1
|
Wei Y, Meng Y, Jia K, Lu W, Huang Y, Lu H. Dimethomorph induces heart and vascular developmental defects by disrupting thyroid hormone in zebrafish embryos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 289:117413. [PMID: 39693786 DOI: 10.1016/j.ecoenv.2024.117413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024]
Abstract
Dimethomorph (DMT) is a widely-used selective active fungicide that effectively controls downy mildew, crown rot, and late blight in crops. The extensive application of DMT raises concerns about its ecological impact on non-target organisms in the environment. However, there is limited understanding of the toxicological properties of DMT on these organisms. In this study, we utilized zebrafish as an animal model to assess the toxicity of DMT induced by exposure 5.5-72 hours post-fertilization (hpf). During this period, we monitored and evaluated the development of the zebrafish heart and vascular system. Additionally, embryo samples were collected to perform molecular-level detection of PCNA, oxidative stress, and related genes. The results showed a concentration-dependent decrease in survival rate and hatching rate, shortened body length, slowed heart rate, and pericardial edema, body curvature and reduced eye size as DMT exposure concentration increased. Furthermore, DMT exposure led to impairments in the development of the heart, vascular, along with change in the expression levels of relevant genes. It also caused a decrease in cell proliferation and an increase in oxidative stress levels. Moreover, DMT disrupts the normal development of thyroid follicular cells, leading to a reduction in T3 levels. Thyroid hormone supplementation partially reverses the toxicity induced by DMT, increasing eye size, restoring body length, reducing spine curvature, and reducing pericardial edema. Therefore, we speculate that DMT likely affects the development of zebrafish embryos by disrupting normal thyroid follicle development.
Collapse
Affiliation(s)
- You Wei
- Center for Clinical Medicine Research, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China; Center for drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi 341000, China
| | - Yunlong Meng
- Center for Clinical Medicine Research, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China; Center for drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi 341000, China
| | - Kun Jia
- Center for drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi 341000, China
| | - Weijian Lu
- Center for Clinical Medicine Research, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yushan Huang
- Center for Evidence Based Medical and Clinical Research, First Affiliated Hospital of Gannan Medical University, Ganzhou, China.
| | - Huiqiang Lu
- Center for Clinical Medicine Research, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China.
| |
Collapse
|
2
|
Martínez Sanmiguel JJ, Rincón-López JA, Rangel-López R, Hermann-Muñoz JA, Franco Molina M, Rodríguez Padilla C, Alvarado-Orozco JM, Zárate Triviño DG. In Vitro and In Vivo Biological Properties of Calcium Silicophosphate-Based Bone Grafts: Silicocarnotite and Nagelschmidtite. ACS APPLIED BIO MATERIALS 2024; 7:5318-5336. [PMID: 39029129 DOI: 10.1021/acsabm.4c00538] [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] [Indexed: 07/21/2024]
Abstract
Accidents, trauma, bone defects, and oncological processes significantly impact patients' health and quality of life. While calcium phosphates and bioactive glasses are commonly used as bone fillers to facilitate bone regeneration in orthopedics and traumatology, they exhibit certain disadvantages compared to calcium silicophosphate phases. This study evaluates the in vitro cytocompatibility and in vivo osteogenic properties of two-third-generation ceramic phases: silicocarnotite (SC) and nagelschmidtite (Nagel). These phases were synthesized via a solid-state reaction and characterized using X-ray diffraction and scanning electron microscopy. In vitro behavior was assessed through bioactivity tests, cell viability, proliferation, and inflammatory profiles by detecting cytokines and reactive oxygen species. Osteogenic properties were evaluated by detecting bone-associated proteins in MG-G3, hFOB1.19, and MC3T3-E1 cell lines after 3, 7, and 14 days. 45S5 Bioactive glass (BG), hydroxyapatite (HAp), and osteogenic medium were employed as control standards for bone formation. SC and Nagel phases exhibited higher viability percentages as well as osteoconductive and osteoinductive behavior. Finally, SC and Nagel bone grafts were implanted in a Wistar rat model to assess their in vivo ability to induce bone formation, demonstrating complete osseointegration after 12 weeks. Histological evaluation revealed osteocytes forming osteons and the presence of blood vessels, particularly in rats implanted with Nagel. Given their favorable biological performance, SC and Nagel emerge as promising candidates for bone grafts in orthopedics, traumatology, and maxillofacial surgery.
Collapse
Affiliation(s)
- Juan José Martínez Sanmiguel
- Centro de Ingeniería y Desarrollo Industrial, Avenida Playa Pie de la Cuesta No. 702 Desarrollo San Pablo, Querétaro 76125, México
| | - July Andrea Rincón-López
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad Querétaro Libramiento Norponiente #2000, Querétaro C.P. 76230, México
| | - Raúl Rangel-López
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León 66455, México
| | - Jennifer Andrea Hermann-Muñoz
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad Querétaro Libramiento Norponiente #2000, Querétaro C.P. 76230, México
| | - Moisés Franco Molina
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León 66455, México
| | - Cristina Rodríguez Padilla
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León 66455, México
| | - Juan Manuel Alvarado-Orozco
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad Querétaro Libramiento Norponiente #2000, Querétaro C.P. 76230, México
| | - Diana G Zárate Triviño
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León 66455, México
| |
Collapse
|
3
|
Wu S, Huang J, Li Y. A novel hypoxic lncRNA, LOC110520012 sponges miR-206-y to regulate angiogenesis and liver cell proliferation in rainbow trout (Oncorhynchus mykiss) by targeting vegfaa. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 280:116554. [PMID: 38878335 DOI: 10.1016/j.ecoenv.2024.116554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/28/2024] [Accepted: 06/04/2024] [Indexed: 06/25/2024]
Abstract
Long non-coding RNA (lncRNA) is a novel emerging type of competitive endogenous RNA (ceRNA) that performs key functions in multiple biological processes. However, little is known about the roles of lncRNA under hypoxia stress in fish. Here, vascular endothelial growth factor-Aa (vegfaa) was cloned in rainbow trout (Oncorhynchus mykiss), with the complete cDNA sequence of 2914 bp, encoding 218 amino acids. The molecular weight of the protein was approximately 25.33 kDa, and contained PDGF and VEGF_C domains. Time-course and spatial expression patterns revealed that LOC110520012 was a key regulator of rainbow trout in response to hypoxia stress, and LOC110520012, miR-206-y and vegfaa exhibited a ceRNA regulatory relationship in liver, gill, muscle and rainbow trout liver cells treated with acute hypoxia. Subsequently, the targeting relationship of LOC110520012 and vegfaa with miR-206-y was confirmed by dual-luciferase reporter analysis, and overexpression of LOC110520012 mediated the inhibition of miR-206-y expression in rainbow trout liver cells, while the opposite results were obtained after LOC110520012 silencing with siRNA. We also proved that vegfaa was a target of miR-206-y in vitro and in vivo, and the vegfaa expression and anti-proliferative effect on rainbow trout liver cells regulated by miR-206-y mimics could be reversed by LOC110520012. These results suggested that LOC110520012 can positively regulate vegfaa expression by sponging miR-206-y under hypoxia stress in rainbow trout, which facilitate in-depth understanding of the molecular mechanisms of fish adaptation and tolerance to hypoxia.
Collapse
Affiliation(s)
- Shenji Wu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Jinqiang Huang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China.
| | - Yongjuan Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; College of Science, Gansu Agricultural University, Lanzhou 730070, China
| |
Collapse
|
4
|
Xiao D, Zeng T, Zhu W, Yu ZZ, Huang W, Yi H, Lu SS, Feng J, Feng XP, Wu D, Wen Q, Zhou JH, Yuan L, Zhuang W, Xiao ZQ. ANXA1 Promotes Tumor Immune Evasion by Binding PARP1 and Upregulating Stat3-Induced Expression of PD-L1 in Multiple Cancers. Cancer Immunol Res 2023; 11:1367-1383. [PMID: 37566399 DOI: 10.1158/2326-6066.cir-22-0896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 05/10/2023] [Accepted: 08/08/2023] [Indexed: 08/12/2023]
Abstract
The deregulation of Annexin A1 (ANXA1), a regulator of inflammation and immunity, leads to cancer growth and metastasis. However, whether ANXA1 is involved in cancer immunosuppression is still unclear. Here, we report that ANXA1 knockdown (i) dramatically downregulates programmed cell death-ligand 1 (PD-L1) expression in breast cancer, lung cancer, and melanoma cells; (ii) promotes T cell-mediated killing of cancer cells in vitro; and (iii) inhibits cancer immune escape in immune-competent mice via downregulating PD-L1 expression and increasing the number and killing activity of CD8+ T cells. Mechanistically, ANXA1 functioned as a sponge molecule for interaction of PARP1 and Stat3. Specifically, binding of ANXA1 to PARP1 decreased PARP1's binding to Stat3, which reduced poly(ADP-ribosyl)ation and dephosphorylation of Stat3 and thus, increased Stat3's transcriptional activity, leading to transcriptionally upregulated expression of PD-L1 in multiple cancer cells. In clinical samples, expression of ANXA1 and PD-L1 was significantly higher in breast cancer, non-small cell lung cancer, and skin cutaneous melanoma compared with corresponding normal tissues and positively correlated in cancer tissues. Moreover, using both ANXA1 and PD-L1 proteins for predicting efficacy of anti-PD-1 immunotherapy and patient prognosis was superior to using individual proteins. Our data suggest that ANXA1 promotes cancer immune escape via binding PARP1 and upregulating Stat3-induced expression of PD-L1, that ANXA1 is a potential new target for cancer immunotherapy, and combination of ANXA1 and PD-L1 expression is a potential marker for predicting efficacy of anti-PD-1 immunotherapy in multiple cancers.
Collapse
Affiliation(s)
- Ding Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, China
- The Higher Educational Key Laboratory for Cancer Proteomics and Translational Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ting Zeng
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, China
- The Higher Educational Key Laboratory for Cancer Proteomics and Translational Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Zhu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Zheng-Zheng Yu
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, China
- The Higher Educational Key Laboratory for Cancer Proteomics and Translational Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Huang
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, China
- The Higher Educational Key Laboratory for Cancer Proteomics and Translational Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Hong Yi
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, China
- The Higher Educational Key Laboratory for Cancer Proteomics and Translational Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Shan-Shan Lu
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, China
- The Higher Educational Key Laboratory for Cancer Proteomics and Translational Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Juan Feng
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, China
- The Higher Educational Key Laboratory for Cancer Proteomics and Translational Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Xue-Ping Feng
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, China
- The Higher Educational Key Laboratory for Cancer Proteomics and Translational Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Di Wu
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, China
- The Higher Educational Key Laboratory for Cancer Proteomics and Translational Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Qi Wen
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, China
- The Higher Educational Key Laboratory for Cancer Proteomics and Translational Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Jian-Hua Zhou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Li Yuan
- Department of Nuclear Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wei Zhuang
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhi-Qiang Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, China
- The Higher Educational Key Laboratory for Cancer Proteomics and Translational Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
5
|
Multi-component immune knockout: A strategy for studying the effective components of traditional Chinese medicine. J Chromatogr A 2023; 1692:463853. [PMID: 36780848 DOI: 10.1016/j.chroma.2023.463853] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 02/09/2023]
Abstract
Periploca forrestii Schltr., a traditional Chinese medicine (TCM), is commonly used to treat autoimmune diseases such as rheumatoid arthritis (RA). However, its mechanism, involving a variety of cardiac glycosides, remains largely unknown. The immune knockout strategy can highly selectively deplete target components by immunoaffinity chromatography (IAC). We aimed to identify the common structural features of cardiac glycosides in P. forrestii and design IAC to specifically recognize these features to achieve the multi-component knockout of potential active substances from the extracts of P. forrestii. A content detection experiment confirmed that the content of a compound with periplogenin structure (CPS) in the extract of P. forrestii was reduced by 45% by IAC of periplogenin. The immunosuppressive ability of the extract on H9 human T lymphocytic cells was weakened after CPS knockout from P. forrestii extract. Molecular biology experiments showed that mRNA expression of interferon-γ (IFN-γ), interleukin-2 (IL-2), and interleukin-6 (IL-6) in H9 cells was up-regulated after CPS knockout, while no significant changes in the expression of interleukin-4 (IL-4) were found. CPS knockout from P. forrestii extract did not cause significant changes in the proliferation of lipopolysaccharide (LPS)-stimulated RAW264.7 macrophage cells incubated with this extract. These results indicate that CPS exhibited immunosuppressive effects via inhibiting the T helper 1 (Th1) cell immune response and not via the anti-inflammatory components in P. forrestii. This is the first use of IAC to achieve multi-component knockout in TCM extracts for identifying effective compounds. This method is effective and reliable and warrants further exploration.
Collapse
|
6
|
Enhancing spray-drying tolerance of Lactobacillus bulgaricus via non-sporeforming dormancy induction. INNOV FOOD SCI EMERG 2023. [DOI: 10.1016/j.ifset.2023.103309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
|
7
|
Liu MM, Zhang FF, Liu H, Wu MJ, Liu ZJ, Huang PF. Cell viability and drug evaluation biosensing system based on disposable AuNPs/MWCNT nanocomposite modified screen-printed electrode for exocytosis dopamine detection. Talanta 2023; 254:124118. [PMID: 36470018 DOI: 10.1016/j.talanta.2022.124118] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 11/27/2022]
Abstract
Cell viability, as an important index to evaluate drug effects, usually was measured by tetrazolium colorimetric assay, playing a key role in drug development and drug screening. Tedious operating procedures, unsatisfactory sensitivity and abominable environments perplex researchers to acquire more detailed in vivo-relevant biological information. Herein, a simple and low-cost cell viability and drug evaluation biosensing system-based on multiwalled carbon nanotubes, gold nanoparticles and Nafion modified screen-printed electrode (SPE) biosensor was constructed for detection of dopamine (DA) released from living cells to evaluate cytotoxicity of antineoplastic drugs such as cisplatin and resveratrol. The biosensing system was demonstrated to display exceptional selectivity, excellent flexibility and good stability toward DA measurement in complex bio-samples. Additionally, the satisfactory recoveries of DA in real samples revealed the reliability and accuracy of the biosensing system in practical application. The IC50 curves respectively obtained by the biosensing system and tetrazolium colorimetric assay provided similar IC50 value but distinctly different dose-effect relationship, which confirmed the enormous potential of the biosensor in cell viability and described drug efficacy profiles in cell function. In short, the cell viability and drug evaluation system using SPE biosensor paves a new way in drug screening and pharmaceutical application to measure bioactive molecule such as DA.
Collapse
Affiliation(s)
- Meng-Meng Liu
- Department of Pharmacy, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Feng-Feng Zhang
- Department of Pharmacy, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Hui Liu
- Department of Pharmaceutical Analysis, Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Faculty of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Mei-Juan Wu
- Department of Pharmacy, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Zhou-Jie Liu
- Department of Pharmacy, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
| | - Pin-Fang Huang
- Department of Pharmacy, First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
| |
Collapse
|
8
|
Krassnig SC, Mäser M, Probst NA, Werner J, Schlett C, Schumann N, von Scheven G, Mangerich A, Bürkle A. Comparative analysis of chlorambucil-induced DNA lesion formation and repair in a spectrum of different human cell systems. Toxicol Rep 2023; 10:171-189. [PMID: 36714466 PMCID: PMC9881385 DOI: 10.1016/j.toxrep.2023.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/21/2023] Open
Abstract
Chlorambucil (CLB) belongs to the class of nitrogen mustards (NMs), which are highly reactive bifunctional alkylating agents and were the first chemotherapeutic agents developed. They form DNA interstrand crosslinks (ICLs), which cause a blockage of DNA strand separation, inhibiting essential processes in DNA metabolism like replication and transcription. In fast replicating cells, e.g., tumor cells, this can induce cell death. The upregulation of ICL repair is thought to be a key factor for the resistance of tumor cells to ICL-inducing cytostatic agents including NMs. To monitor induction and repair of CLB-induced ICLs, we adjusted the automated reversed fluorometric analysis of alkaline DNA unwinding assay (rFADU) for the detection of ICLs in adherent cells. For the detection of monoalkylated DNA bases we established an LC-MS/MS method. We performed a comparative analysis of adduct formation and removal in five human cell lines and in peripheral blood mononuclear cells (PBMCs) after treatment with CLB. Dose-dependent increases in adduct formation were observed, and suitable treatment concentrations were identified for each cell line, which were then used for monitoring the kinetics of adduct formation. We observed significant differences in the repair kinetics of the cell lines tested. For example, in A2780 cells, hTERT immortalized VH10 cells, and in PBMCs a time-dependent repair of the two main monoalkylated DNA-adducts was confirmed. Regarding ICLs, repair was observed in all cell systems except for PBMCs. In conclusion, LC-MS/MS analyses combined with the rFADU technique are powerful tools to study the molecular mechanisms of NM-induced DNA damage and repair. By applying these methods to a spectrum of human cell systems of different origin and transformation status, we obtained insight into the cell-type specific repair of different CLB-induced DNA lesions, which may help identify novel resistance mechanisms of tumors and define molecular targets for therapeutic interventions.
Collapse
Key Words
- BER, base excision repair
- CLB, chlorambucil
- Chlorambucil
- DNA repair kinetics
- ICL, interstrand crosslink
- Interstrand crosslink
- MS, mass spectrometry
- Mass spectrometry
- Monoalkylated DNA adducts
- NER, nucleotide excision repair
- NM, Nitrogen mustard
- Nitrogen mustard
- PBMCs, peripheral blood mononuclear cells
- PI, propidium iodide
- RPE-1, human retinal pigment epithelial
- SD, standard deviation
- VH10, human foreskin fibroblasts
- dG, 2'-deoxyguanosine
- hTERT, human telomerase reverse transcriptase
- rFADU, reverse fluorometric analysis of alkaline DNA unwinding
Collapse
Affiliation(s)
- Sarah Ceylan Krassnig
- Molecular Toxicology, Department of Biology, University of Konstanz, D-78464 Konstanz, Germany
| | - Marina Mäser
- Molecular Toxicology, Department of Biology, University of Konstanz, D-78464 Konstanz, Germany
| | - Nicola Anna Probst
- Molecular Toxicology, Department of Biology, University of Konstanz, D-78464 Konstanz, Germany
| | - Jens Werner
- Molecular Toxicology, Department of Biology, University of Konstanz, D-78464 Konstanz, Germany
| | - Charlotte Schlett
- Molecular Toxicology, Department of Biology, University of Konstanz, D-78464 Konstanz, Germany
| | - Nina Schumann
- Molecular Toxicology, Department of Biology, University of Konstanz, D-78464 Konstanz, Germany
| | - Gudrun von Scheven
- Molecular Toxicology, Department of Biology, University of Konstanz, D-78464 Konstanz, Germany
| | - Aswin Mangerich
- Molecular Toxicology, Department of Biology, University of Konstanz, D-78464 Konstanz, Germany
- Nutritional Toxicology, Institute of Nutritional Science, University of Potsdam, D-14558 Nuthetal, Germany
| | - Alexander Bürkle
- Molecular Toxicology, Department of Biology, University of Konstanz, D-78464 Konstanz, Germany
| |
Collapse
|
9
|
Knockdown of Girdin Induced Apoptosis of Glioblastoma Cells via the Mitochondrion Signaling Pathway. Neuroscience 2022; 500:95-105. [PMID: 35952993 DOI: 10.1016/j.neuroscience.2022.07.025] [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: 02/17/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/23/2022]
Abstract
Glioblastoma is the most common primary brain tumor with poor survival rate and without effective treatment strategy. However, the influence of Girdin on human glioblastoma and the underlying molecular mechanisms have yet to be uncovered. We mainly investigated the role of Girdin in glioblastoma cells apoptosis. First, we examined Girdin expression in 90 glioma patients by using immunohistochemistry (IHC) and analyzed its association with patients' prognosis. The results showed that the expression of Girdin was positively associated with the histological grade of glioma, and glioma patients with high Girdin expression had a poor prognosis. Next, proliferation assay, Colony formation assay and Cell Counting Kit-8 (CCK-8) results showed that knockdown of Girdin suppressed proliferation and reduced cell survival rate. Flow cytometry and DAPI staining results showed that knockdown of Girdin induced apoptosis in LN229 cells. Western blot results suggested that reduction of Gridin increased the level of Cytochrome C (Cyt-C) and Bad while decreased the expression of Bcl-2 and p-AKT. Moreover, subcutaneous mouse xenograft model was used to validate the role of Girdin in glioblastoma apoptosis. Consistently, in vivo assays showed that knockdown of Girdin inhibited the growth of the grafted tumor and increased the level of Cyt-C and Bad. These findings demonstrated that knockdown of Girdin may induce Bad expression and reduce Bcl-2 expression by inhibiting the activation of AKT, leading to the release of Cyt-C from mitochondria, thereby promoting glioblastoma cells apoptosis.
Collapse
|
10
|
Aini S, Bolati S, Ding W, Liu S, Su P, Aili S, Naman Y, Xuekelaiti K. LncRNA SNHG10 suppresses the development of doxorubicin resistance by downregulating miR-302b in triple-negative breast cancer. Bioengineered 2022; 13:11430-11439. [PMID: 35506202 PMCID: PMC9275935 DOI: 10.1080/21655979.2022.2063592] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Unlike other types of breast cancer, triple negative breast cancer (TNBC) does not respond to therapies targeting human epidermal growth factor receptor-2 (HER2) or hormone therapy, and the prognosis of patients with TNBC is usually poor. The role of long non-coding RNA (lncRNA) small nucleolar RNA host gene 10 (SNHG10) has been investigated in many types of cancer, but its role in TNBC is unknown. This study aimed to explore the role of SNHG10 in TNBC in the context of doxorubicin treatment, a common therapy for TNBC. Analysis of the TCGA dataset revealed the downregulation of SNHG10 in TNBC. The downregulation of SNHG10 of TNBC in TNBC was further confirmed by detecting its expression in 60 patients with TNBC by qPCR. The expression of SNHG10 was further downregulated after doxorubicin treatment. In TNBC, microRNA-302b (miR-302b) was downregulated and was positively correlated with SNHG10. In TNBC cells, overexpression of SNHG10 resulted in upregulation of miR-302b, and methylation-specific PCR analysis showed that SNHG10 negatively regulates the methylation of miR-302b. In addition, doxorubicin treatment resulted in the downregulation of SNHG10 in TNBC cells, and overexpression of SNHG10 and miR-302b promoted apoptosis of doxorubicin-treated TNBC cells. Furthermore, overexpression of both SNHG10 and miR-302b had a stronger effect on apoptosis than that of overexpression of SNHG10 alone. Our study showed that SNHG10 could inhibit the development of resistance to doxorubicin by upregulating miR-302b in TNBC through methylation. Our findings suggested that SNHG10 might serve as a molecular target for intervening in TBNC metastasis.
Collapse
Affiliation(s)
| | | | - Wei Ding
- Department of Mammary Gland and Thyroid Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
| | - Siyin Liu
- Department of Mammary Gland and Thyroid Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
| | - Pengcheng Su
- Department of Mammary Gland and Thyroid Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
| | - Saiding Aili
- Department of Mammary Gland and Thyroid Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
| | - Yimin Naman
- Department of Mammary Gland and Thyroid Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
| | - Kuerban Xuekelaiti
- Department of Mammary Gland and Thyroid Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
| |
Collapse
|
11
|
Lemieszek MB, Findlay SD, Siegers GM. CellTrace™ Violet Flow Cytometric Assay to Assess Cell Proliferation. Methods Mol Biol 2022; 2508:101-114. [PMID: 35737236 DOI: 10.1007/978-1-0716-2376-3_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
CellTrace™ Violet (CTV) is a powerful tool for tracking cell proliferation by permanently binding cellular proteins and rendering the cell fluorescent. After cell division, each daughter cell contains half of the parent cell's fluorescence, enabling quantification of proliferation via flow cytometry. This method enables monitoring of several generations of cell division and tracking of different cell populations in co-culture. Here we describe the use of CellTrace™ Violet in different cell types, and we share important observations we made during protocol optimization.
Collapse
Affiliation(s)
- Marina B Lemieszek
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Scott D Findlay
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | | |
Collapse
|
12
|
Boibessot C, Molina O, Lachance G, Tav C, Champagne A, Neveu B, Pelletier J, Pouliot F, Fradet V, Bilodeau S, Fradet Y, Bergeron A, Toren P. Subversion of infiltrating prostate macrophages to a mixed immunosuppressive tumor-associated macrophage phenotype. Clin Transl Med 2022; 12:e581. [PMID: 35075795 PMCID: PMC8786699 DOI: 10.1002/ctm2.581] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 09/05/2021] [Accepted: 09/07/2021] [Indexed: 12/21/2022] Open
Abstract
Tumor-associated macrophages (TAMs) support tumor progression within the tumor microenvironment (TME). Many questions remain as to the origin, development, and function of TAMs within the prostate TME. Evaluation of TAMs in prostate cancer (PCa) patients identified the immunosuppressive TAM marker CD163 in adjacent normal epithelium as an independent predictor of metastases or PCa death. Flow cytometry analyses identified prostate TAMs as frequently expressing both proinflammatory M1 (CCR7+) and immunosuppressive M2 (CD163+) markers. In vitro, we demonstrate PCa cells similarly subvert human M1 macrophages toward a mixed M1/M2 macrophage phenotype favoring tumor growth. Further the cytokine milieu-induced transition between immunosuppressive M2 to proinflammatory M1 (M2→M1) macrophages is abrogated by the presence of PCa cells. RNA sequencing suggests alterations in chemokine expression in prostate TAMs due to the presence of PCa cells. Together, our results suggest that prostate TAMs originate from inflammatory infiltrating macrophages, which are then reprogrammed mainly by PCa cells, but also the cytokine milieu. A better understanding of this subversion of macrophages within the prostate may lead to novel treatment strategies.
Collapse
Affiliation(s)
- Clovis Boibessot
- Centre de recherche du CHU de Québec—Université LavalAxe OncologieQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
| | - Oscar Molina
- Centre de recherche du CHU de Québec—Université LavalAxe OncologieQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
| | - Gabriel Lachance
- Centre de recherche du CHU de Québec—Université LavalAxe OncologieQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
| | - Christophe Tav
- Centre de recherche du CHU de Québec—Université LavalAxe OncologieQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
- Centre de Recherche en Données Massives de l'Université LavalQuébecCanada
| | - Audrey Champagne
- Centre de recherche du CHU de Québec—Université LavalAxe OncologieQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
| | - Bertrand Neveu
- Centre de recherche du CHU de Québec—Université LavalAxe OncologieQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
| | - Jean‐François Pelletier
- Centre de recherche du CHU de Québec—Université LavalAxe OncologieQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
| | - Frédéric Pouliot
- Centre de recherche du CHU de Québec—Université LavalAxe OncologieQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
- Département de chirurgieUniversité LavalQuébecCanada
| | - Vincent Fradet
- Centre de recherche du CHU de Québec—Université LavalAxe OncologieQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
- Département de chirurgieUniversité LavalQuébecCanada
| | - Steve Bilodeau
- Centre de recherche du CHU de Québec—Université LavalAxe OncologieQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
- Centre de Recherche en Données Massives de l'Université LavalQuébecCanada
- Département de biologie moléculairebiochimie médicale et pathologieFaculté de MédecineUniversité LavalQuébecCanada
| | - Yves Fradet
- Centre de recherche du CHU de Québec—Université LavalAxe OncologieQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
- Département de chirurgieUniversité LavalQuébecCanada
| | - Alain Bergeron
- Centre de recherche du CHU de Québec—Université LavalAxe OncologieQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
- Département de chirurgieUniversité LavalQuébecCanada
| | - Paul Toren
- Centre de recherche du CHU de Québec—Université LavalAxe OncologieQuébecCanada
- Centre de recherche sur le cancer de l'Université LavalQuébecCanada
- Département de chirurgieUniversité LavalQuébecCanada
| |
Collapse
|
13
|
Xu B, Liu F, Gao Y, Sun J, Li Y, Lin Y, Liu X, Wen Y, Yi S, Dang J, Tu P, Wang Y. High Expression of IKZF2 in Malignant T Cells Promotes Disease Progression in Cutaneous T Cell Lymphoma. Acta Derm Venereol 2021; 101:adv00613. [PMID: 34853863 PMCID: PMC9472098 DOI: 10.2340/actadv.v101.570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Cutaneous T cell lymphoma is a generally indolent disease derived from skin-homing mature T cells. However, in advanced stages, cutaneous T cell lymphoma may manifest aggressive clinical behaviour and lead to a poor prognosis. The mechanism of disease progression in cutaneous T cell lymphoma remains unknown. This study, based on a large clinical cohort, found that IKZF2, an essential transcription factor during T cell development and differentiation, showed stage-dependent overexpression in the malignant T cells in mycosis fungoides lesions. IKZF2 is specifically over-expressed in advanced-stage mycosis fungoides lesions, and correlates with poor prognosis. Mechanistically, overexpression of IKZF2 promotes cutaneous T cell lymphoma progression via inhibiting malignant cell apoptosis and may contribute to tumour immune escape by downregulating major histocompatibility complex II molecules and up-regulating the production of anti-inflammatory cytokine interleukin-10 by malignant T cells. These results demonstrate the important role of IKZF2 in high-risk cutaneous T cell lymphoma and pave the way for future targeted therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Yang Wang
- Department of Dermatology and Venereology, Peking University First Hospital, No.8 Xishiku Street, Xi Cheng District, Beijing 100034, China.
| |
Collapse
|
14
|
Karimnia N, Wilson AL, Green E, Matthews A, Jobling TW, Plebanski M, Bilandzic M, Stephens AN. Chemoresistance is mediated by ovarian cancer leader cells in vitro. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:276. [PMID: 34470672 PMCID: PMC8408956 DOI: 10.1186/s13046-021-02086-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/25/2021] [Indexed: 11/26/2022]
Abstract
Background Leader cells are a subset of cancer cells that coordinate the complex cell-cell and cell-matrix interactions required for ovarian cancer migration, invasion, tumour deposition and are negatively associated with progression-free survival and response to therapy. Emerging evidence suggests leader cells may be enriched in response to chemotherapy, underlying disease recurrence following treatment. Methods CRISPR was used to insert a bicistronic T2A-GFP cassette under the native KRT14 (leader cell) promoter. 2D and 3D drug screens were completed in the presence of chemotherapies used in ovarian cancer management. Leader cell; proliferative (Ki67); and apoptotic status (Cleaved Caspase 3) were defined by live cell imaging and flow cytometry. Quantitative real-time PCR defined “stemness” profiles. Proliferation was assessed on the xCELLigence real time cell analyser. Statistical Analysis was performed using unpaired non-parametric t-tests or one-way ANOVA and Tukey’s multiple comparison post hoc. Results Leader cells represent a transcriptionally plastic subpopulation of ovarian cancer cells that arise independently of cell division or DNA replication, and exhibit a “stemness” profile that does not correlate with epithelial-to-mesenchymal transition. Chemotherapeutics increased apoptosis-resistant leader cells in vitro, who retained motility and expressed known chemo-resistance markers including ALDH1, Twist and CD44v6. Functional impairment of leader cells restored chemosensitivity, with leader cell-deficient lines failing to recover following chemotherapeutic intervention. Conclusions Our data demonstrate that ovarian cancer leader cells are resistant to a diverse array of chemotherapeutic agents, and are likely to play a critical role in the recurrence of chemo-resistant disease as drivers of poor treatment outcomes. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02086-3.
Collapse
Affiliation(s)
- Nazanin Karimnia
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, VIC, 3168, Australia
| | - Amy L Wilson
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, VIC, 3168, Australia
| | - Emma Green
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, VIC, 3168, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| | - Amelia Matthews
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, VIC, 3168, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| | - Thomas W Jobling
- Monash Medical Centre, Department of Gynaecology Oncology, Monash Health, Moorabbin, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | - Maree Bilandzic
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, VIC, 3168, Australia.
| | - Andrew N Stephens
- Hudson Institute of Medical Research, 27-31 Wright St, Clayton, VIC, 3168, Australia
| |
Collapse
|
15
|
Ziółkowska-Suchanek I, Podralska M, Żurawek M, Łaczmańska J, Iżykowska K, Dzikiewicz-Krawczyk A, Rozwadowska N. Hypoxia-Induced FAM13A Regulates the Proliferation and Metastasis of Non-Small Cell Lung Cancer Cells. Int J Mol Sci 2021; 22:ijms22094302. [PMID: 33919074 PMCID: PMC8122400 DOI: 10.3390/ijms22094302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/13/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022] Open
Abstract
Hypoxia in non-small cell lung cancer (NSCLC) affects cancer progression, metastasis and metabolism. We previously showed that FAM13A was induced by hypoxia in NSCLC but the biological function of this gene has not been fully elucidated. This study aimed to investigate the role of hypoxia-induced FAM13A in NSCLC progression and metastasis. Lentiviral shRNAs were used for FAM13A gene silencing in NSCLC cell lines (A549, CORL-105). MTS assay, cell tracking VPD540 dye, wound healing assay, invasion assay, BrdU assay and APC Annexin V staining assays were performed to examine cell proliferation ability, migration, invasion and apoptosis rate in NSCLC cells. The results of VPD540 dye and MTS assays showed a significant reduction in cell proliferation after FAM13A knockdown in A549 cells cultured under normal and hypoxia (1% O2) conditions (p < 0.05), while the effect of FAM13A downregulation on CORL-105 cells was observed after 96 h exposition to hypoxia. Moreover, FAM13A inhibition induced S phase cell cycle arrest in A549 cells under hypoxia conditions. Silencing of FAM13A significantly suppressed migration of A549 and CORL-105 cells in both oxygen conditions, especially after 72 and 96 h (p < 0.001 in normoxia, p < 0.01 after hypoxia). It was showed that FAM13A reduction resulted in disruption of the F-actin cytoskeleton altering A549 cell migration. Cell invasion rates were significantly decreased in A549 FAM13A depleted cells compared to controls (p < 0.05), mostly under hypoxia. FAM13A silencing had no effect on apoptosis induction in NSCLC cells. In the present study, we found that FAM13A silencing has a negative effect on proliferation, migration and invasion activity in NSCLC cells in normal and hypoxic conditions. Our data demonstrated that FAM13A depleted post-hypoxic cells have a decreased cell proliferation ability and metastatic potential, which indicates FAM13A as a potential therapeutic target in lung cancer.
Collapse
|
16
|
Long noncoding RNA GAS8-AS1: A novel biomarker in human diseases. Biomed Pharmacother 2021; 139:111572. [PMID: 33838502 DOI: 10.1016/j.biopha.2021.111572] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/21/2021] [Accepted: 03/31/2021] [Indexed: 12/16/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) represent a group of ncRNAs with more than 200 nucleotides. These RNAs can specifically regulate gene expression at both the transcriptional and the post-transcriptional levels, and increasing evidence indicates that they play vital roles in a variety of disease-related cellular processes. The lncRNA GAS8 antisense RNA 1 (GAS8-AS1, also known as C16orf3) is located in the second intron of GAS8 and has been reported to be both abnormally expressed in several diseases and closely correlated with many clinical characteristics. GAS8-AS1 has been shown to affect many biological functions, including cell proliferation, migration, invasiveness, and autophagy using several signaling pathways. In this review, we have summarized current studies on GAS8-AS1 roles in disease and discuss its potential clinical utility. GAS8-AS1 may be a promising biomarker for both diagnoses and prognoses, and a novel target for many disease therapies.
Collapse
|
17
|
Partscht P, Uddin B, Schiebel E. Human cells lacking CDC14A and CDC14B show differences in ciliogenesis but not in mitotic progression. J Cell Sci 2021; 134:224108. [PMID: 33328327 DOI: 10.1242/jcs.255950] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/10/2020] [Indexed: 11/20/2022] Open
Abstract
The budding yeast phosphatase Cdc14 has a central role in mitotic exit and cytokinesis. Puzzlingly, a uniform picture for the three human CDC14 paralogues CDC14A, CDC14B and CDC14C in cell cycle control has not emerged to date. Redundant functions between the three CDC14 phosphatases could explain this unclear picture. To address the possibility of redundancy, we tested expression of CDC14 and analysed cell cycle progression of cells with single and double deletions in CDC14 genes. Our data suggest that CDC14C is not expressed in human RPE1 cells, excluding a function in this cell line. Single- and double-knockouts (KO) of CDC14A and CDC14B in RPE1 cells indicate that both phosphatases are not important for the timing of mitotic phases, cytokinesis and cell proliferation. However, cycling CDC14A KO and CDC14B KO cells show altered ciliogenesis compared to wild-type cells. The cilia of cycling CDC14A KO cells are longer, whereas CDC14B KO cilia are more frequent and disassemble faster. In conclusion, this study demonstrates that the cell cycle functions of CDC14 proteins are not conserved between yeast and human cells.
Collapse
Affiliation(s)
- Patrick Partscht
- Zentrum für Molekulare Biologie, Universität Heidelberg, DKFZ-ZMBH Allianz, Heidelberg 69120, Germany.,Heidelberg Biosciences International Graduate School (HBIGS), Universität Heidelberg, Heidelberg, Germany
| | - Borhan Uddin
- Zentrum für Molekulare Biologie, Universität Heidelberg, DKFZ-ZMBH Allianz, Heidelberg 69120, Germany
| | - Elmar Schiebel
- Zentrum für Molekulare Biologie, Universität Heidelberg, DKFZ-ZMBH Allianz, Heidelberg 69120, Germany
| |
Collapse
|
18
|
DEAD-Box Helicase 4 (Ddx4) + Stem Cells Sustain Tumor Progression in Non-Serous Ovarian Cancers. Int J Mol Sci 2020; 21:ijms21176096. [PMID: 32847044 PMCID: PMC7503840 DOI: 10.3390/ijms21176096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/14/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022] Open
Abstract
DEAD-Box Helicase 4 (Ddx4)+ ovarian stem cells are able to differentiate into several cell types under appropriate stimuli. Ddx4 expression has been correlated with poor prognosis of serous ovarian cancer (OC), while the potential role of Ddx4+ cells in non-serous epithelial OC (NS-EOC) is almost unexplored. The aim of this study was to demonstrate the presence of Ddx4+ cells in NS-EOC and investigate the effect of follicle-stimulating hormone (FSH) on this population. Increased Ddx4 expression was demonstrated in samples from patients with advanced NS-EOC, compared to those with early-stage disease. Under FSH stimulation, OC-derived Ddx4+ cells differentiated into mesenchymal-like (ML) cells, able to deregulate genes involved in cell migration, invasiveness, stemness and chemoresistance in A2780 OC cells. This effect was primarily induced by ML-cells deriving from advanced NS-EOC, suggesting that a tumor-conditioned germ cell niche inhabits its microenvironment and is able to modulate, in a paracrine manner, tumor cell behavior through transcriptome modulation.
Collapse
|
19
|
Hanna CB, Mudaliar D, John K, Allen CL, Sun L, Hawkinson JE, Schönbrunn E, Georg GI, Jensen JT. Development of WEE2 kinase inhibitors as novel non-hormonal female contraceptives that target meiosis†. Biol Reprod 2020; 103:368-377. [PMID: 32667031 PMCID: PMC7401407 DOI: 10.1093/biolre/ioaa097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 05/26/2020] [Indexed: 11/24/2022] Open
Abstract
WEE2 oocyte meiosis inhibiting kinase is a well-conserved oocyte specific kinase with a dual regulatory role during meiosis. Active WEE2 maintains immature, germinal vesicle stage oocytes in prophase I arrest prior to the luteinizing hormone surge and facilitates exit from metaphase II arrest at fertilization. Spontaneous mutations at the WEE2 gene locus in women have been linked to total fertilization failure indicating that selective inhibitors to this kinase could function as non-hormonal contraceptives. Employing co-crystallization with WEE1 G2 checkpoint kinase inhibitors, we revealed the structural basis of action across WEE kinases and determined type I inhibitors were not selective to WEE2 over WEE1. In response, we performed in silico screening by FTMap/FTSite and Schrodinger SiteMap analysis to identify potential allosteric sites, then used an allosterically biased activity assay to conduct high-throughput screening of a 26 000 compound library containing scaffolds of known allosteric inhibitors. Resulting hits were validated and a selective inhibitor that binds full-length WEE2 was identified, designated GPHR-00336382, along with a fragment-like inhibitor that binds the kinase domain, GPHR-00355672. Additionally, we present an in vitro testing workflow to evaluate biological activity of candidate WEE2 inhibitors including; (1) enzyme-linked immunosorbent assays measuring WEE2 phosphorylation activity of cyclin dependent kinase 1 (CDK1; also known as cell division cycle 2 kinase, CDC2), (2) in vitro fertilization of bovine ova to determine inhibition of metaphase II exit, and (3) cell-proliferation assays to look for off-target effects against WEE1 in somatic (mitotic) cells.
Collapse
Affiliation(s)
- Carol B Hanna
- Oregon National Primate Research Center, Beaverton, Division of Reproductive & Developmental Sciences OR, USA
- Correspondence: Oregon Health & Science University, 505 NW 185th Ave, Beaverton, OR 97006, USA. Tel: +1-503-346-5000; Fax: +1-503-346-5585; E-mail:
| | - Deepti Mudaliar
- University of Minnesota, Department of Obstetrics & Gynecology, Minneapolis, MN, USA
| | - Kristen John
- University of Minnesota, Department of Obstetrics & Gynecology, Minneapolis, MN, USA
| | - C Leigh Allen
- University of Minnesota, Department of Obstetrics & Gynecology, Minneapolis, MN, USA
| | - Luxin Sun
- Moffitt Cancer Center, Drug Discovery Department, Tampa, FL, USA
| | - Jon E Hawkinson
- University of Minnesota, Department of Obstetrics & Gynecology, Minneapolis, MN, USA
| | - Ernst Schönbrunn
- Moffitt Cancer Center, Drug Discovery Department, Tampa, FL, USA
| | - Gunda I Georg
- University of Minnesota, Department of Obstetrics & Gynecology, Minneapolis, MN, USA
| | - Jeffrey T Jensen
- Oregon National Primate Research Center, Beaverton, Division of Reproductive & Developmental Sciences OR, USA
- Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
20
|
Pereira TF, Levin G, DeOcesano-Pereira C, Caodaglio AS, Fujita A, Tonso A, Sogayar MC. Fluorescence-based method is more accurate than counting-based methods for plotting growth curves of adherent cells. BMC Res Notes 2020; 13:57. [PMID: 32019595 PMCID: PMC7001368 DOI: 10.1186/s13104-020-4914-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 01/22/2020] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE Cell growth curves constitute one of the primary assays employed to analyze cell proliferation dynamics of in vitro cultured cells under specific culture conditions. From the cell growth curve, it is possible to assess the behavior of proliferating cells under different conditions, such as drug treatment and genomic editions. Traditionally, growth curves for adherent cells are obtained by seeding the cells in multiple-well plates and counting the total number of cells at different time points. Here, we compare this traditional method to the fluorescence-based method, which is based on the CFSE fluorescence decay over time. RESULTS The fluorescence-based method is not dependent on the determination of the total number of cells, but rather is approached by assessing the fluorescence of a sample of single cells from a cell population at different time points after plating. Therefore, this method is not biased due to either cell loss during harvesting or to the presence of cellular debris and cell clumps. Moreover, the fluorescence-based method displays lower variation among different measurements of the same time point, which increases the reliability on the determination of lag, log and stationary phase transitions.
Collapse
Affiliation(s)
- Túlio Felipe Pereira
- Cell and Molecular Therapy Center (NUCEL), Department of Internal Medicine, School of Medicine, University of São Paulo, Rua Pangaré, 100, Cidade Universitária, São Paulo, SP, 05360-130, Brazil
- Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, SP, Brazil
| | - Gabriel Levin
- Cell and Molecular Therapy Center (NUCEL), Department of Internal Medicine, School of Medicine, University of São Paulo, Rua Pangaré, 100, Cidade Universitária, São Paulo, SP, 05360-130, Brazil
| | - Carlos DeOcesano-Pereira
- Cell and Molecular Therapy Center (NUCEL), Department of Internal Medicine, School of Medicine, University of São Paulo, Rua Pangaré, 100, Cidade Universitária, São Paulo, SP, 05360-130, Brazil
- Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo, SP, Brazil
| | - Amanda Schiersner Caodaglio
- Cell and Molecular Therapy Center (NUCEL), Department of Internal Medicine, School of Medicine, University of São Paulo, Rua Pangaré, 100, Cidade Universitária, São Paulo, SP, 05360-130, Brazil
| | - André Fujita
- Department of Computer Science, Institute of Mathematics and Statistics, University of São Paulo, São Paulo, SP, Brazil
| | - Aldo Tonso
- Department of Chemical Engineering, Polytechnic School, University of São Paulo, São Paulo, SP, Brazil
| | - Mari Cleide Sogayar
- Cell and Molecular Therapy Center (NUCEL), Department of Internal Medicine, School of Medicine, University of São Paulo, Rua Pangaré, 100, Cidade Universitária, São Paulo, SP, 05360-130, Brazil.
- Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
21
|
Chen S, Gong P, Zhang J, Shan Y, Han X, Zhang L. Quantitative analysis of Lactobacillus delbrueckii subsp. bulgaricus cell division and death using fluorescent dye tracking. J Microbiol Methods 2020; 169:105832. [DOI: 10.1016/j.mimet.2020.105832] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/29/2019] [Accepted: 01/01/2020] [Indexed: 12/30/2022]
|
22
|
Novel [1,2,3]triazolo[1,5-a]pyridine derivatives are trypanocidal by sterol biosynthesis pathway alteration. Future Med Chem 2019; 11:1137-1155. [PMID: 31280672 DOI: 10.4155/fmc-2018-0242] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Aim: To study a new series of [1,2,3]triazolo[1,5-α]pyridine derivatives as trypanocidal agents because current antichagasic pharmacologic therapy is only partially effective. Materials & methods: The effect of the series upon Trypanosoma cruzi epimastigotes and murine macrophages viability, cell cycle, cell death and on the metabolites of the sterol biosynthesis pathway was measured; also, docking in 14α-demethylase was analyzed. Results: Compound 16 inhibits 14α-demethylase producing an imbalance in the cholesterol/ergosterol synthesis pathway, as suggested by a metabolic control and theoretical docking analysis. Consequently, it prevented cell proliferation, stopping the cellular cycle at the G2/M phase, inducing cell death. Conclusion: Although the exact cell death mechanism remained elusive, this series can be used for the further rational design of novel antiparasitic molecules.
Collapse
|
23
|
Effect of single and multiple doses of low-level laser therapy on viability and proliferation of stem cells from human exfoliated deciduous teeth (SHED). Lasers Med Sci 2019; 34:1917-1924. [PMID: 31267320 DOI: 10.1007/s10103-019-02836-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 06/25/2019] [Indexed: 01/09/2023]
Abstract
The present study aimed to evaluate in vitro whether the low-level laser (LLL) delivering fractionated total energy (multiple irradiation) or single irradiation stimulates regeneration-associated events (viability and proliferation) in stem cells from human exfoliated deciduous teeth (SHED). Cells received LLL irradiation (InGaAlP-660 nm), according to the following experimental groups: G1 (single irradiation 2.5 J/cm2, 10 mW, 10 s, 0.10 J), G2 (single irradiation 5.0 J/cm2, 10 mW, 20 s, 0.20 J), G3 (single irradiation 7.5 J/cm2, 10 mW, 30 s, 0.30 J), G4 (two irradiations 2.5 J/cm2, 10 mW, 10 s; total energy 0.20 J), G5 (three irradiations 2.5 J/cm2, 10 mW, 10 s; total energy 0.30 J), and G6 (non-irradiated). Cell viability was assessed by MTT and trypan blue exclusion (TBE) methods, while cell proliferation was evaluated by crystal violet (CV) and sulforhodamine B (SRB) assays after 24, 48, and 72 h after the first irradiation. By MTT, there was no difference between groups at 24 and 72 h. At 48 h, the groups subjected to multiple irradiation (G4 and G5) presented higher cell viability rates. The average percentages of viable cells for all groups by TBE method were 91.04%, 96.63%, and 97.48% at 24, 48, and 72 h, respectively. By CV, there was no significant difference between groups at 24 and 48 h; at 72 h, G2, G3, and G4 presented higher cell proliferation. By SRB, G1 and G4 presented lower proliferation rates in all the periods. When the groups presenting the same total energy were compared, G2 (0.20 J) presented lower cell viability rates and higher cell proliferation rates in comparison with G4; G3 (0.30 J) presented similar results to those of G5, with higher cell viability and proliferation. The application of laser delivering fractionated total energy (two or three applications of 2.5 J/cm2) induced higher cell viability at 48 h, while the single irradiation with 2.5 J/cm2 did not stimulate metabolic activity in such period and the proliferation over time. The 5.0 and 7.5 J/cm2 single doses and the three applications of 2.5 J/cm2 maintained cell viability and stimulated proliferation of SHED at 72 h.
Collapse
|
24
|
Abstract
The prevalence of fungal infections has seen a rise in the past decades due to advances in modern medicine leading to an expanding population of device-associated and immunocompromised patients. Furthermore, the spectrum of pathogenic fungi has changed, with the emergence of multidrug-resistant strains such as C. auris. High mortality related to fungal infections points to major limitations of current antifungal therapy and an unmet need for new antifungal drugs. We screened a library of repurposed FDA-approved inhibitors to identify compounds with activities against a diverse range of fungi in varied phases of growth. The assays identified alexidine dihydrochloride (AXD) to have pronounced antifungal activity, including against preformed biofilms, at concentrations lower than mammalian cell toxicity. AXD potentiated the activity of fluconazole and amphotericin B against Candida biofilms in vitro and prevented biofilm growth in vivo. Thus, AXD has the potential to be developed as a pan-antifungal, antibiofilm drug. Invasive fungal infections due to Candida albicans, Aspergillus fumigatus, and Cryptococcus neoformans constitute a substantial threat to hospitalized immunocompromised patients. Further, the presence of drug-recalcitrant biofilms on medical devices and emergence of drug-resistant fungi, such as Candida auris, introduce treatment challenges with current antifungal drugs. Worse, currently there is no approved drug capable of obviating preformed biofilms, which increase the chance of infection relapses. Here, we screened a small-molecule New Prestwick Chemical Library, consisting of 1,200 FDA-approved off-patent drugs against C. albicans, C. auris, and A. fumigatus, to identify those that inhibit growth of all three pathogens. Inhibitors were further prioritized for their potency against other fungal pathogens and their ability to kill preformed biofilms. Our studies identified the bis-biguanide alexidine dihydrochloride (AXD) as a drug with the highest antifungal and antibiofilm activity against a diverse range of fungal pathogens. Finally, AXD significantly potentiated the efficacy of fluconazole against biofilms, displayed low mammalian cell toxicity, and eradicated biofilms growing in mouse central venous catheters in vivo, highlighting its potential as a pan-antifungal drug. IMPORTANCE The prevalence of fungal infections has seen a rise in the past decades due to advances in modern medicine leading to an expanding population of device-associated and immunocompromised patients. Furthermore, the spectrum of pathogenic fungi has changed, with the emergence of multidrug-resistant strains such as C. auris. High mortality related to fungal infections points to major limitations of current antifungal therapy and an unmet need for new antifungal drugs. We screened a library of repurposed FDA-approved inhibitors to identify compounds with activities against a diverse range of fungi in varied phases of growth. The assays identified alexidine dihydrochloride (AXD) to have pronounced antifungal activity, including against preformed biofilms, at concentrations lower than mammalian cell toxicity. AXD potentiated the activity of fluconazole and amphotericin B against Candida biofilms in vitro and prevented biofilm growth in vivo. Thus, AXD has the potential to be developed as a pan-antifungal, antibiofilm drug.
Collapse
|
25
|
Podszywalow-Bartnicka P, Kominek A, Wolczyk M, Kolba MD, Swatler J, Piwocka K. Characteristics of live parameters of the HS-5 human bone marrow stromal cell line cocultured with the leukemia cells in hypoxia, for the studies of leukemia-stroma cross-talk. Cytometry A 2018; 93:929-940. [PMID: 30247803 DOI: 10.1002/cyto.a.23580] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/18/2018] [Accepted: 07/20/2018] [Indexed: 12/15/2022]
Abstract
The unique bone marrow microenvironment is created by stromal cells and such physical conditions as hypoxia. Both hypoxia and interactions with stromal cells have a significant impact on the biology of leukemia cells, changing their sensitivity to antileukemic therapies. Thus, it is crucial to introduce biological systems, which enable the investigation of leukemia-stroma cross-talk and verification of novel therapies effectiveness under such bone marrow niche-mimicking conditions. Here, we have established an experimental setup based on the hypoxic co-culture of stromal cells with different cell lines derived from various leukemia patients. Flow cytometry enables simultaneous fluorescent tracking of viable cells and analysis of fundamental cellular processes, also to monitor the basal vital state of cells in the hypoxic co-culture. This is critically important, as the stromal cells deliver a big variability of signals to protect leukemia cells and provide drug resistance. Therefore, keeping stromal cells at the healthy state is crucial during experimental procedures. In the proposed studies, viability, apoptosis, proliferation, ROS production, and mitochondrial membrane potential were monitored in both cell types, which were separated on the basis of the fluorescence of a cell tracker. We have shown that the proposed hypoxic co-culture conditions do not affect basal live parameters of stromal cells, indicating the relevance of proposed model. Finally, we utilized this experimental setup to monitor the stroma-mediated protection of leukemia cells from the imatinib-induced cell death, which contributes to the leukemia progression and development of therapy resistance. Altogether, we recommend such flow cytometric strategy as an elementary screen of the vital state of stromal cells, which should be performed when using the co-culture hypoxic models. The proposed approach can also be broadly used for other studies of the leukemia-stroma cross-talk and of the part played by the leukemic microenvironment in drug screening studies.
Collapse
Affiliation(s)
| | - Agata Kominek
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Magdalena Wolczyk
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Marta D Kolba
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Julian Swatler
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
26
|
Varshosaz J, Davoudi MA, Rasoul-Amini S. Docetaxel-loaded nanostructured lipid carriers functionalized with trastuzumab (Herceptin) for HER2-positive breast cancer cells. J Liposome Res 2017; 28:285-295. [DOI: 10.1080/08982104.2017.1370471] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Jaleh Varshosaz
- Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Ali Davoudi
- Department of Medical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Rasoul-Amini
- Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
27
|
Altea-Manzano P, Unciti-Broceta JD, Cano-Cortes V, Ruiz-Blas MP, Valero-Griñan T, Diaz-Mochon JJ, Sanchez-Martin R. Tracking cell proliferation using a nanotechnology-based approach. Nanomedicine (Lond) 2017; 12:1591-1605. [PMID: 28513331 DOI: 10.2217/nnm-2017-0118] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
AIM To develop an efficient nanotechnology fluorescence-based method to track cell proliferation to avoid the limitations of current cell-labeling dyes. MATERIAL & METHODS Synthesis, PEGylation, bifunctionalization and labeling with a fluorophore (Cy5) of 200 nm polystyrene nanoparticles (NPs) were performed. These NPs were characterized and assessed for in vitro long-term monitoring of cell proliferation. RESULTS The optimization and validation of this method to track long-term cell proliferation assays have been achieved with high reproducibility, without cell cycle disruption. This method has been successfully applied in several adherent and suspension cells including hard-to-transfect cells and isolated human primary lymphocytes. CONCLUSION A novel approach to track efficiently cellular proliferation by flow cytometry using fluorescence labeled NPs has been successfully developed. [Formula: see text].
Collapse
Affiliation(s)
- Patricia Altea-Manzano
- GENYO:Pfizer - Universidad de Granada-Junta de Andalucía Centre for Genomics & Oncological Research, Health Science Technological Park (PTS), Avenida de la Ilustración 114, 18016 Granada, Spain.,R&D Deparment, NanoGetic S. L. Granada HealthScienceTechnological Park (PTS), Avenida de la Innovación 1, Edificio BIC, 18016 Granada, Spain
| | - Juan Diego Unciti-Broceta
- R&D Deparment, NanoGetic S. L. Granada HealthScienceTechnological Park (PTS), Avenida de la Innovación 1, Edificio BIC, 18016 Granada, Spain
| | - Victoria Cano-Cortes
- GENYO:Pfizer - Universidad de Granada-Junta de Andalucía Centre for Genomics & Oncological Research, Health Science Technological Park (PTS), Avenida de la Ilustración 114, 18016 Granada, Spain.,Department of Medicinal & Organic Chemistry, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - María Paz Ruiz-Blas
- GENYO:Pfizer - Universidad de Granada-Junta de Andalucía Centre for Genomics & Oncological Research, Health Science Technological Park (PTS), Avenida de la Ilustración 114, 18016 Granada, Spain.,Department of Medicinal & Organic Chemistry, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Teresa Valero-Griñan
- GENYO:Pfizer - Universidad de Granada-Junta de Andalucía Centre for Genomics & Oncological Research, Health Science Technological Park (PTS), Avenida de la Ilustración 114, 18016 Granada, Spain.,Department of Medicinal & Organic Chemistry, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Juan Jose Diaz-Mochon
- GENYO:Pfizer - Universidad de Granada-Junta de Andalucía Centre for Genomics & Oncological Research, Health Science Technological Park (PTS), Avenida de la Ilustración 114, 18016 Granada, Spain.,Department of Medicinal & Organic Chemistry, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| | - Rosario Sanchez-Martin
- GENYO:Pfizer - Universidad de Granada-Junta de Andalucía Centre for Genomics & Oncological Research, Health Science Technological Park (PTS), Avenida de la Ilustración 114, 18016 Granada, Spain.,Department of Medicinal & Organic Chemistry, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
| |
Collapse
|