1
|
Salis Torres A, Lee JE, Caporali A, Semple RK, Horrocks MH, MacRae VE. Mitochondrial Dysfunction as a Potential Mechanism Mediating Cardiac Comorbidities in Parkinson's Disease. Int J Mol Sci 2024; 25:10973. [PMID: 39456761 PMCID: PMC11507255 DOI: 10.3390/ijms252010973] [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: 09/11/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Individuals diagnosed with Parkinson's disease (PD) often exhibit heightened susceptibility to cardiac dysfunction, reflecting a complex interaction between these conditions. The involvement of mitochondrial dysfunction in the development and progression of cardiac dysfunction and PD suggests a plausible commonality in some aspects of their molecular pathogenesis, potentially contributing to the prevalence of cardiac issues in PD. Mitochondria, crucial organelles responsible for energy production and cellular regulation, play important roles in tissues with high energetic demands, such as neurons and cardiac cells. Mitochondrial dysfunction can occur in different and non-mutually exclusive ways; however, some mechanisms include alterations in mitochondrial dynamics, compromised bioenergetics, biogenesis deficits, oxidative stress, impaired mitophagy, and disrupted calcium balance. It is plausible that these factors contribute to the increased prevalence of cardiac dysfunction in PD, suggesting mitochondrial health as a potential target for therapeutic intervention. This review provides an overview of the physiological mechanisms underlying mitochondrial quality control systems. It summarises the diverse roles of mitochondria in brain and heart function, highlighting shared pathways potentially exhibiting dysfunction and driving cardiac comorbidities in PD. By highlighting strategies to mitigate dysfunction associated with mitochondrial impairment in cardiac and neural tissues, our review aims to provide new perspectives on therapeutic approaches.
Collapse
Affiliation(s)
- Agustina Salis Torres
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RH, UK; (A.S.T.); (J.-E.L.)
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, UK;
| | - Ji-Eun Lee
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RH, UK; (A.S.T.); (J.-E.L.)
- IRR Chemistry Hub, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Andrea Caporali
- Centre for Cardiovascular Science, Queen’s Medical Research Institute (QMRI), The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; (A.C.); (R.K.S.)
| | - Robert K. Semple
- Centre for Cardiovascular Science, Queen’s Medical Research Institute (QMRI), The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; (A.C.); (R.K.S.)
| | - Mathew H. Horrocks
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, UK;
- MRC Human Genetics Unit, Institute for Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Vicky E. MacRae
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RH, UK; (A.S.T.); (J.-E.L.)
| |
Collapse
|
2
|
Lu MJ, Zhang JQ, Nie ZY, Yan TH, Cao YB, Zhang LC, Li L. Monocyte/macrophage-mediated venous thrombus resolution. Front Immunol 2024; 15:1429523. [PMID: 39100675 PMCID: PMC11297357 DOI: 10.3389/fimmu.2024.1429523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/03/2024] [Indexed: 08/06/2024] Open
Abstract
Venous thromboembolism (VTE) poses a notable risk of morbidity and mortality. The natural resolution of the venous thrombus might be a potential alternative treatment strategy for VTE. Monocytes/macrophages merge as pivotal cell types in the gradual resolution of the thrombus. In this review, the vital role of macrophages in inducing inflammatory response, augmenting neovascularization, and facilitating the degradation of fibrin and collagen during thrombus resolution was described. The two phenotypes of macrophages involved in thrombus resolution and their dual functions were discussed. Macrophages expressing various factors, including cytokines and their receptors, adhesion molecules, chemokine receptors, vascular endothelial growth factor receptors, profibrinolytic- or antifibrinolytic-related enzymes, and other elements, are explored for their potential to promote or attenuate thrombus resolution. Furthermore, this review provides a comprehensive summary of new and promising therapeutic candidate drugs associated with monocytes/macrophages that have been demonstrated to promote or impair thrombus resolution. However, further clinical trials are essential to validate their efficacy in VTE therapy.
Collapse
Affiliation(s)
- Meng-Jiao Lu
- Institute of Vascular Disease, Shanghai TCM- Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Physiology and Pharmacology, China Pharmaceutic University, Nanjing, China
| | - Jia-Qi Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhou-Yu Nie
- Institute of Vascular Disease, Shanghai TCM- Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tian-Hua Yan
- Department of Physiology and Pharmacology, China Pharmaceutic University, Nanjing, China
| | - Yong-Bing Cao
- Institute of Vascular Disease, Shanghai TCM- Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li-Chao Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ling Li
- Institute of Vascular Disease, Shanghai TCM- Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
3
|
Hong GL, Kim KH, Cho SP, Lee HJ, Kim YJ, Jung JY. Korean red ginseng alleviates benign prostatic hyperplasia by dysregulating androgen receptor signaling and inhibiting DRP1-mediated mitochondrial fission. Chin J Nat Med 2024; 22:599-607. [PMID: 39059829 DOI: 10.1016/s1875-5364(24)60671-0] [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: 02/12/2024] [Indexed: 07/28/2024]
Abstract
Panax ginseng (C.A. Mey.) has been traditionally employed in Korea and China to alleviate fatigue and digestive disorders. In particular, Korean red ginseng (KRG), derived from streamed and dried P. ginseng, is known for its anti-aging and anti-inflammatory properties. However, its effects on benign prostatic hyperplasia (BPH), a representative aging-related disease, and the underlying mechanisms remain unclear. This study aims to elucidate the therapeutic effects of KRG on BPH, with a particular focus on mitochondrial dynamics, including fission and fusion processes. The effects of KRG on cell proliferation, apoptosis, and mitochondrial dynamics and morphology were evaluated in a rat model of testosterone propionate (TP)-induced BPH and TP-treated LNCaP cells, with mdivi-1 as a control. The results revealed that KRG treatment reduced the levels of androgen receptors (AR) and prostate-specific antigens in the BPH group. KRG inhibited cell proliferation by downregulating cyclin D and proliferating cell nuclear antigen (PCNA) levels, and it promoted apoptosis by increasing the ratio of B-cell lymphoma protein 2 (Bcl-2)-associated X protein (Bax) to Bcl-2 expression. Notably, KRG treatment enhanced the phosphorylation of dynamin-related protein 1 (DRP-1, serine 637) compared with that in the BPH group, which inhibited mitochondrial fission and led to mitochondrial elongation. This modulation of mitochondrial dynamics was associated with decreased cell proliferation and increased apoptosis. By dysregulating AR signaling and inhibiting mitochondrial fission through enhanced DRP-1 (ser637) phosphorylation, KRG effectively reduced cell proliferation and induced apoptosis. These findings suggest that KRG's regulation of mitochondrial dynamics offers a promising clinical approach for the treatment of BPH.
Collapse
Affiliation(s)
- Geum-Lan Hong
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon 34134, Korea; Department of Anatomy, College of Medicine, Konyang University, Daejeon 35365, Korea
| | - Kyung-Hyun Kim
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon 34134, Korea
| | - Sung-Pil Cho
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon 34134, Korea
| | - Hui-Ju Lee
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon 34134, Korea
| | - Yae-Ji Kim
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon 34134, Korea
| | - Ju-Young Jung
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon 34134, Korea.
| |
Collapse
|
4
|
He Y, Li R, Yu Y, Huang C, Xu Z, Wang T, Chen M, Huang H, Qi Z. Human neural stem cells promote mitochondrial genesis to alleviate neuronal damage in MPTP-induced cynomolgus monkey models. Neurochem Int 2024; 175:105700. [PMID: 38417589 DOI: 10.1016/j.neuint.2024.105700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/14/2024] [Accepted: 02/18/2024] [Indexed: 03/01/2024]
Abstract
Currently, there is no effective treatment for Parkinson's disease (PD), and the regenerative treatment of neural stem cells (NSCs) is considered the most promising method. This study aimed to investigate the protective effect and mechanism of NSCs on neurons in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induced cynomolgus monkey (Macaca fascicularis) model of PD. We first found that injecting NSCs into the subarachnoid space relieved motor dysfunction in PD cynomolgus monkeys, as well as reduced dopaminergic neuron loss and neuronal damage in the substantia nigra (SN) and striatum. Besides, NSCs decreased 17-estradiol (E2) level, an estrogen, in the cerebrospinal fluid (CSF) of PD cynomolgus monkeys, which shows NSCs may provide neuro-protection by controlling estrogen levels in the CSF. Furthermore, NSCs elevated proliferator-activated receptor gamma coactivator-1 alpha (PGC-1a), mitofusin 2 (MFN2), and optic atrophy 1 (OPA1) expression, three genes mediating mitochondrial biogenesis, in the SN and striatum of PD monkeys. In addition, NSCs suppress reactive oxygen species (ROS) production caused by MPTP, as well as mitochondrial autophagy, therefore preserving dopaminergic neurons. In summary, our findings show that NSCs may preserve dopaminergic and neuronal cells in an MPTP-induced PD cynomolgus monkey model. These protective benefits might be attributed to NSCs' ability of modulating estrogen balance, increasing mitochondrial biogenesis, and limiting oxidative stress and mitochondrial autophagy. These findings add to our understanding of the mechanism of NSC treatment and shed light on further clinical treatment options.
Collapse
Affiliation(s)
- Ying He
- Medical College, Guangxi University, Nanning, Guangxi, 530004, China; The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, 545007, China
| | - Ruicheng Li
- Medical College, Guangxi University, Nanning, Guangxi, 530004, China
| | - Yuxi Yu
- Medical College, Guangxi University, Nanning, Guangxi, 530004, China
| | - Chusheng Huang
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530004, China
| | - Zhiran Xu
- Translational Medicine Research Center, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, 530011, China
| | - Tianbao Wang
- Medical College, Guangxi University, Nanning, Guangxi, 530004, China
| | - Ming Chen
- Jinjiang Municipal Hospital (Shanghai Sixth People's Hospital Fujian Campus), Quanzhou, Fujian, 362200, China
| | - Hongri Huang
- Guangxi Taimei Rensheng Biotechnology Co., Ltd., Nanning, Guangxi, 530011, China
| | - Zhongquan Qi
- Medical College, Guangxi University, Nanning, Guangxi, 530004, China.
| |
Collapse
|
5
|
Bukato K, Kostrzewa T, Gammazza AM, Gorska-Ponikowska M, Sawicki S. Endogenous estrogen metabolites as oxidative stress mediators and endometrial cancer biomarkers. Cell Commun Signal 2024; 22:205. [PMID: 38566107 PMCID: PMC10985914 DOI: 10.1186/s12964-024-01583-0] [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: 11/29/2023] [Accepted: 03/23/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Endometrial cancer is the most common gynecologic malignancy found in developed countries. Because therapy can be curative at first, early detection and diagnosis are crucial for successful treatment. Early diagnosis allows patients to avoid radical therapies and offers conservative management options. There are currently no proven biomarkers that predict the risk of disease occurrence, enable early identification or support prognostic evaluation. Consequently, there is increasing interest in discovering sensitive and specific biomarkers for the detection of endometrial cancer using noninvasive approaches. CONTENT Hormonal imbalance caused by unopposed estrogen affects the expression of genes involved in cell proliferation and apoptosis, which can lead to uncontrolled cell growth and carcinogenesis. In addition, due to their ability to cause oxidative stress, estradiol metabolites have both carcinogenic and anticarcinogenic properties. Catechol estrogens are converted to reactive quinones, resulting in oxidative DNA damage that can initiate the carcinogenic process. The molecular anticancer mechanisms are still not fully understood, but it has been established that some estradiol metabolites generate reactive oxygen species and reactive nitrogen species, resulting in nitro-oxidative stress that causes cancer cell cycle arrest or cell death. Therefore, identifying biomarkers that reflect this hormonal imbalance and the presence of endometrial cancer in minimally invasive or noninvasive samples such as blood or urine could significantly improve early detection and treatment outcomes.
Collapse
Affiliation(s)
- Katarzyna Bukato
- Department of Obstetrics and Gynecology, Oncological Gynecology and Gynecological Endocrinology, Medical University of Gdansk, Smoluchowskiego 17, Gdańsk, 80-214, Poland
| | - Tomasz Kostrzewa
- Department of Medical Chemistry, Faculty of Medicine, Medical University of Gdansk, Dębinki 1, Gdansk, 80-211, Poland
| | - Antonella Marino Gammazza
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, 90127, Italy
| | - Magdalena Gorska-Ponikowska
- Department of Medical Chemistry, Faculty of Medicine, Medical University of Gdansk, Dębinki 1, Gdansk, 80-211, Poland.
- IEMEST Istituto Euro-Mediterraneo di Scienza e Tecnologia, Palermo, 90127, Italy.
- Department of Biophysics, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, 70174, Stuttgart, Germany.
| | - Sambor Sawicki
- Department of Obstetrics and Gynecology, Oncological Gynecology and Gynecological Endocrinology, Medical University of Gdansk, Smoluchowskiego 17, Gdańsk, 80-214, Poland.
| |
Collapse
|
6
|
Bastian P, Konieczna L, Dulski J, Daca A, Jacewicz D, Płoska A, Knap N, Sławek J, Bączek T, Kalinowski L, Drzeżdżon J, Roszmann A, Belka M, Górska-Ponikowska M. 2-Methoxyestradiol and Hydrogen Peroxide as Promising Biomarkers in Parkinson's Disease. Mol Neurobiol 2024; 61:148-166. [PMID: 37589832 PMCID: PMC10791893 DOI: 10.1007/s12035-023-03575-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/12/2023] [Indexed: 08/18/2023]
Abstract
Estrogens function in numerous physiological processes including controlling brain cell growth and differentiation. 2-Methoxestradiol (2-ME2), a 17β-estradiol (E2) metabolite, is known for its anticancer effects as observed both in vivo and in vitro. 2-ME2 affects all actively dividing cells, including neurons. The study aimed to determine whether 2-ME2 is a potentially cancer-protective or rather neurodegenerative agent in a specific tissue culture model as well as a clinical setup. In this study, 2-ME2 activity was determined in a Parkinson's disease (PD) in vitro model based on the neuroblastoma SH-SY5Y cell line. The obtained results suggest that 2-ME2 generates nitro-oxidative stress and controls heat shock proteins (HSP), resulting in DNA strand breakage and apoptosis. On the one hand, it may affect intensely dividing cells preventing cancer development; however, on the other hand, this kind of activity within the central nervous system may promote neurodegenerative diseases like PD. Thus, the translational value of 2-ME2's neurotoxic activity in a PD in vitro model was also investigated. LC-MS/MS technique was used to evaluate estrogens and their derivatives, namely, hydroxy and methoxyestrogens, in PD patients' blood, whereas the stopped-flow method was used to assess hydrogen peroxide (H2O2) levels. Methoxyestrogens and H2O2 levels were increased in patients' blood as compared to control subjects, but hydoxyestrogens were simultaneously decreased. From the above, we suggest that the determination of plasma levels of methoxyestrogens and H2O2 may be a novel PD biomarker. The presented research is the subject of the pending patent application "The use of hydrogen peroxide and 17β-estradiol and its metabolites as biomarkers in the diagnosis of neurodegenerative diseases," no. P.441360.
Collapse
Affiliation(s)
- Paulina Bastian
- Department of Medical Chemistry, Medical University of Gdansk, 80-210, Gdansk, Poland.
| | - Lucyna Konieczna
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416, Gdansk, Poland
| | - Jarosław Dulski
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
- Department of Neurological-Psychiatric Nursing, Medical University of Gdansk, 80-211, Gdansk, Poland
- Neurology & Stroke Dpt. St. Adalbert Hospital, "Copernicus" Ltd, 80-462, Gdansk, Poland
| | - Agnieszka Daca
- Department of Pathology and Experimental Rheumatology, Medical University of Gdansk, 80-210, Gdansk, Poland
| | - Dagmara Jacewicz
- Department of Environmental Technology, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Faculty of Pharmacy, Medical University of Gdansk, 80-211, Gdansk, Poland
| | - Narcyz Knap
- Department of Medical Chemistry, Medical University of Gdansk, 80-210, Gdansk, Poland
| | - Jarosław Sławek
- Department of Neurological-Psychiatric Nursing, Medical University of Gdansk, 80-211, Gdansk, Poland
- Neurology & Stroke Dpt. St. Adalbert Hospital, "Copernicus" Ltd, 80-462, Gdansk, Poland
| | - Tomasz Bączek
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416, Gdansk, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Faculty of Pharmacy, Medical University of Gdansk, 80-211, Gdansk, Poland
- BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, Narutowicza Street 11/12, 80-233, Gdansk, Poland
| | - Joanna Drzeżdżon
- Department of Environmental Technology, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland
| | - Anna Roszmann
- Department of Neurological-Psychiatric Nursing, Medical University of Gdansk, 80-211, Gdansk, Poland
- Neurology & Stroke Dpt. St. Adalbert Hospital, "Copernicus" Ltd, 80-462, Gdansk, Poland
| | - Mariusz Belka
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416, Gdansk, Poland
| | - Magdalena Górska-Ponikowska
- Department of Medical Chemistry, Medical University of Gdansk, 80-210, Gdansk, Poland.
- Department of Biophysics, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, 70569, Stuttgart, Germany.
- Euro-Mediterranean Institute of Science and Technology, 90139, Palermo, Italy.
| |
Collapse
|
7
|
Bastian PE, Daca A, Płoska A, Kuban-Jankowska A, Kalinowski L, Gorska-Ponikowska M. 2-Methoxyestradiol Damages DNA in Glioblastoma Cells by Regulating nNOS and Heat Shock Proteins. Antioxidants (Basel) 2022; 11:2013. [PMID: 36290736 PMCID: PMC9598669 DOI: 10.3390/antiox11102013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/02/2022] [Accepted: 10/10/2022] [Indexed: 08/18/2023] Open
Abstract
Gliomas are the most prevalent primary tumors of the central nervous system (CNS), accounting for over fifty percent of all primary intracranial neoplasms. Glioblastoma (GBM) is the most prevalent form of malignant glioma and is often incurable. The main distinguishing trait of GBM is the presence of hypoxic regions accompanied by enhanced angiogenesis. 2-Methoxyestradiol (2-ME) is a well-established antiangiogenic and antiproliferative drug. In current clinical studies, 2-ME, known as Panzem, was examined for breast, ovarian, prostate, and multiple myeloma. The SW1088 grade III glioma cell line was treated with pharmacological and physiological doses of 2-ME. The induction of apoptosis and necrosis, oxidative stress, cell cycle arrest, and mitochondrial membrane potential were established by flow cytometry. Confocal microscopy was used to detect DNA damage. The Western blot technique determined the level of nitric oxide synthase and heat shock proteins. Here, for the first time, 2-ME is shown to induce nitro-oxidative stress with the concomitant modulation of heat shock proteins (HSPs) in the SW1088 grade III glioma cell line. Crucial therapeutic strategies for GMB should address both cell proliferation and angiogenesis, and due to the above, 2-ME seems to be a perfect candidate for GBM therapy.
Collapse
Affiliation(s)
| | - Agnieszka Daca
- Department of Pathology and Experimental Rheumatology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Faculty of Pharmacy, Medical University of Gdansk, 80-211 Gdansk, Poland
| | | | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Faculty of Pharmacy, Medical University of Gdansk, 80-211 Gdansk, Poland
- BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, Narutowicza Street 11/12, 80-233 Gdansk, Poland
| | - Magdalena Gorska-Ponikowska
- Department of Medical Chemistry, Medical University of Gdansk, 80-210 Gdansk, Poland
- Department of Biophysics, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, D-70569 Stuttgart, Germany
- Euro-Mediterranean Institute of Science and Technology, 90139 Palermo, Italy
| |
Collapse
|
8
|
Musial C, Knap N, Zaucha R, Bastian P, Barone G, Lo Bosco G, Lo-Celso F, Konieczna L, Belka M, Bączek T, Gammazza AM, Kuban-Jankowska A, Cappello F, Nussberger S, Gorska-Ponikowska M. Induction of 2-hydroxycatecholestrogens O-methylation: A missing puzzle piece in diagnostics and treatment of lung cancer. Redox Biol 2022; 55:102395. [PMID: 35841627 PMCID: PMC9289866 DOI: 10.1016/j.redox.2022.102395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/15/2022] [Accepted: 07/02/2022] [Indexed: 02/07/2023] Open
Abstract
Lung cancer is one of the most common cancers worldwide, causing nearly one million deaths each year. Herein, we present the effect of 2-methoxyestradiol (2-ME), the endogenous metabolite of 17β-estradiol (E2), on non-small cell lung cancer (NSCLC) cells. We observed that 2-ME reduced the viability of lung adenocarcinoma in two-dimensional (2D) and three-dimensional (3D) spheroidal A549 cell culture models. Molecular modeling was carried out aiming to visualize amino acid residues within binding pockets of the acyl-protein thioesterases, namely 1 (APT1) and 2 (APT2), and thus to identify which ones were more likely involved in the interaction with 2-ME. Our findings suggest that 2-ME acts as an APT1 inhibitor enhancing protein palmitoylation and oxidative stress phenomena in the lung cancer cell. In order to support our data, metabolomics of blood serum from NSCLC patients was also performed. Moreover, computational analysis suggests that 2-ME as compared to other estrogen metabolism intermediates is relatively safe in terms of its possible non-receptor bioactivity within healthy human cells due to a very low electrophilic potential and hence no substantial risk of spontaneous covalent modification of biologically protective nucleophiles. We propose that 2-ME can be used as a selective tumor biomarker in the course of certain types of lung cancers and possibly as a therapeutic adjuvant or neoadjuvant.
Collapse
Affiliation(s)
- Claudia Musial
- Department of Medical Chemistry, Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland
| | - Narcyz Knap
- Department of Medical Chemistry, Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland
| | - Renata Zaucha
- Department of Clinical Oncology and Radiotherapy, Medical University of Gdansk, 80-214, Gdansk, Poland
| | - Paulina Bastian
- Department of Medical Chemistry, Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland
| | - Giampaolo Barone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90128, Palermo, Italy
| | - Giosuè Lo Bosco
- Department of Mathematics and Computer Science, University of Palermo, 90133, Palermo, Italy; Euro-Mediterranean Institute of Science and Technology, 90139, Palermo, Italy
| | - Fabrizio Lo-Celso
- Department of Physics and Chemistry 'Emilio Segrè', University of Palermo, 90128, Palermo, Italy
| | - Lucyna Konieczna
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416, Gdansk, Poland
| | - Mariusz Belka
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416, Gdansk, Poland
| | - Tomasz Bączek
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416, Gdansk, Poland
| | - Antonella Marino Gammazza
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, 90127, Palermo, Italy
| | - Alicja Kuban-Jankowska
- Department of Medical Chemistry, Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland
| | - Francesco Cappello
- Euro-Mediterranean Institute of Science and Technology, 90139, Palermo, Italy; Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, 90127, Palermo, Italy
| | - Stephan Nussberger
- Department of Biophysics, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, 70569, Stuttgart, Germany
| | - Magdalena Gorska-Ponikowska
- Department of Medical Chemistry, Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland; Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90128, Palermo, Italy; Euro-Mediterranean Institute of Science and Technology, 90139, Palermo, Italy; Department of Biophysics, Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, 70569, Stuttgart, Germany.
| |
Collapse
|