1
|
Marranci A, Maresca L, Lodovichi S, Luserna di Rorà AG, Stecca B, Poliseno L. PARP1 in melanoma: Mechanistic insights and implications for basic and clinical research. Cancer Lett 2025; 617:217599. [PMID: 40024566 DOI: 10.1016/j.canlet.2025.217599] [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: 11/25/2024] [Revised: 02/04/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Targeted therapies and immunotherapies have revolutionized the treatment of metastatic melanoma and have set a successful example for the treatment of other cancers. A similar breakthrough was achieved with the advent of PARP inhibitors (PARPi) in breast and ovarian cancer. Recent evidence highlights the critical role of PARP1 in melanoma initiation and progression. High PARP1 expression correlates with aggressive melanoma characteristics and poor patient outcomes. Preclinical and clinical data suggest that PARPi, alone or in combination, can effectively reduce melanoma cell viability and inhibit tumor growth. However, integrating PARPi with current treatment approaches and identifying patients who could benefit the most from such combinations remain underexplored areas of investigation. This review highlights the need for further basic and clinical research on PARP1 in melanoma, to better understand its role and to tackle major challenges in the field, such as resistance to targeted therapies and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Andrea Marranci
- Oncohematology Unit, Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, 56017, Pisa, Italy. http://www.fpscience.it/
| | - Luisa Maresca
- Tumor Cell Biology Unit, Core Research Laboratory (CRL), Institute for Cancer Research and Prevention (ISPRO), 50139, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, 50139, Florence, Italy
| | - Samuele Lodovichi
- Department of Biosciences, University of Milan, 20133, Milan, Italy; Institute of Clinical Physiology, National Research Council (CNR-IFC), 56124, Pisa, Italy
| | | | - Barbara Stecca
- Tumor Cell Biology Unit, Core Research Laboratory (CRL), Institute for Cancer Research and Prevention (ISPRO), 50139, Florence, Italy
| | - Laura Poliseno
- Institute of Clinical Physiology, National Research Council (CNR-IFC), 56124, Pisa, Italy; Oncogenomics Unit, Core Research Laboratory, Institute for Cancer Research and Prevention (ISPRO), 56124, Pisa, Italy.
| |
Collapse
|
2
|
顾 庆, 李 剑, 陈 裕, 汪 林, 李 义, 王 子, 王 一, 杨 民. [Mechanism of sodium valproate in inhibiting ferroptosis of bone marrow mesenchymal stem cells via the adenosine monophosphate-activated protein kinase/Sirtuin 1 axis]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2025; 39:215-223. [PMID: 39971368 PMCID: PMC11839301 DOI: 10.7507/1002-1892.202411089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/22/2025] [Indexed: 02/21/2025]
Abstract
Objective To investigate the effects of sodium valproate (VPA) in inhibiting Erastin-induced ferroptosis in bone marrow mesenchymal stem cells (BMSCs) and its underlying mechanisms. Methods BMSCs were isolated from bone marrow of 8-week-old Spragur Dawley rats and identified [cell surface antigens CD90, CD44, and CD45 were analyzed by flow cytometry, and osteogenic and adipogenic differentiation abilities were assessed by alizarin red S (ARS) and oil red O staining, respectively]. Cells of passage 3 were used for the Erastin-induced ferroptosis model, with different concentrations of VPA for intervention. The optimal drug concentration was determined using the cell counting kit 8 assay. The experiment was divided into 4 groups: group A, cells were cultured in osteogenic induction medium for 24 hours; group B, cells were cultured in osteogenic induction medium containing optimal concentration Erastin for 24 hours; group C, cells were cultured in osteogenic induction medium containing optimal concentration Erastin and VPA for 24 hours; group D, cells were cultured in osteogenic induction medium containing optimal concentration Erastin and VPA, and 8 μmol/L EX527 for 24 hours. The mitochondrial state of the cells was evaluated, including the levels of malondialdehyde (MDA), glutathione (GSH), and reactive oxygen species (ROS). Osteogenic capacity was assessed by alkaline phosphatase (ALP) activity and ARS staining. Western blot analysis was performed to detect the expressions of osteogenic-related proteins [Runt-related transcription factor 2 (RUNX2) and osteopontin (OPN)], ferroptosis-related proteins [glutathione peroxidase 4 (GPX4), ferritin heavy chain 1 (FTH1), and solute carrier family 7 member 11 (SLC7A11)], and pathway-related proteins [adenosine monophosphate-activated protein kinase (AMPK) and Sirtuin 1 (SIRT1)]. Results The cultured cells were identified as BMSCs. VPA inhibited Erastin-induced ferroptosis and the decline of osteogenic ability in BMSCs, acting through the activation of the AMPK/SIRT1 pathway. VPA significantly reduced the levels of ROS and MDA in Erastin-treated BMSCs and significantly increased GSH levels. Additionally, the expression levels of ferroptosis-related proteins (GPX4, FTH1, and SLC7A11) significantly decreased. VPA also upregulated the expressions of osteogenic-related proteins (RUNX2 and OPN), enhanced mineralization and osteogenic differentiation, and increased the expressions of pathway-related proteins (AMPK and SIRT1). These effects could be reversed by the SIRT1 inhibitor EX527. Conclusion VPA inhibits ferroptosis in BMSCs through the AMPK/SIRT1 axis and promotes osteogenesis.
Collapse
Affiliation(s)
- 庆松 顾
- 皖南医学院第一附属医院/弋矶山医院创伤骨科(安徽芜湖 241001)Department of Traumatic Orthopedics, the First Affiliated Hospital of Wannan Medical College/Yijishan Hospital, Wuhu Anhui, 241001, P. R. China
| | - 剑桥 李
- 皖南医学院第一附属医院/弋矶山医院创伤骨科(安徽芜湖 241001)Department of Traumatic Orthopedics, the First Affiliated Hospital of Wannan Medical College/Yijishan Hospital, Wuhu Anhui, 241001, P. R. China
| | - 裕虎 陈
- 皖南医学院第一附属医院/弋矶山医院创伤骨科(安徽芜湖 241001)Department of Traumatic Orthopedics, the First Affiliated Hospital of Wannan Medical College/Yijishan Hospital, Wuhu Anhui, 241001, P. R. China
| | - 林辉 汪
- 皖南医学院第一附属医院/弋矶山医院创伤骨科(安徽芜湖 241001)Department of Traumatic Orthopedics, the First Affiliated Hospital of Wannan Medical College/Yijishan Hospital, Wuhu Anhui, 241001, P. R. China
| | - 义恒 李
- 皖南医学院第一附属医院/弋矶山医院创伤骨科(安徽芜湖 241001)Department of Traumatic Orthopedics, the First Affiliated Hospital of Wannan Medical College/Yijishan Hospital, Wuhu Anhui, 241001, P. R. China
| | - 子儒 王
- 皖南医学院第一附属医院/弋矶山医院创伤骨科(安徽芜湖 241001)Department of Traumatic Orthopedics, the First Affiliated Hospital of Wannan Medical College/Yijishan Hospital, Wuhu Anhui, 241001, P. R. China
| | - 一聪 王
- 皖南医学院第一附属医院/弋矶山医院创伤骨科(安徽芜湖 241001)Department of Traumatic Orthopedics, the First Affiliated Hospital of Wannan Medical College/Yijishan Hospital, Wuhu Anhui, 241001, P. R. China
| | - 民 杨
- 皖南医学院第一附属医院/弋矶山医院创伤骨科(安徽芜湖 241001)Department of Traumatic Orthopedics, the First Affiliated Hospital of Wannan Medical College/Yijishan Hospital, Wuhu Anhui, 241001, P. R. China
| |
Collapse
|
3
|
Liu J, Yu Y, Xu B, Liang Q, Fang T, Zhou N, Sun G. NOTCH1 regulates the DNA damage response and sorafenib resistance by activating ATM in hepatocellular carcinoma. Am J Transl Res 2024; 16:7317-7329. [PMID: 39822534 PMCID: PMC11733373 DOI: 10.62347/eafu3015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 10/30/2024] [Indexed: 01/19/2025]
Abstract
OBJECTIVE This study investigates the mechanism underlying sorafenib resistance in hepatocellular carcinoma cells (HCC), focusing on DNA damage repair (DDR) pathways to develop targeted therapeutic strategies. METHODS Bioinformatics analysis was used to screen genes associated with sorafenib resistance, which was further demonstrated by western blotting. Cell proliferation was determined using the EdU assay. The presence of binding sites between valproic acid (VPA) and NOTCH1 was analyzed by molecular docking. Comet and flow cytometry assays evaluated DNA damage and cell cycle arrest induced by VPA in sorafenib-resistant cells, with further mechanistic insights gained via western blotting and co-immunoprecipitation (Co-IP). RESULTS We found that NOTCH1/ATM axis plays a vital role in the prognosis of patients with liver cancer and in the behavior of sorafenib-resistant cells. HCC resistant to sorafenib exhibited enhanced cell proliferation ability. Moreover, overexpression of NOTCH1 in sorafenib-sensitive HCC cells significantly increased liver cancer cell proliferation. Conversely, silencing NOTCH1 expression in sorafenib-resistant HCC cell lines reduced their proliferative activity. Additionally, VPA enhanced the therapeutic efficacy against sorafenib-resistance cells by modulating NOTCH1/ATM/p-BRCA1/p-CHK2/γ-H2AX signaling axis and homologous recombination (HR) activity. CONCLUSION Targeting NOTCH1 and ATM is a promising strategy to overcome sorafenib resistance in HCC, particularly through the combined use of VPA and sorafenib.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pharmacy, Shanghai Fifth People’s Hospital, Fudan University801 Heqing Road, Shanghai 200240, China
| | - Yan Yu
- Department of Pharmacy, Shanghai Fifth People’s Hospital, Fudan University801 Heqing Road, Shanghai 200240, China
| | - Bin Xu
- Department of Ultrasonic Medicine, Shanghai Fifth People’s Hospital, Fudan University801 Heqing Road, Shanghai 200240, China
| | - Qing Liang
- Department of Pharmacy, Shanghai Fifth People’s Hospital, Fudan University801 Heqing Road, Shanghai 200240, China
| | - Tingting Fang
- Department of Pharmacy, The Shanghai University of Medicine and Health Sciences279 Zhouzhu Highway, Pudong New Area, Shanghai 201318, China
| | - Ningming Zhou
- Department of Ultrasonic Medicine, Shanghai Fifth People’s Hospital, Fudan University801 Heqing Road, Shanghai 200240, China
| | - Guangchun Sun
- Department of Pharmacy, Shanghai Fifth People’s Hospital, Fudan University801 Heqing Road, Shanghai 200240, China
| |
Collapse
|
4
|
Barszczewska-Pietraszek G, Czarny P, Drzewiecka M, Błaszczyk M, Radek M, Synowiec E, Wigner-Jeziorska P, Sitarek P, Szemraj J, Skorski T, Śliwiński T. Polθ Inhibitor (ART558) Demonstrates a Synthetic Lethal Effect with PARP and RAD52 Inhibitors in Glioblastoma Cells. Int J Mol Sci 2024; 25:9134. [PMID: 39273083 PMCID: PMC11395082 DOI: 10.3390/ijms25179134] [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: 06/18/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/15/2024] Open
Abstract
DNA repair proteins became the popular targets in research on cancer treatment. In our studies we hypothesized that inhibition of DNA polymerase theta (Polθ) and its combination with Poly (ADP-ribose) polymerase 1 (PARP1) or RAD52 inhibition and the alkylating drug temozolomide (TMZ) has an anticancer effect on glioblastoma cells (GBM21), whereas it has a low impact on normal human astrocytes (NHA). The effect of the compounds was assessed by analysis of cell viability, apoptosis, proliferation, DNA damage and cell cycle distribution, as well as gene expression. The main results show that Polθ inhibition causes a significant decrease in glioblastoma cell viability. It induces apoptosis, which is accompanied by a reduction in cell proliferation and DNA damage. Moreover, the effect was stronger when dual inhibition of Polθ with PARP1 or RAD52 was applied, and it is further enhanced by addition of TMZ. The impact on normal cells is much lower, especially when considering cell viability and DNA damage. In conclusion, we would like to highlight that Polθ inhibition used in combination with PARP1 or RAD52 inhibition has great potential to kill glioblastoma cells, and shows a synthetic lethal effect, while sparing normal astrocytes.
Collapse
Affiliation(s)
- Gabriela Barszczewska-Pietraszek
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (G.B.-P.)
| | - Piotr Czarny
- Department of Medical Biochemistry, Medical University of Lodz, 92-216 Lodz, Poland
| | - Małgorzata Drzewiecka
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (G.B.-P.)
| | - Maciej Błaszczyk
- Department of Neurosurgery, Surgery of Spine and Peripheral Nerves, Medical University of Lodz, University Hospital WAM-CSW, 90-549 Lodz, Poland
| | - Maciej Radek
- Department of Neurosurgery, Surgery of Spine and Peripheral Nerves, Medical University of Lodz, University Hospital WAM-CSW, 90-549 Lodz, Poland
| | - Ewelina Synowiec
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (G.B.-P.)
| | - Paulina Wigner-Jeziorska
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (G.B.-P.)
| | - Przemysław Sitarek
- Department of Medical Biology, Medical University of Lodz, 92-151 Lodz, Poland
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, 92-216 Lodz, Poland
| | - Tomasz Skorski
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Tomasz Śliwiński
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (G.B.-P.)
| |
Collapse
|
5
|
Valdez-Salazar F, Jiménez-Del Rio LA, Padilla-Gutiérrez JR, Valle Y, Muñoz-Valle JF, Valdés-Alvarado E. Advances in Melanoma: From Genetic Insights to Therapeutic Innovations. Biomedicines 2024; 12:1851. [PMID: 39200315 PMCID: PMC11351162 DOI: 10.3390/biomedicines12081851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Advances in melanoma research have unveiled critical insights into its genetic and molecular landscape, leading to significant therapeutic innovations. This review explores the intricate interplay between genetic alterations, such as mutations in BRAF, NRAS, and KIT, and melanoma pathogenesis. The MAPK and PI3K/Akt/mTOR signaling pathways are highlighted for their roles in tumor growth and resistance mechanisms. Additionally, this review delves into the impact of epigenetic modifications, including DNA methylation and histone changes, on melanoma progression. The tumor microenvironment, characterized by immune cells, stromal cells, and soluble factors, plays a pivotal role in modulating tumor behavior and treatment responses. Emerging technologies like single-cell sequencing, CRISPR-Cas9, and AI-driven diagnostics are transforming melanoma research, offering precise and personalized approaches to treatment. Immunotherapy, particularly immune checkpoint inhibitors and personalized mRNA vaccines, has revolutionized melanoma therapy by enhancing the body's immune response. Despite these advances, resistance mechanisms remain a challenge, underscoring the need for combined therapies and ongoing research to achieve durable therapeutic responses. This comprehensive overview aims to highlight the current state of melanoma research and the transformative impacts of these advancements on clinical practice.
Collapse
Affiliation(s)
| | | | | | | | | | - Emmanuel Valdés-Alvarado
- Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Ciencias Biomédicas (IICB), Universidad de Guadalajara, Guadalajara 44340, Mexico; (F.V.-S.)
| |
Collapse
|
6
|
Chong ZX, Ho WY, Yeap SK. Decoding the tumour-modulatory roles of LIMK2. Life Sci 2024; 347:122609. [PMID: 38580197 DOI: 10.1016/j.lfs.2024.122609] [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: 12/13/2023] [Revised: 03/19/2024] [Accepted: 04/01/2024] [Indexed: 04/07/2024]
Abstract
LIM domains kinase 2 (LIMK2) is a 72 kDa protein that regulates actin and cytoskeleton reorganization. Once phosphorylated by its upstream activator (ROCK1), LIMK2 can phosphorylate cofilin to inactivate it. This relieves the levering stress on actin and allows polymerization to occur. Actin rearrangement is essential in regulating cell cycle progression, apoptosis, and migration. Dysregulation of the ROCK1/LIMK2/cofilin pathway has been reported to link to the development of various solid cancers such as breast, lung, and prostate cancer and liquid cancer like leukemia. This review aims to assess the findings from multiple reported in vitro, in vivo, and clinical studies on the potential tumour-regulatory role of LIMK2 in different human cancers. The findings of the selected literature unraveled that activated AKT, EGF, and TGF-β pathways can upregulate the activities of the ROCK1/LIMK2/cofilin pathway. Besides cofilin, LIMK2 can modulate the cellular levels of other proteins, such as TPPP1, to promote microtubule polymerization. The tumour suppressor protein p53 can transactivate LIMK2b, a splice variant of LIMK2, to induce cell cycle arrest and allow DNA repair to occur before the cell enters the next phase of the cell cycle. Additionally, several non-coding RNAs, such as miR-135a and miR-939-5p, could also epigenetically regulate the expression of LIMK2. Since the expression of LIMK2 is dysregulated in several human cancers, measuring the tissue expression of LIMK2 could potentially help diagnose cancer and predict patient prognosis. As LIMK2 could play tumour-promoting and tumour-inhibiting roles in cancer development, more investigation should be conducted to carefully evaluate whether introducing a LIMK2 inhibitor in cancer patients could slow cancer progression without posing clinical harms.
Collapse
Affiliation(s)
- Zhi Xiong Chong
- Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia.
| | - Wan Yong Ho
- Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia.
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, 43900 Sepang, Selangor, Malaysia.
| |
Collapse
|
7
|
Man E, Evran S. Deacetylation of Histones and Non-histone Proteins in Inflammatory Diseases and Cancer Therapeutic Potential of Histone Deacetylase Inhibitors. Curr Genomics 2023; 24:136-145. [PMID: 38178983 PMCID: PMC10761333 DOI: 10.2174/0113892029265046231011100327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/18/2023] [Accepted: 08/26/2023] [Indexed: 01/06/2024] Open
Abstract
Epigenetic changes play an important role in the pathophysiology of autoimmune diseases such as allergic asthma, multiple sclerosis, lung diseases, diabetes, cystic fibrosis, atherosclerosis, rheumatoid arthritis, and COVID-19. There are three main classes of epigenetic alterations: post-translational modifications of histone proteins, control by non-coding RNA and DNA methylation. Since histone modifications can directly affect chromatin structure and accessibility, they can regulate gene expression levels. Abnormal expression and activity of histone deacetylases (HDACs) have been reported in immune mediated diseases. Increased acetylated levels of lysine residues have been suggested to be related to the overexpression of inflammatory genes. This review focuses on the effect of HDAC modifications on histone and non-histone proteins in autoimmune diseases. Furthermore, we discuss the potential therapeutic effect of HDAC inhibitors (HDACi) used in these diseases.
Collapse
Affiliation(s)
- Ezgi Man
- Department of Biochemistry, Faculty of Science, Ege University, 35100, İzmir, Türkiye
- EGE SCIENCE PRO Scientific Research Inc., Ege University, IdeEGE Technology Development Zone, 35100, Bornova-Izmir, Türkiye
| | - Serap Evran
- Department of Biochemistry, Faculty of Science, Ege University, 35100, İzmir, Türkiye
| |
Collapse
|
8
|
Drzewiecka M, Jaśniak D, Barszczewska-Pietraszek G, Czarny P, Kobrzycka A, Wieczorek M, Radek M, Szemraj J, Skorski T, Śliwiński T. Class I HDAC Inhibition Leads to a Downregulation of FANCD2 and RAD51, and the Eradication of Glioblastoma Cells. J Pers Med 2023; 13:1315. [PMID: 37763083 PMCID: PMC10532614 DOI: 10.3390/jpm13091315] [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: 06/23/2023] [Revised: 07/26/2023] [Accepted: 08/16/2023] [Indexed: 09/29/2023] Open
Abstract
HDAC inhibitors (HDACi) hold great potential as anticancer therapies due to their ability to regulate the acetylation of both histone and non-histone proteins, which is frequently disrupted in cancer and contributes to the development and advancement of the disease. Additionally, HDACi have been shown to enhance the cytotoxic effects of DNA-damaging agents such as radiation and cisplatin. In this study, we found that histone deacetylase inhibits valproic acid (VPA), synergized with PARP1 inhibitor (PARPi), talazoparib (BMN-673), and alkylating agent, and temozolomide (TMZ) to induce DNA damage and reduce glioblastoma multiforme. At the molecular level, VPA leads to a downregulation of FANCD2 and RAD51, and the eradication of glioblastoma cells. The results of this study indicate that combining HDACi with PARPi could potentially enhance the treatment of glioblastoma, the most aggressive type of cancer that originates in the brain.
Collapse
Affiliation(s)
- Małgorzata Drzewiecka
- Laboratory of Medical Genetics Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland (G.B.-P.)
| | - Dominika Jaśniak
- Laboratory of Medical Genetics Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland (G.B.-P.)
| | - Gabriela Barszczewska-Pietraszek
- Laboratory of Medical Genetics Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland (G.B.-P.)
| | - Piotr Czarny
- Department of Medical Biochemistry, Medical University of Lodz, 92-216 Lodz, Poland; (P.C.)
| | - Anna Kobrzycka
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Łodz, 90-236 Lodz, Poland
| | - Marek Wieczorek
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Łodz, 90-236 Lodz, Poland
| | - Maciej Radek
- Department of Neurosurgery, Surgery of Spine and Peripheral Nerves, Medical University of Lodz, University Hospital WAM-CSW, 90-236 Lodz, Poland
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, 92-216 Lodz, Poland; (P.C.)
| | - Tomasz Skorski
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Tomasz Śliwiński
- Laboratory of Medical Genetics Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland (G.B.-P.)
| |
Collapse
|