ and acts as an apoptosis modulator. Increased expression of SIG1R is associated with poor prognosis in breast cancers (BC), and SIG1R antagonists like BD1047 induce apoptosis. As a heavy metal, cadmium (Cd2+) is competitive with Ca2+ due to its physicochemical similarities and may trigger apoptosis at low concentrations. Our study investigated the SIG1R protein expression in 74 BC patients and found a significant increase in SIG1R expression in the triple-negative BC subtype. We also examined the apoptotic and anti-cancer effects of BD1047 in combination with CdCl2 in MCF7 and MDA-MB-213 cells. Cells were treated with CdCl2 at doses of 1 μM, 25 μM, and 50 μM, along with BD1047. Higher doses of CdCl2 were cytotoxic on both cancer cells and significantly increased DNA breaks. However, low-dose CdCl2 with BD1047 increased cell death and the apoptotic index in BC cells, although it did not exhibit cytotoxic effects on HUVEC cells. Co-administration of low-dose CdCl2 with BD1047 also reduced the migration and colony-forming ability of BC cells. Moreover, the expression of SIG1R protein in these groups decreased significantly compared to groups treated with BD1047 or low-dose CdCl2 alone. In conclusion, low-dose CdCl2 is thought to increase the apoptotic ability of BD1047 in BC cells by reducing SIG1R expression.
Collapse
Affiliation(s)
- Barış Yıldız
- Institute of Health Sciences, Department of Physiology, Kafkas University, 36100, Kars, Turkey
| | - Ramazan Demirel
- Department of Bioengineering, Institute of Natural and Applied Sciences, Kafkas University, 36100, Kars, Turkey
| | - Hatice Beşeren Havadar
- Deparment of Medical Pathology, Centre of Health Research and Training Hospital, Kafkas University, 36100, Kars, Turkey
| | - Gülden Yıldız
- Deparment of Medical Pathology, Centre of Health Research and Training Hospital, Kafkas University, 36100, Kars, Turkey
| | - Cem Öziç
- Department of Medical Biology, School of Medicine, Kafkas University, 36100, Kars, Turkey
| | - Nadide Nabil Kamiloğlu
- Department of Physiology, Faculty of Veterinary Medicine, Kafkas University, 36100, Kars, Turkey
| | - Özkan Özden
- Department of Bioengineering, Faculty of Engineering and Architecture, Kafkas University, 36100, Kars, Turkey.
| |
Collapse
|
3
|
Dashtestani P, Karami L. The molecular mechanism of the effects of the anti-neuropathic ligands on the modulation of the Sigma-2 receptor: An in-silico study. Int J Biol Macromol 2024; 254:127925. [PMID: 37944735 DOI: 10.1016/j.ijbiomac.2023.127925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 11/03/2023] [Accepted: 11/04/2023] [Indexed: 11/12/2023]
Abstract
Neuropathic pain (NP) is a prevalent medical condition that lacks an effective treatment. Recently, the Sigma-2 receptor (S2R) has been proposed as a potential therapeutic target for NP. Some highly-selective S2R ligands (UKH1114, CM398, and YTD) have shown promising results in vivo, but the molecular interaction between the S2R and these ligands has been scarcely investigated. This work explores changes in the S2R upon interaction with the three mentioned ligands using in silico approaches. The results indicated that the ICL1, H1, ICL2, and ECL are the most dynamic regions of S2R in all systems. Binding interaction analysis identified amino acids with significant contribution to the binding free energy. Notably, the UKH1114-S2R simulation trajectory revealed that small alterations in the ICL1, H1, ICL2, and ECL form a new stable opening in the S2R, linking the occluded S2R binding pocket to the endoplasmic reticulum lumen, providing more evidence for the assumptions about the EBP and S2R mechanism of function. Further, the agreement between the membrane parameters in our study and experimental values confirms the validity of the MD simulations. Overall, this study provides new insights into the interaction between anti-NP ligands and the S2R.
Collapse
Affiliation(s)
- Parisa Dashtestani
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Leila Karami
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| |
Collapse
|
4
|
Li J, Xian L, Zhu Z, Wang Y, Zhang W, Zheng R, Xue W, Li J. Role of CELF2 in ferroptosis: Potential targets for cancer therapy (Review). Int J Mol Med 2023; 52:88. [PMID: 37594127 PMCID: PMC10500222 DOI: 10.3892/ijmm.2023.5291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/19/2023] [Indexed: 08/19/2023] Open
Abstract
Ferroptosis is a novel form of regulated cellular necrosis that plays a critical role in promoting cancer progression and developing drug resistance. The main characteristic of ferroptosis is iron‑dependent lipid peroxidation caused by excess intracellular levels of reactive oxygen species. CUGBP ELAV‑like family number 2 (CELF2) is an RNA‑binding protein that is downregulated in various types of cancer and is associated with poor patient prognoses. CELF2 can directly bind mRNA to a variety of ferroptosis control factors; however, direct evidence of the regulatory role of CELF2 in ferroptosis is currently limited. The aim of the present review was to summarise the findings of previous studies on CELF2 and its role in regulating cellular redox homeostasis. The present review may provide insight into the possible mechanisms through which CELF2 affects ferroptosis and to provide recommendations for future studies.
Collapse
Affiliation(s)
- Jiahao Li
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Lei Xian
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zifeng Zhu
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yang Wang
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wenlei Zhang
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ruipeng Zheng
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wang Xue
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jiarui Li
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
5
|
Sorbi C, Belluti S, Atene CG, Marocchi F, Linciano P, Roy N, Paradiso E, Casarini L, Ronsisvalle S, Zanocco-Marani T, Brasili L, Lanfrancone L, Imbriano C, Di Rocco G, Franchini S. BS148 Reduces the Aggressiveness of Metastatic Melanoma via Sigma-2 Receptor Targeting. Int J Mol Sci 2023; 24:ijms24119684. [PMID: 37298633 DOI: 10.3390/ijms24119684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/18/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
The management of advanced-stage melanoma is clinically challenging, mainly because of its resistance to the currently available therapies. Therefore, it is important to develop alternative therapeutic strategies. The sigma-2 receptor (S2R) is overexpressed in proliferating tumor cells and represents a promising vulnerability to target. Indeed, we have recently identified a potent S2R modulator (BS148) that is effective in melanoma. To elucidate its mechanism of action, we designed and synthesized a BS148 fluorescent probe that enters SK-MEL-2 melanoma cells as assessed using confocal microscopy analysis. We show that S2R knockdown significantly reduces the anti-proliferative effect induced by BS148 administration, indicating the engagement of S2R in BS148-mediated cytotoxicity. Interestingly, BS148 treatment showed similar molecular effects to S2R RNA interference-mediated knockdown. We demonstrate that BS148 administration activates the endoplasmic reticulum stress response through the upregulation of protein kinase R-like ER kinase (PERK), activating transcription factor 4 (ATF4) genes, and C/EBP homologous protein (CHOP). Furthermore, we show that BS148 treatment downregulates genes related to the cholesterol pathway and activates the MAPK signaling pathway. Finally, we translate our results into patient-derived xenograft (PDX) cells, proving that BS148 treatment reduces melanoma cell viability and migration. These results demonstrate that BS148 is able to inhibit metastatic melanoma cell proliferation and migration through its interaction with the S2R and confirm its role as a promising target to treat cancer.
Collapse
Affiliation(s)
- Claudia Sorbi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Silvia Belluti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Claudio Giacinto Atene
- Hematology Section, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Federica Marocchi
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy
| | - Pasquale Linciano
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Neena Roy
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Ospedale di Baggiovara, 41126 Modena, Italy
| | - Elia Paradiso
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Ospedale di Baggiovara, 41126 Modena, Italy
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Ospedale di Baggiovara, 41126 Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Simone Ronsisvalle
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy
| | - Tommaso Zanocco-Marani
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Livio Brasili
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Luisa Lanfrancone
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy
| | - Carol Imbriano
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giulia Di Rocco
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Silvia Franchini
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
6
|
Dichiara M, Ambrosio FA, Barbaraci C, González-Cano R, Costa G, Parenti C, Marrazzo A, Pasquinucci L, Cobos EJ, Alcaro S, Amata E. Synthesis, Computational Insights, and Evaluation of Novel Sigma Receptors Ligands. ACS Chem Neurosci 2023; 14:1845-1858. [PMID: 37155827 DOI: 10.1021/acschemneuro.3c00074] [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: 05/10/2023] Open
Abstract
The development of diazabicyclo[4.3.0]nonane and 2,7-diazaspiro[3.5]nonane derivatives as sigma receptors (SRs) ligands is reported. The compounds were evaluated in S1R and S2R binding assays, and modeling studies were carried out to analyze the binding mode. The most notable compounds, 4b (AD186, KiS1R = 2.7 nM, KiS2R = 27 nM), 5b (AB21, KiS1R = 13 nM, KiS2R = 102 nM), and 8f (AB10, KiS1R = 10 nM, KiS2R = 165 nM), have been screened for analgesic effects in vivo, and their functional profile was determined through in vivo and in vitro models. Compounds 5b and 8f reached the maximum antiallodynic effect at 20 mg/kg. The selective S1R agonist PRE-084 completely reversed their action, indicating that the effects are entirely dependent on the S1R antagonism. Conversely, compound 4b sharing the 2,7-diazaspiro[3.5]nonane core as 5b was completely devoid of antiallodynic effect. Interestingly, compound 4b fully reversed the antiallodynic effect of BD-1063, indicating that 4b induces an S1R agonistic in vivo effect. The functional profiles were confirmed by the phenytoin assay. Our study might establish the importance of 2,7-diazaspiro[3.5]nonane core for the development of S1R compounds with specific agonist or antagonist profile and the role of the diazabicyclo[4.3.0]nonane in the development of novel SR ligands.
Collapse
Affiliation(s)
- Maria Dichiara
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Francesca Alessandra Ambrosio
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi "Magna Græcia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100 Catanzaro, Italy
| | - Carla Barbaraci
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Rafael González-Cano
- Departamento de Farmacología e Instituto de Neurociencias, Facultad de Medicina, Universitad de Granada e Instituto de Investigación Biosanitaria de Granada ibs.GRANADA, Avenida de la Investigación 11, 18016 Granada, Spain
| | - Giosuè Costa
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100 Catanzaro, Italy
- Net4Science Academic Spin-Off, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100 Catanzaro, Italy
| | - Carmela Parenti
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Agostino Marrazzo
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Lorella Pasquinucci
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Enrique J Cobos
- Departamento de Farmacología e Instituto de Neurociencias, Facultad de Medicina, Universitad de Granada e Instituto de Investigación Biosanitaria de Granada ibs.GRANADA, Avenida de la Investigación 11, 18016 Granada, Spain
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100 Catanzaro, Italy
- Net4Science Academic Spin-Off, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100 Catanzaro, Italy
| | - Emanuele Amata
- Dipartimento di Scienze del Farmaco e della Salute, Università degli Studi di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| |
Collapse
|
7
|
Stavroulaki D, Kyroglou I, Skourtis D, Athanasiou V, Thimi P, Sofianopoulou S, Kazaryan D, Fragouli PG, Labrianidou A, Dimas K, Patias G, Haddleton DM, Iatrou H. Influence of the Topology of Poly(L-Cysteine) on the Self-Assembly, Encapsulation and Release Profile of Doxorubicin on Dual-Responsive Hybrid Polypeptides. Pharmaceutics 2023; 15:790. [PMID: 36986652 PMCID: PMC10055909 DOI: 10.3390/pharmaceutics15030790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/16/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
Τhe synthesis of a series of novel hybrid block copolypeptides based on poly(ethylene oxide) (PEO), poly(l-histidine) (PHis) and poly(l-cysteine) (PCys) is presented. The synthesis of the terpolymers was achieved through a ring-opening polymerization (ROP) of the corresponding protected N-carboxy anhydrides of Nim-Trityl-l-histidine and S-tert-butyl-l-cysteine, using an end-amine-functionalized poly(ethylene oxide) (mPEO-NH2) as macroinitiator, followed by the deprotection of the polypeptidic blocks. The topology of PCys was either the middle block, the end block or was randomly distributed along the PHis chain. These amphiphilic hybrid copolypeptides assemble in aqueous media to form micellar structures, comprised of an outer hydrophilic corona of PEO chains, and a pH- and redox-responsive hydrophobic layer based on PHis and PCys. Due to the presence of the thiol groups of PCys, a crosslinking process was achieved further stabilizing the nanoparticles (NPs) formed. Dynamic light scattering (DLS), static light scattering (SLS) and transmission electron microscopy (TEM) were utilized to obtain the structure of the NPs. Moreover, the pH and redox responsiveness in the presence of the reductive tripeptide of glutathione (GSH) was investigated at the empty as well as the loaded NPs. The ability of the synthesized polymers to mimic natural proteins was examined by Circular Dichroism (CD), while the study of zeta potential revealed the "stealth" properties of NPs. The anticancer drug doxorubicin (DOX) was efficiently encapsulated in the hydrophobic core of the nanostructures and released under pH and redox conditions that simulate the healthy and cancer tissue environment. It was found that the topology of PCys significantly altered the structure as well as the release profile of the NPs. Finally, in vitro cytotoxicity assay of the DOX-loaded NPs against three different breast cancer cell lines showed that the nanocarriers exhibited similar or slightly better activity as compared to the free drug, rendering these novel NPs very promising materials for drug delivery applications.
Collapse
Affiliation(s)
- Dimitra Stavroulaki
- Industrial Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, GR-15771 Athens, Greece
| | - Iro Kyroglou
- Industrial Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, GR-15771 Athens, Greece
| | - Dimitrios Skourtis
- Industrial Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, GR-15771 Athens, Greece
| | - Varvara Athanasiou
- Industrial Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, GR-15771 Athens, Greece
| | - Pandora Thimi
- Industrial Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, GR-15771 Athens, Greece
| | - Sosanna Sofianopoulou
- Hellenic Police Headquarters, Forensic Science Division, Chemical and Physical Examinations Department, GR-10442 Athens, Greece
| | - Diana Kazaryan
- Industrial Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, GR-15771 Athens, Greece
| | - Panagiota G. Fragouli
- DIDPE, Dyeing, Finishing, Dyestuffs and Advanced Polymers Laboratory, University of West Attica, 250 Thevon Street, GR-12241 Athens, Greece
| | - Andromahi Labrianidou
- Laboratory of Pharmacology, Faculty of Medicine, University of Thessaly, Viopolis, GR-41500 Larissa, Greece
| | - Konstantinos Dimas
- Laboratory of Pharmacology, Faculty of Medicine, University of Thessaly, Viopolis, GR-41500 Larissa, Greece
| | - Georgios Patias
- Department of Chemistry, University of Warwick, Gibbet Hill, Coventry CV4 7AL, UK
| | - David M. Haddleton
- Department of Chemistry, University of Warwick, Gibbet Hill, Coventry CV4 7AL, UK
| | - Hermis Iatrou
- Industrial Chemistry Laboratory, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, GR-15771 Athens, Greece
| |
Collapse
|
8
|
Asong GM, Voshavar C, Amissah F, Bricker B, Lamango NS, Ablordeppey SY. An Evaluation of the Anticancer Properties of SYA014, a Homopiperazine-Oxime Analog of Haloperidol in Triple Negative Breast Cancer Cells. Cancers (Basel) 2022; 14:6047. [PMID: 36551533 PMCID: PMC9776707 DOI: 10.3390/cancers14246047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Triple negative breast cancer (TNBC) is a type of breast cancer associated with early metastasis, poor prognosis, high relapse rates, and mortality. Previously, we demonstrated that SYA013, a selective σ2RL, could inhibit cell proliferation, suppress migration, reduce invasion, and induce mitochondria-mediated apoptosis in MDA-MB-231 cell lines, although we were unable to demonstrate the direct involvement of sigma receptors. This study aimed to determine the anticancer properties and mechanisms of action of SYA014, [4-(4-(4-chlorophenyl)-1,4-diazepan-1-yl)-1-(4-fluorophenyl)butan-1-one oxime], an oxime analogue of SYA013, the contribution of its sigma-2 receptor (σ2R) binding, and its possible synergistic use with cisplatin to improve anticancer properties in two TNBC cell lines, MDA-MB-231 (Caucasian) and MDA-MB-468 (Black). In the present investigation, we have shown that SYA014 displays anticancer properties against cell proliferation, survival, metastasis and apoptosis in the two TNBC cell lines. Furthermore, a mechanistic investigation was conducted to identify the apoptotic pathway by which SYA014 induces cell death in MDA-MB-231 cells. Since SYA014 has a higher binding affinity for σ2R compared to σ1R, we tested the role of σ2R on the antiproliferative property of SYA014 with a σ2R blockade. We also attempted to evaluate the combination effect of SYA014 with cisplatin in TNBC cells.
Collapse
Affiliation(s)
- Gladys M. Asong
- College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA
| | - Chandrashekhar Voshavar
- College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA
| | - Felix Amissah
- College of Pharmacy, Ferris State University, Big Rapids, MI 49307, USA
| | - Barbara Bricker
- College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA
| | - Nazarius S. Lamango
- College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA
| | - Seth Y. Ablordeppey
- College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA
| |
Collapse
|
9
|
Mao D, Zhang X, Wang Z, Xu G, Zhang Y. TMEM97 is transcriptionally activated by YY1 and promotes colorectal cancer progression via the GSK-3β/β-catenin signaling pathway. Hum Cell 2022; 35:1535-1546. [PMID: 35907137 DOI: 10.1007/s13577-022-00759-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/21/2022] [Indexed: 11/25/2022]
Abstract
Transmembrane protein 97 (TMEM97) is a conserved integral membrane protein highly expressed in various human cancers, including colorectal cancer (CRC), and it exhibits pro-tumor roles in breast cancer, gastric cancer, and glioma. However, whether TMEM97 participates in CRC progression is not fully understood. The expression of mRNA and protein was evaluated by real-time qPCR, western blotting, immunofluorescent, and immunohistochemical staining. TMEM97 functions in cell proliferation, apoptosis, migration, and invasion were assessed by CCK-8, flow cytometry, and transwell assays. The roles of TMEM97 in CRC cells in vivo was investigated using a subcutaneous xenograft model. The transcriptional regulation of TMEM97 was explored by luciferase reporter and ChIP assays. The silencing of TMEM97 inhibited migration and invasion of CRC cells in vitro and led to suppressed growth and enhanced apoptosis in CRC cells and xenografts, whereas overexpression of TMEM97 displayed opposite effects. Mechanistically, TMEM97 knockdown caused a reduction of the proliferating marker PCNA and an increase of pro-apoptotic proteins (cleaved caspase 8/3/7 and cleaved PARP) in CRC cells. TMEM97 also positively regulated the β-catenin signaling pathway in CRC cells and xenografts by modulating the phosphorylated-GSK-3β and active (non-phospho) β-catenin levels. Interestingly, YY1, a well-recognized oncogenic transcription factor, was identified to bind to the TMEM97 promoter and enhance its transcriptional activity, and silencing of TMEM97 abolished YY1-mediated pro-tumor effects on CRC cells. Our results suggest that TMEM97 is transcriptionally activated by YY1 and promotes CRC progression via the GSK-3β/β-catenin signaling pathway, providing that TMEM97 might be a novel therapeutic target for preventing CRC development.
Collapse
Affiliation(s)
- Dong Mao
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, No. 2, The Fifth Section of Renmin Street, Jinzhou, Liaoning Province, China
| | - Xiaowei Zhang
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, No. 2, The Fifth Section of Renmin Street, Jinzhou, Liaoning Province, China
| | - Zhaoping Wang
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, No. 2, The Fifth Section of Renmin Street, Jinzhou, Liaoning Province, China
| | - Guannan Xu
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, No. 2, The Fifth Section of Renmin Street, Jinzhou, Liaoning Province, China
| | - Yun Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinzhou Medical University, No. 2, The Fifth Section of Renmin Street, Jinzhou, Liaoning Province, China.
| |
Collapse
|
10
|
Oflaz FE, Koshenov Z, Hirtl M, Rost R, Malli R, Graier WF. Sigma-1 Receptor Modulation by Ligands Coordinates Cancer Cell Energy Metabolism. Biomolecules 2022; 12:762. [PMID: 35740887 PMCID: PMC9221035 DOI: 10.3390/biom12060762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/16/2022] [Accepted: 05/27/2022] [Indexed: 02/06/2023] Open
Abstract
Sigma-1 receptor (S1R) is an important endoplasmic reticulum chaperone with various functions in health and disease. The purpose of the current work was to elucidate the involvement of S1R in cancer energy metabolism under its basal, activated, and inactivated states. For this, two cancer cell lines that differentially express S1R were treated with S1R agonist, (+)-SKF10047, and antagonist, BD1047. The effects of the agonist and antagonist on cancer energy metabolism were studied using single-cell fluorescence microscopy analysis of real-time ion and metabolite fluxes. Our experiments revealed that S1R activation by agonist increases mitochondrial bioenergetics of cancer cells while decreasing their reliance on aerobic glycolysis. S1R antagonist did not have a major impact on mitochondrial bioenergetics of tested cell lines but increased aerobic glycolysis of S1R expressing cancer cell line. Our findings suggest that S1R plays an important role in cancer energy metabolism and that S1R ligands can serve as tools to modulate it.
Collapse
Affiliation(s)
- Furkan E. Oflaz
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (R.M.)
| | - Zhanat Koshenov
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (R.M.)
| | - Martin Hirtl
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (R.M.)
| | - Rene Rost
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (R.M.)
| | - Roland Malli
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (R.M.)
- BioTechMed Graz, 8010 Graz, Austria
| | - Wolfgang F. Graier
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (R.M.)
- BioTechMed Graz, 8010 Graz, Austria
| |
Collapse
|
11
|
Abatematteo FS, Niso M, Lacivita E, Abate C. σ 2 Receptor and Its Role in Cancer with Focus on a MultiTarget Directed Ligand (MTDL) Approach. Molecules 2021; 26:3743. [PMID: 34205334 PMCID: PMC8235595 DOI: 10.3390/molecules26123743] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 11/19/2022] Open
Abstract
Sigma-2 (σ2) is an endoplasmic receptor identified as the Endoplasmic Reticulum (ER) transmembrane protein TMEM97. Despite its controversial identity, which was only recently solved, this protein has gained scientific interest because of its role in the proliferative status of cells; many tumor cells from different organs overexpress the σ2 receptor, and many σ2 ligands display cytotoxic actions in (resistant) cancer cells. These properties have shed light on the σ2 receptor as a potential druggable target to be bound/activated for the diagnosis or therapy of tumors. Additionally, diverse groups have shown how the σ2 receptor can be exploited for the targeted delivery of the anticancer drugs to tumors. As the cancer disease is a multifactorial pathology with multiple cell populations, a polypharmacological approach is very often needed. Instead of the simultaneous administration of different classes of drugs, the use of one molecule that interacts with diverse pharmacological targets, namely MultiTarget Directed Ligand (MTDL), is a promising and currently pursued strategy, that may overcome the pharmacokinetic problems associated with the administration of multiple molecules. This review aims to point out the progress regarding the σ2 ligands in the oncology field, with a focus on MTDLs directed towards σ2 receptors as promising weapons against (resistant) cancer diseases.
Collapse
Affiliation(s)
| | | | | | - Carmen Abate
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari ALDO MORO, Via Orabona 4, 70125 Bari, Italy; (F.S.A.); (M.N.); (E.L.)
| |
Collapse
|