1
|
Zeng X, Luo D, Zhang S, Cui Z, Wang Y, Chen J, Zhang S, Teng L, Hu Z, Liu L, Zhou S, Zeng Z, Long J. High-dose radiation-induced immunogenic cell death of bladder cancer cells leads to dendritic cell activation. PLoS One 2024; 19:e0307024. [PMID: 39231199 PMCID: PMC11373825 DOI: 10.1371/journal.pone.0307024] [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: 01/31/2024] [Accepted: 06/27/2024] [Indexed: 09/06/2024] Open
Abstract
Radiotherapy is a commonly used method in the treatment of bladder cancers (BC). Radiation-induced immunogenic cell death (ICD) is related to the immune response against cancers and their prognoses. Even though dendritic cells (DC) act as powerful antigen-presenting cells in the body, their precise role in this ICD process remains unclear. Accordingly, an in vitro study was undertaken to ascertain whether high-dose radiation-induced ICD of BC cells could regulate the immune response of DC. The results indicated that high-dose radiation treatments of BC cells significantly increased their levels of apoptosis, blocked their cell cycle in the G2/M phase, increased their expression of ICD-related proteins, and upregulated their secretion of CCL5 and CCL21 which control the directed migration of DC. It was also noted that expression of CD80, CD86, CCR5, and CCR7 on DC was upregulated in the medium containing the irradiated cells. In conclusion, the present findings illustrate that high-dose radiation can induce the occurrence of ICD within BC cells, concomitantly resulting in the activation of DC. Such findings could be of great significance in increasing the understanding how radiotherapy of BC may work to bring about reductions in cell activity and how these processes in turn lead to immunoregulation of the function of DC.
Collapse
Affiliation(s)
- Xianlin Zeng
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Daiqin Luo
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Shuai Zhang
- Department of Interventional Radiology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhonghui Cui
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Yun Wang
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Jin Chen
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Shichao Zhang
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Lijing Teng
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Zuquan Hu
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Lina Liu
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Shi Zhou
- Department of Interventional Radiology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhu Zeng
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
| | - Jinhua Long
- Department of Head and Neck, Affiliated Tumor Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
2
|
Barbonari S, D'Amore A, Hanbashi AA, Palombi F, Riccioli A, Parrington J, Filippini A. Endolysosomal two-pore channel 2 plays opposing roles in primary and metastatic malignant melanoma cells. Cell Biol Int 2024; 48:521-540. [PMID: 38263578 DOI: 10.1002/cbin.12129] [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: 07/22/2023] [Revised: 11/10/2023] [Accepted: 01/01/2024] [Indexed: 01/25/2024]
Abstract
The ion channel two-pore channel 2 (TPC2), localised on the membranes of acidic organelles such as endo-lysosomes and melanosomes, has been shown to play a role in pathologies including cancer, and it is differently expressed in primary versus metastatic melanoma cells. Whether TPC2 plays a pro- or anti-oncogenic role in different tumour conditions is a relevant open question which we have explored in melanoma at different stages of tumour progression. The behaviour of primary melanoma cell line B16F0 and its metastatic subline B16F10 were compared in response to TPC2 modulation by silencing (by small interfering RNA), knock-out (by CRISPR/Cas9) and overexpression (by mCherry-TPC2 transfected plasmid). TPC2 silencing increased cell migration, epithelial-to-mesenchymal transition and autophagy in the metastatic samples, but abated them in the silenced primary ones. Interestingly, while TPC2 inactivation failed to affect markers of proliferation in both samples, it strongly enhanced the migratory behaviour of the metastatic cells, again suggesting that in the more aggressive phenotype TPC2 plays a specific antimetastatic role. In line with this, overexpression of TPC2 in B16F10 cells resulted in phenotype rescue, that is, a decrease in migratory ability, thus collectively resuming traits of the B16F0 primary cell line. Our research shows a novel role of TPC2 in melanoma cells that is intriguingly different in initial versus late stages of cancer progression.
Collapse
Affiliation(s)
- Samantha Barbonari
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Unit of Histology and Medical Embryology, Sapienza University, Rome, Italy
| | | | - Ali A Hanbashi
- Department of Pharmacology, University of Oxford, Oxford, UK
- Department of Pharmacology, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Fioretta Palombi
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Unit of Histology and Medical Embryology, Sapienza University, Rome, Italy
| | - Anna Riccioli
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Unit of Histology and Medical Embryology, Sapienza University, Rome, Italy
| | - John Parrington
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Antonio Filippini
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Unit of Histology and Medical Embryology, Sapienza University, Rome, Italy
| |
Collapse
|
3
|
Sun X, Spellman RA, Engel M, Rubitski E, Schuler M. Comparative analysis of micronucleus induction and DNA damage biomarkers in TK6 and A375 cells using flow cytometry. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2024; 65:25-46. [PMID: 38333939 DOI: 10.1002/em.22585] [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: 08/25/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 02/10/2024]
Abstract
Previously, we introduced an alternative adherent A375 cell line for clastogenicity and aneugenicity testing using a high content imaging platform. To further characterize the performance of A375 cells, we investigated the sensitivity and specificity of A375 and TK6 cells by directly comparing micronucleus (MN) induction, cytotoxicity (relative cell counts, viability, and apoptosis), clastogenicity (γH2AX), and aneuploidy markers (pH 3, MPM-2, and polyploidy) using flow cytometric methods. We evaluated 14 compounds across different mechanisms (non-genotoxic apoptosis inducers, clastogens, and aneugens with either tubulin binding or aurora kinase inhibiting phenotypes) at 4-h and 24-h post treatment. Both aneugens and clastogens tested positive for micronucleus induction in both cell lines. Apoptosis continued to be a confounding factor for flow cytometry-based micronuclei assessment in TK6 cells as evidenced by positive responses by the three cytotoxicants. Conversely, A375 cells were not affected by apoptosis-related false positive signals and did not produce a positive response in the in vitro micronucleus assay. Benchmark dose response (BMD) analysis showed that the induction of micronuclei and biomarkers occurred at similar concentrations in both cell lines for clastogens and aneugens. By showing that A375 cells have similar sensitivity to TK6 cells but a greater specificity, these results provide additional support for A375 cells to be used as an alternative adherent cell line for in vitro genetic toxicology assessment.
Collapse
Affiliation(s)
- Xiaowen Sun
- Pfizer Research and Development, Groton, Connecticut, USA
| | | | - Maria Engel
- Pfizer Research and Development, Groton, Connecticut, USA
| | | | - Maik Schuler
- Pfizer Research and Development, Groton, Connecticut, USA
| |
Collapse
|
4
|
Fitzgerald MC, O'Halloran PJ, Kerrane SA, Ní Chonghaile T, Connolly NMC, Murphy BM. The identification of BCL-XL and MCL-1 as key anti-apoptotic proteins in medulloblastoma that mediate distinct roles in chemotherapy resistance. Cell Death Dis 2023; 14:705. [PMID: 37898609 PMCID: PMC10613306 DOI: 10.1038/s41419-023-06231-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 09/25/2023] [Accepted: 10/18/2023] [Indexed: 10/30/2023]
Abstract
Medulloblastoma is the most common malignant paediatric brain tumour, representing 20% of all paediatric intercranial tumours. Current aggressive treatment protocols and the use of radiation therapy in particular are associated with high levels of toxicity and significant adverse effects, and long-term sequelae can be severe. Therefore, improving chemotherapy efficacy could reduce the current reliance on radiation therapy. Here, we demonstrated that systems-level analysis of basal apoptosis protein expression and their signalling interactions can differentiate between medulloblastoma cell lines that undergo apoptosis in response to chemotherapy, and those that do not. Combining computational predictions with experimental BH3 profiling, we identified a therapeutically-exploitable dependence of medulloblastoma cells on BCL-XL, and experimentally validated that BCL-XL targeting, and not targeting of BCL-2 or MCL-1, can potentiate cisplatin-induced cytotoxicity in medulloblastoma cell lines with low sensitivity to cisplatin treatment. Finally, we identified MCL-1 as an anti-apoptotic mediator whose targeting is required for BCL-XL inhibitor-induced apoptosis. Collectively, our study identifies that BCL-XL and MCL-1 are the key anti-apoptotic proteins in medulloblastoma, which mediate distinct protective roles. While BCL-XL has a first-line role in protecting cells from apoptosis basally, MCL-1 represents a second line of defence that compensates for BCL-XL upon its inhibition. We provide rationale for the further evaluation of BCL-XL and MCL-1 inhibitors in the treatment of medulloblastoma, and together with current efforts to improve the cancer-specificity of BCL-2 family inhibitors, these novel treatment strategies have the potential to improve the future clinical management of medulloblastoma.
Collapse
Affiliation(s)
- Marie-Claire Fitzgerald
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 31A York Street, Dublin, D02 YN77, Ireland
- National Children's Research Centre at the Children's Health Ireland at Crumlin, Dublin, D12 N512, Ireland
| | - Philip J O'Halloran
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 31A York Street, Dublin, D02 YN77, Ireland
- Department of Neurosurgery, Queen Elizabeth Hospital, Birmingham, UK
| | - Sean A Kerrane
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 31A York Street, Dublin, D02 YN77, Ireland
- National Children's Research Centre at the Children's Health Ireland at Crumlin, Dublin, D12 N512, Ireland
| | - Triona Ní Chonghaile
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 31A York Street, Dublin, D02 YN77, Ireland
| | - Niamh M C Connolly
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 31A York Street, Dublin, D02 YN77, Ireland
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, 31A York Street, Dublin, D02 YN77, Ireland
| | - Brona M Murphy
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 31A York Street, Dublin, D02 YN77, Ireland.
- National Children's Research Centre at the Children's Health Ireland at Crumlin, Dublin, D12 N512, Ireland.
| |
Collapse
|
5
|
Lie KCM, Bonturi CR, Salu BR, de Oliveira JR, Bonini Galo M, Paiva PMG, Correia MTDS, Oliva MLV. Impairment of SK-MEL-28 Development-A Human Melanoma Cell Line-By the Crataeva tapia Bark Lectin and Its Sequence-Derived Peptides. Int J Mol Sci 2023; 24:10617. [PMID: 37445794 DOI: 10.3390/ijms241310617] [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: 04/19/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Melanoma is difficult to treat with chemotherapy, prompting the need for new treatments. Protease inhibitors have emerged as promising candidates as tumor cell proteases promote metastasis. Researchers have developed a chimeric form of the Bauhinia bauhinioides kallikrein inhibitor, rBbKIm, which has shown negative effects on prostate tumor cell lines DU145 and PC3. Crataeva tapia bark lectin, CrataBL, targets sulfated oligosaccharides in glycosylated proteins and has also demonstrated deleterious effects on prostate and glioblastoma tumor cells. However, neither rBbKIm nor its derived peptides affected the viability of SK-MEL-28, a melanoma cell line, while CrataBL decreased viability by over 60%. Two peptides, Pep. 26 (Ac-Q-N-S-S-L-K-V-V-P-L-NH2) and Pep. 27 (Ac-L-P-V-V-K-L-S-S-N-Q-NH2), were also tested. Pep. 27 suppressed cell migration and induced apoptosis when combined with vemurafenib, while Pep. 26 inhibited cell migration and reduced nitric oxide and the number of viable cells. Vemurafenib, a chemotherapy drug used to treat melanoma, was found to decrease the release of interleukin 8 and PDGF-AB/BB cytokines and potentiated the effects of proteins and peptides in reducing these cytokines. These findings suggest that protease inhibitors may be effective in blocking melanoma cells and highlight the potential of CrataBL and its derived peptides.
Collapse
Affiliation(s)
| | - Camila Ramalho Bonturi
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo 04044-020, Brazil
| | - Bruno Ramos Salu
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo 04044-020, Brazil
| | | | - Márcia Bonini Galo
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo 04044-020, Brazil
| | | | | | | |
Collapse
|
6
|
Vera J, Lai X, Baur A, Erdmann M, Gupta S, Guttà C, Heinzerling L, Heppt MV, Kazmierczak PM, Kunz M, Lischer C, Pützer BM, Rehm M, Ostalecki C, Retzlaff J, Witt S, Wolkenhauer O, Berking C. Melanoma 2.0. Skin cancer as a paradigm for emerging diagnostic technologies, computational modelling and artificial intelligence. Brief Bioinform 2022; 23:6761961. [PMID: 36252807 DOI: 10.1093/bib/bbac433] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/28/2022] [Accepted: 09/08/2022] [Indexed: 12/19/2022] Open
Abstract
We live in an unprecedented time in oncology. We have accumulated samples and cases in cohorts larger and more complex than ever before. New technologies are available for quantifying solid or liquid samples at the molecular level. At the same time, we are now equipped with the computational power necessary to handle this enormous amount of quantitative data. Computational models are widely used helping us to substantiate and interpret data. Under the label of systems and precision medicine, we are putting all these developments together to improve and personalize the therapy of cancer. In this review, we use melanoma as a paradigm to present the successful application of these technologies but also to discuss possible future developments in patient care linked to them. Melanoma is a paradigmatic case for disruptive improvements in therapies, with a considerable number of metastatic melanoma patients benefiting from novel therapies. Nevertheless, a large proportion of patients does not respond to therapy or suffers from adverse events. Melanoma is an ideal case study to deploy advanced technologies not only due to the medical need but also to some intrinsic features of melanoma as a disease and the skin as an organ. From the perspective of data acquisition, the skin is the ideal organ due to its accessibility and suitability for many kinds of advanced imaging techniques. We put special emphasis on the necessity of computational strategies to integrate multiple sources of quantitative data describing the tumour at different scales and levels.
Collapse
Affiliation(s)
- Julio Vera
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Xin Lai
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Andreas Baur
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Michael Erdmann
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Shailendra Gupta
- Department of Systems Biology and Bioinformatics, Institute of Computer Science, University of Rostock, Rostock 18051, Germany
| | - Cristiano Guttà
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569 Stuttgart, Germany
| | - Lucie Heinzerling
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany.,Department of Dermatology, LMU University Hospital, Munich, Germany
| | - Markus V Heppt
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | | | - Manfred Kunz
- Department of Dermatology, Venereology and Allergology, University of Leipzig, 04103 Leipzig, Germany
| | - Christopher Lischer
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Brigitte M Pützer
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, 18057 Rostock, Germany
| | - Markus Rehm
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569 Stuttgart, Germany.,Stuttgart Research Center Systems Biology, University of Stuttgart, 70569 Stuttgart, Germany
| | - Christian Ostalecki
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | - Jimmy Retzlaff
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| | | | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, Institute of Computer Science, University of Rostock, Rostock 18051, Germany
| | - Carola Berking
- Department of Dermatology, FAU Erlangen-Nürnberg, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen and Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
| |
Collapse
|
7
|
Wang L, Zhu Z, Liang Q, Tao Y, Jin G, Zhong Y, Dai J, Dai R, Wang Z, Chen J, Zhou L, Ke S, Zheng B, Lan L, Lin X, Chen T. A novel small molecule glycolysis inhibitor WZ35 exerts anti-cancer effect via metabolic reprogramming. J Transl Med 2022; 20:530. [DOI: 10.1186/s12967-022-03758-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 11/05/2022] [Indexed: 11/19/2022] Open
Abstract
Abstract
Background
Liver cancer is the fifth leading cause of cancer death worldwide, but early diagnosis and treatment of liver cancer remains a clinical challenge. How to screen and diagnose liver cancer early and prolong the survival rate is still the focus of researchers.
Methods
Cell experiments were used to detect the effect of WZ35 on the colony formation ability and proliferation activity of hepatoma cells, nude mouse experiment to observe the in vivo anticancer activity and toxic side effects of WZ35; metabolomics analysis, glucose metabolism experiment and Seahorse analysis of liver cancer cells treated with WZ35; cell experiments combined with bioinformatics analysis to explore the mechanism of WZ35-mediated metabolic reprogramming to exert anticancer activity; tissue microarray and case analysis to evaluate the clinical significance of biomarkers for early diagnosis, treatment and prognosis evaluation of liver cancer.
Results
WZ35 inhibited the proliferation activity of various cell lines of liver cancer, and showed good therapeutic effect in nude mice model of hepatocellular carcinoma without obvious toxic and side effects; WZ35 inhibited the absorption of glucose in hepatoma cells, and the drug effect glycolysis, phosphorylation and purine metabolism are relatively seriously damaged; WZ35 mainly inhibits YAP from entering the nucleus as a transcription factor activator by activating oxidative stress in liver cancer cells, reducing the transcription of GLUT1, and finally reducing its GLUT1. Tissue microarray and case analysis showed that GLUT1 and YAP were highly expressed and correlated in liver cancer patients, and were associated with poor patient prognosis. The GLUT1-YAP risk model had a high score in predicting prognosis.
Conclusion
The study confirms that WZ35 is a small molecule glycolysis inhibitor, and through its properties, it mediates metabolic reprogramming dominated by impaired glycolysis, oxidative phosphorylation and purine metabolism to inhibit the proliferation activity of liver cancer cells. Our findings present novel insights into the pathology of liver cancer and potential targets for new therapeutic strategies. GLUT1-YAP has important reference significance for predicting the stages of disease progression in liver cancer patients and have the potential to serve as novel biomarkers for the diagnosis and treatment of liver cancer.
Collapse
|
8
|
Gcanga L, Tamgue O, Ozturk M, Pillay S, Jacobs R, Chia JE, Mbandi SK, Davids M, Dheda K, Schmeier S, Alam T, Roy S, Suzuki H, Brombacher F, Guler R. Host-Directed Targeting of LincRNA-MIR99AHG Suppresses Intracellular Growth of Mycobacterium tuberculosis. Nucleic Acid Ther 2022; 32:421-437. [PMID: 35895506 PMCID: PMC7613730 DOI: 10.1089/nat.2022.0009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) kills 1.6 million people worldwide every year, and there is an urgent need for targeting host-pathogen interactions as a strategy to reduce mycobacterial resistance to current antimicrobials. Noncoding RNAs are emerging as important regulators of numerous biological processes and avenues for exploitation in host-directed therapeutics. Although long noncoding RNAs (lncRNAs) are abundantly expressed in immune cells, their functional role in gene regulation and bacterial infections remains understudied. In this study, we identify an immunoregulatory long intergenic noncoding RNA, lincRNA-MIR99AHG, which is upregulated in mouse and human macrophages upon IL-4/IL-13 stimulation and downregulated after clinical Mtb HN878 strain infection and in peripheral blood mononuclear cells from active TB patients. To evaluate the functional role of lincRNA-MIR99AHG, we used antisense locked nucleic acid (LNA) GapmeR-mediated antisense oligonucleotide (ASO) lncRNA knockdown experiments. Knockdown of lincRNA-MIR99AHG with ASOs significantly reduced intracellular Mtb growth in mouse and human macrophages and reduced pro-inflammatory cytokine production. In addition, in vivo treatment of mice with MIR99AHG ASOs reduced the mycobacterial burden in the lung and spleen. Furthermore, in macrophages, lincRNA-MIR99AHG is translocated to the nucleus and interacts with high affinity to hnRNPA2/B1 following IL-4/IL-13 stimulation and Mtb HN878 infection. Together, these findings identify lincRNA-MIR99AHG as a positive regulator of inflammation and macrophage polarization to promote Mtb growth and a possible target for adjunctive host-directed therapy against TB.
Collapse
Affiliation(s)
- Lorna Gcanga
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Department of Pathology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa.,Immunology of Infectious Diseases, Faculty of Health Sciences, South African Medical Research Council (SAMRC) University of Cape Town, Cape Town, South Africa
| | - Ousman Tamgue
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Department of Pathology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa.,Immunology of Infectious Diseases, Faculty of Health Sciences, South African Medical Research Council (SAMRC) University of Cape Town, Cape Town, South Africa.,Department of Biochemistry, Faculty of Sciences, University of Douala, Douala, Cameroon
| | - Mumin Ozturk
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Department of Pathology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa.,Immunology of Infectious Diseases, Faculty of Health Sciences, South African Medical Research Council (SAMRC) University of Cape Town, Cape Town, South Africa
| | - Shandre Pillay
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Department of Pathology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa.,Immunology of Infectious Diseases, Faculty of Health Sciences, South African Medical Research Council (SAMRC) University of Cape Town, Cape Town, South Africa
| | - Raygaana Jacobs
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Department of Pathology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa.,Immunology of Infectious Diseases, Faculty of Health Sciences, South African Medical Research Council (SAMRC) University of Cape Town, Cape Town, South Africa
| | - Julius Ebua Chia
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Department of Pathology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa.,Immunology of Infectious Diseases, Faculty of Health Sciences, South African Medical Research Council (SAMRC) University of Cape Town, Cape Town, South Africa
| | - Stanley Kimbung Mbandi
- Division of Immunology, Department of Pathology, South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Malika Davids
- Division of Pulmonology, Department of Medicine, Centre for Lung Infection and Immunology, UCT Lung Institute, University of Cape Town, Cape Town, South Africa.,South African MRC/UCT Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa
| | - Keertan Dheda
- Division of Pulmonology, Department of Medicine, Centre for Lung Infection and Immunology, UCT Lung Institute, University of Cape Town, Cape Town, South Africa.,South African MRC/UCT Centre for the Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South Africa.,Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical medicine, London, United Kingdom
| | - Sebastian Schmeier
- College of Science, School of Natural and Computational Sciences, Massey University, Auckland, New Zealand
| | - Tanvir Alam
- Information and Computing Technology Division, College of Science and Engineering, Hamad Bin Khalifa University, Doha, Qatar
| | - Sugata Roy
- RIKEN Center for Integrative Medical Sciences, Cellular Function Conversion Technology Team, Yokohama, Japan
| | - Harukazu Suzuki
- RIKEN Center for Integrative Medical Sciences, Cellular Function Conversion Technology Team, Yokohama, Japan
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Department of Pathology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa.,Immunology of Infectious Diseases, Faculty of Health Sciences, South African Medical Research Council (SAMRC) University of Cape Town, Cape Town, South Africa.,Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Address correspondence to: Frank Brombacher, PhD, International Centre for Genetic Engineering and Biotechnology (ICGEB) Department of Pathology, Cape Town Component, Cape Town 7925, South Africa
| | - Reto Guler
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Department of Pathology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa.,Immunology of Infectious Diseases, Faculty of Health Sciences, South African Medical Research Council (SAMRC) University of Cape Town, Cape Town, South Africa.,Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Reto Guler, PhD, Division of Immunology, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town 7925, South Africa
| |
Collapse
|
9
|
Fu W, Ma G. Significance of immunogenic cell death-related genes in prognosis prediction and immune microenvironment landscape of patients with cutaneous melanoma. Front Genet 2022; 13:988821. [PMID: 36212143 PMCID: PMC9532744 DOI: 10.3389/fgene.2022.988821] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Cutaneous melanoma (CM) is one of the most life-threatening tumors. Although targeted therapies and immune checkpoint inhibitors have significantly improved patient outcomes over the past decades, they still have their efficacy limitations. Immunogenic cell death (ICD) induces regulated cell death through immunogenic signal secretion and exposure. Accumulated evidence suggests that the ICD process is an effective target for the treatment of a variety of tumor types, including CM. However, the research on ICD in CM is far from complete, and its clinical value has not been widely concerned. By analyzing the Cancer Genome Atlas (TCGA) database, we constructed a new risk model based on 4 ICD-related genes and validated its ability to predict the prognosis of CM patients. In addition, we comprehensively analyzed the tumor microenvironment (TME) of CM patients and showed a significant immunosuppressive TME in the high-risk group compared with the low-risk group. By Immunophenoscore (IPS), we further explored the correlation between the model and immunotherapy response. The data of Genomics of Drug Sensitivity in Cancer (GDSC) database were further extracted to analyze drug sensitivity and evaluate its correlation with the established risk model. In the end, differential expressed genes (DEGs) were analyzed by Gene Set Variation Analysis (GSVA) to preliminarily explore the possible signaling pathways related to the prognosis of ICD and CM. The results of this study provide new perspectives and insights for individualized and accurate treatment strategies for CM patients.
Collapse
|
10
|
Shao R, Wang H, Wang J, Lu S, He R, Lu Y. Comprehensive analysis of a pyroptosis-related gene signature of clinical and biological value in acute myeloid leukaemia. Int Immunopharmacol 2022; 108:108802. [DOI: 10.1016/j.intimp.2022.108802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/31/2022] [Accepted: 04/23/2022] [Indexed: 11/05/2022]
|
11
|
Li JJ, Wang CM, Wang YJ, Yang Q, Cai WY, Li YJ, Song M, Zang YL, Cui XH, Li Q, Chen Y, Weng XG, Zhu XX. Network pharmacology analysis and experimental validation to explore the mechanism of Shenlian extract on myocardial ischemia. JOURNAL OF ETHNOPHARMACOLOGY 2022; 288:114973. [PMID: 34990768 DOI: 10.1016/j.jep.2022.114973] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/30/2021] [Accepted: 01/02/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shenlian extract (SL), extracted from Salvia miltiorrhiza Bunge and Andrographis paniculata (Burm. f.) Nees, has been proved to be effective in the prevention and treatment of atherosclerosis. Recently, we have partially elucidated the mechanisms involved in the therapeutic effects of SL on myocardial ischemia (MI). However, the underlying mechanisms remain largely unclear. AIM OF THE STUDY This study aims to explore the potential molecular mechanism of SL on MI on the basis of network pharmacology. MATERIALS AND METHODS First, the main active ingredients of SL were screened in the Traditional Chinese Medicine Integrated Database, and the MI-associated targets were collected from the DisGeNET database. Then, we used compound-target and target-pathway networks to uncover the therapeutic mechanisms of SL. On the basis of network pharmacology analysis results, we assessed the effects of SL in MI rat model and oxygen glucose deprivation model of H9c2 cells and validated the possible molecular mechanisms of SL on myocardial injury in vivo and in vitro. RESULTS The network pharmacology results showed that 37 potential targets were recognized, including TNF-α, Bcl-2, STAT3, PI3K and MMP2. These results revealed that the possible targets of SL were involved in the regulation of inflammation and apoptosis signaling pathway. Then, in vivo experiments indicated that SL significantly reduced the myocardial infarction size of MI rats. Serum CK-MB, cTnT, CK, LDH, and AST levels were significantly decreased by SL (P < 0.05 or P < 0.01). In vitro, SL significantly increased H9c2 cell viability. The levels of inflammation factors including TNF-α and MMP2 were significantly decreased by SL (P < 0.05 or P < 0.01). TUNEL and Annexin V/propidium iodide assays indicated that SL could significantly decrease the cell apoptotic rate in vivo and in vitro (P < 0.05 or P < 0.01). The remarkable upregulation of anti-apoptotic Bcl-2 and downregulation of pro-apoptotic Bax protein level further confirmed this result. Kyoto Encyclopedia of Genes and Genomes pathway analysis showed that the PI3K-AKT and JAK2-STAT3 pathways were significantly enriched in SL. Compared with the model group, SL treatment significantly activated the PI3K-AKT and JAK2-STAT3 pathways in vivo and in vitro according to Western blot analyses. CONCLUSION SL could protect the myocardium from MI injury. The underlying mechanism may be related to the reduction of inflammation and apoptosis by activating the PI3K/AKT and JAK2/STAT3 pathways.
Collapse
Affiliation(s)
- Jing-Jing Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China
| | - Chun-Miao Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China
| | - Ya-Jie Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China.
| | - Qing Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China
| | - Wei-Yan Cai
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China
| | - Yu-Jie Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China
| | - Min Song
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China
| | - Yuan-Long Zang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China
| | - Xi-He Cui
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China
| | - Qi Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China
| | - Xiao-Gang Weng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China
| | - Xiao-Xin Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Science, Beijing, 100700, China.
| |
Collapse
|
12
|
Spontaneous activity of the mitochondrial apoptosis pathway drives chromosomal defects, the appearance of micronuclei and cancer metastasis through the Caspase-Activated DNAse. Cell Death Dis 2022; 13:315. [PMID: 35393399 PMCID: PMC8990075 DOI: 10.1038/s41419-022-04768-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 03/16/2022] [Accepted: 03/25/2022] [Indexed: 01/15/2023]
Abstract
Micronuclei are DNA-containing structures separate from the nucleus found in cancer cells. Micronuclei are recognized by the immune sensor axis cGAS/STING, driving cancer metastasis. The mitochondrial apoptosis apparatus can be experimentally triggered to a non-apoptotic level, and this can drive the appearance of micronuclei through the Caspase-activated DNAse (CAD). We tested whether spontaneously appearing micronuclei in cancer cells are linked to sub-lethal apoptotic signals. Inhibition of mitochondrial apoptosis or of CAD reduced the number of micronuclei in tumor cell lines as well as the number of chromosomal misalignments in tumor cells and intestinal organoids. Blockade of mitochondrial apoptosis or deletion of CAD reduced, while experimental activation CAD, STING-dependently, enhanced aggressive growth of tumor cells in vitro. Deletion of CAD from human cancer cells reduced metastasis in xenograft models. CAD-deficient cells displayed a substantially altered gene-expression profile, and a CAD-associated gene expression ‘signature’ strongly predicted survival in cancer patients. Thus, low-level activity in the mitochondrial apoptosis apparatus operates through CAD-dependent gene-induction and STING-activation and has substantial impact on metastasis in cancer.
Collapse
|
13
|
Haak VM, Huang S, Panigrahy D. Debris-stimulated tumor growth: a Pandora's box? Cancer Metastasis Rev 2021; 40:791-801. [PMID: 34665387 PMCID: PMC8524220 DOI: 10.1007/s10555-021-09998-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/01/2021] [Indexed: 12/24/2022]
Abstract
Current cancer therapies aim at eradicating cancer cells from the body. However, killing cells generates cell “debris” which can promote tumor progression. Thus, therapy can be a double-edged sword. Specifically, injury and debris generated by cancer therapies, including chemotherapy, radiation, and surgery, may offset their benefit by promoting the secretion of pro-tumorigenic factors (e.g., eicosanoid-driven cytokines) that stimulate regrowth and metastasis of surviving cells. The debris produced by cytotoxic cancer therapy can also contribute to a tumor microenvironment that promotes tumor progression and recurrence. Although not well understood, several molecular mechanisms have been implicated in debris-stimulated tumor growth that we review here, such as the involvement of extracellular vesicles, exosomal miR-194-5p, Bax, Bak, Smac, HMGB1, cytokines, and caspase-3. We discuss the cases of pancreatic and other cancer types where debris promotes postoperative tumor recurrence and metastasis, thus offering a new opportunity to prevent cancer progression intrinsically linked to treatment by stimulating resolution of tumor-promoting debris.
Collapse
Affiliation(s)
- Victoria M Haak
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| | - Sui Huang
- Institute for Systems Biology, Seattle, WA, USA
| | - Dipak Panigrahy
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
14
|
Czyżowska A, Barbasz A, Rudolphi-Szydło E, Dyba B. The cell membrane as the barrier in the defense against nanoxenobiotics: Zinc oxide nanoparticles interactions with native and model membrane of melanoma cells. J Appl Toxicol 2021; 42:334-341. [PMID: 34235764 DOI: 10.1002/jat.4216] [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: 04/09/2021] [Revised: 06/01/2021] [Accepted: 06/17/2021] [Indexed: 11/06/2022]
Abstract
Currently, we are dealing with ever-increasing pollution of the environment with metal and metal oxide nanoparticles. One type of these, zinc oxide nanoparticles (ZnO-NPs), are increasingly used in areas such as cosmetology, electrical engineering, medicine, and even in the food and textile industries. As a consequence, ZnO-NPs may enter the human body in many ways. Their influence on the body is still not clear. Here, we define the mechanism of the initial toxicity of ZnO-NPs to cells based on interaction with the lipid part of the native and model cell membrane. The selected cell lines react differently to contact with nanoparticles. We found a disruption of the native membranes of B16-F0 cells and to a lesser extent of COLO 679. In turn, the membrane of COLO 679 cells was more peroxidated, and cell viability was much lower. A model of the lipid part of the membrane was created for B16-F0 cells and compared with previously published studies on immune cells. On the basis of physicochemical parameters obtained for individual lipids and a mix representing the native membrane of the tested cells, we concluded that exposure to nanoparticles resulted in a change within the model membranes (specifically with the polar parts of lipids). The greatest interaction has been noticed between ZnO-NPs and zwitterionic phospholipids (PC and PE), cholesterol, and negatively charged phosphatidylglycerol. Assessing the interactions between the membrane and nanoparticles will help to better understand the first steps of its toxicity mechanism.
Collapse
Affiliation(s)
| | - Anna Barbasz
- Institute of Biology, Pedagogical University of Cracow, Cracow, Poland
| | | | - Barbara Dyba
- Institute of Biology, Pedagogical University of Cracow, Cracow, Poland
| |
Collapse
|
15
|
Espinosa M, Lizárraga F, Vázquez-Santillán K, Hidalgo-Miranda A, Piña-Sánchez P, Torres J, García-Ramírez RA, Maldonado V, Melendez-Zajgla J, Ceballos-Cancino G. Coexpression of Smac/DIABLO and Estrogen Receptor in breast cancer. Cancer Biomark 2021; 30:429-446. [PMID: 33492282 DOI: 10.3233/cbm-200535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Smac/DIABLO is a proapoptotic protein deregulated in breast cancer, with a controversial role as a tumor marker, possibly due to a lack of correlative mRNA and protein analyses. OBJECTIVE To investigate the association of Smac/DIABLO gene and protein levels with clinical variables in breast cancer patients. METHODS Smac/DIABLO mRNA expression was analyzed by qPCR in 57 frozen tissues, whereas protein levels were assessed by immunohistochemistry in 82 paraffin-embedded tissues. Survivin mRNA levels were also measured. In vitro assays were performed to investigate possible regulators of Smac/DIABLO. RESULTS Higher levels of Smac/DIABLO mRNA and protein were found in estrogen receptor (ER)-positive samples (p= 0.0054 and p= 0.0043, respectively) in comparison to ER-negative tumors. A negligible positive association was found between Smac/DIABLO and survivin expression. In vitro assays showed that Smac/DIABLO is not regulated by ER and, conversely, it does not participate in ER expression modulation. CONCLUSIONS mRNA and protein levels of Smac/DIABLO were increased in ER-positive breast tumors in comparison with ER-negative samples, although the mechanism of this regulation is still unknown. Public databases showed a possible clinical relevance for this association.
Collapse
Affiliation(s)
- Magali Espinosa
- Instituto Nacional de Medicina Genómica, Department of Basic Research, Functional Cancer Genomics Laboratory, Mexico City, Mexico
| | - Floria Lizárraga
- Instituto Nacional de Medicina Genómica, Department of Basic Research, Epigenetic Laboratory, Mexico City, Mexico
| | - Karla Vázquez-Santillán
- Instituto Nacional de Medicina Genómica, Department of Basic Research, Epigenetic Laboratory, Mexico City, Mexico
| | - Alfredo Hidalgo-Miranda
- Instituto Nacional de Medicina Genómica, Department of Basic Research, Cancer Genomics Laboratory, Mexico City, Mexico
| | - Patricia Piña-Sánchez
- Instituto Mexicano del Seguro Social, CMN S XXI, Oncology Research Unit, Molecular Oncology Laboratory, Mexico City, Mexico
| | - Javier Torres
- Instituto Mexicano del Seguro Social, CMN S XXI, Unity of Research in Infectious Diseases, Mexico City, Mexico
| | - Román A García-Ramírez
- Instituto Nacional de Medicina Genómica, Department of Basic Research, Functional Cancer Genomics Laboratory, Mexico City, Mexico
| | - Vilma Maldonado
- Instituto Nacional de Medicina Genómica, Department of Basic Research, Epigenetic Laboratory, Mexico City, Mexico
| | - Jorge Melendez-Zajgla
- Instituto Nacional de Medicina Genómica, Department of Basic Research, Functional Cancer Genomics Laboratory, Mexico City, Mexico
| | - Gisela Ceballos-Cancino
- Instituto Nacional de Medicina Genómica, Department of Basic Research, Functional Cancer Genomics Laboratory, Mexico City, Mexico
| |
Collapse
|
16
|
Trisciuoglio D, Del Bufalo D. New insights into the roles of antiapoptotic members of the Bcl-2 family in melanoma progression and therapy. Drug Discov Today 2021; 26:1126-1135. [PMID: 33545382 DOI: 10.1016/j.drudis.2021.01.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/25/2020] [Accepted: 01/26/2021] [Indexed: 02/07/2023]
Abstract
Prosurvival and antiapoptotic B cell lymphoma-2 (Bcl-2) family proteins are often overexpressed in cutaneous melanoma, one of the most aggressive types of human cancer. They are also implicated in resistance to therapy and participate in melanoma progression by regulating various processes, including cell proliferation, migration, invasion, and crosstalk with the tumor microenvironment. In this review, we summarize recent findings related to prosurvival members of the Bcl-2 family beyond their canonical functions in the apoptotic pathway, mainly focusing on their potential roles as diagnostic and prognostic biomarkers in cutaneous melanoma. We also provide an overview of different approaches used to inhibit Bcl-2 proteins in preclinical and clinical studies, which are mainly based on the inhibition of protein expression or the disruption of their antiapoptotic functions.
Collapse
Affiliation(s)
- Daniela Trisciuoglio
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome, Italy; Institute of Molecular Biology and Pathology, National Research Council, via degli Apuli 4, 00185, Rome, Italy.
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, Rome, Italy.
| |
Collapse
|
17
|
Yin Q, Zhao B, Zhu J, Fei Y, Shen W, Liang B, Zhu X, Li Y. JLX001 improves myocardial ischemia-reperfusion injury by activating Jak2-Stat3 pathway. Life Sci 2020; 257:118083. [PMID: 32673665 DOI: 10.1016/j.lfs.2020.118083] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/22/2020] [Accepted: 07/07/2020] [Indexed: 12/13/2022]
Abstract
AIMS To investigate the preclinical pharmacodynamics and mechanism of JLX001 against myocardial ischemia reperfusion (MI/R) for clinical application. MATERIALS AND METHODS In vivo, SD rats were given intragastric administration for 5 days, and the MI/R model was established by ligating/releasing the left anterior descending coronary artery. In vitro, the oxygen-glucose deprivation/reperfusion (OGD/R) model was established after the drug was pre-incubated for 24 h in H9C2 cells. The infract size was determined by TTC staining. Left ventricular function of MI/R rats was detected by echocardiography. The level of histopathological score was determined by hematoxylin-eosin (HE) staining. The level of superoxide dismutase (SOD), malondialdehyde (MDA), creatine kinase (CK), lactic dehydrogenase (LDH), tumor necrosis factor-α (TNF-α), and interleukin-1β (IL-1β) were determined by relevant kits. The level of apoptosis was measured by Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and Hoechst staining. The expression of p-Jak2, p-Stat3, Bax, Bcl-2, TNF-α, IL-1β protein were determined by western blot. KEY FINDINGS JLX001 can significantly improve left ventricular function, reduce myocardial infract size, histopathological score, the level of MDA, CK, LDH, TNF-α, IL-1β and the expression of Bax protein, significantly increase the activity of SOD, Bcl-2 protein expression, p-Jak2 protein expression, p-Stat3 protein expression in rat heart tissues and H9C2 cells. These effects can be reversed by AG490 which is a specific inhibitor of Jak2-Stat3 pathway. SIGNIFICANCE JLX001 can alleviate MI/R injury by inhibiting myocardial apoptosis, inflammation, and oxidative stress via Jak2-Stat3 pathway in vivo and in vitro.
Collapse
Affiliation(s)
- Qiyang Yin
- State key laboratory of Nature Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Bo Zhao
- State key laboratory of Nature Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jianping Zhu
- State key laboratory of Nature Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yuxiang Fei
- State key laboratory of Nature Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Weiyang Shen
- School of Sciences, China Pharmaceutical University, Nanjing 210009, PR China
| | - Bingwen Liang
- Jiangsu Jinglixin Pharmaceutical Technology Company Limited, Nanjing 211100, PR china
| | - Xiong Zhu
- School of Sciences, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Yuman Li
- State key laboratory of Nature Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
18
|
Bajor M, Graczyk-Jarzynka A, Marhelava K, Kurkowiak M, Rahman A, Aura C, Russell N, Zych AO, Firczuk M, Winiarska M, Gallagher WM, Zagozdzon R. Triple Combination of Ascorbate, Menadione and the Inhibition of Peroxiredoxin-1 Produces Synergistic Cytotoxic Effects in Triple-Negative Breast Cancer Cells. Antioxidants (Basel) 2020; 9:antiox9040320. [PMID: 32316111 PMCID: PMC7222372 DOI: 10.3390/antiox9040320] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/10/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive form of mammary malignancy currently without satisfactory systemic treatment options. Agents generating reactive oxygen species (ROS), such as ascorbate (Asc) and menadione (Men), especially applied in combination, have been proposed as an alternative anticancer modality. However, their effectiveness can be hampered by the cytoprotective effects of elevated antioxidant enzymes (e.g., peroxiredoxins, PRDX) in cancer. In this study, PRDX1 mRNA and protein expression were assessed in TNBC tissues by analysis of the online RNA-seq datasets and immunohistochemical staining of tissue microarray, respectively. We demonstrated that PRDX1 mRNA expression was markedly elevated in primary TNBC tumors as compared to non-malignant controls, with PRDX1 protein staining intensity correlating with favorable survival parameters. Subsequently, PRDX1 functionality in TNBC cell lines or non-malignant mammary cells was targeted by genetic silencing or chemically by auranofin (AUR). The PRDX1-knockdown or AUR treatment resulted in inhibition of the growth of TNBC cells in vitro. These cytotoxic effects were further synergistically potentiated by the incubation with a combination of the prooxidant agents, Asc and Men. In conclusion, we report that the PRDX1-related antioxidant system is essential for maintaining redox homeostasis in TNBC cells and can be an attractive therapeutic target in combination with ROS-generating agents.
Collapse
Affiliation(s)
- Malgorzata Bajor
- Department of Clinical Immunology, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland; (M.B.); (K.M.)
| | - Agnieszka Graczyk-Jarzynka
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5, 02-097 Warsaw, Poland; (A.G.-J.); (A.O.Z.); (M.F.); (M.W.)
| | - Katsiaryna Marhelava
- Department of Clinical Immunology, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland; (M.B.); (K.M.)
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Trojdena 2a, 02-091 Warsaw, Poland
| | - Malgorzata Kurkowiak
- International Centre for Cancer Vaccine Science, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland;
| | - Arman Rahman
- Cancer Biology and Therapeutics Laboratory, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, D04 Dublin 4, Ireland; (A.R.); (C.A.); (N.R.); (W.M.G.)
| | - Claudia Aura
- Cancer Biology and Therapeutics Laboratory, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, D04 Dublin 4, Ireland; (A.R.); (C.A.); (N.R.); (W.M.G.)
| | - Niamh Russell
- Cancer Biology and Therapeutics Laboratory, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, D04 Dublin 4, Ireland; (A.R.); (C.A.); (N.R.); (W.M.G.)
| | - Agata O. Zych
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5, 02-097 Warsaw, Poland; (A.G.-J.); (A.O.Z.); (M.F.); (M.W.)
| | - Malgorzata Firczuk
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5, 02-097 Warsaw, Poland; (A.G.-J.); (A.O.Z.); (M.F.); (M.W.)
| | - Magdalena Winiarska
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5, 02-097 Warsaw, Poland; (A.G.-J.); (A.O.Z.); (M.F.); (M.W.)
| | - William M. Gallagher
- Cancer Biology and Therapeutics Laboratory, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, D04 Dublin 4, Ireland; (A.R.); (C.A.); (N.R.); (W.M.G.)
- OncoMark Ltd., Nova UCD, D04 Dublin 4, Ireland
| | - Radoslaw Zagozdzon
- Department of Clinical Immunology, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland; (M.B.); (K.M.)
- Department of Immunology, Transplantology, and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland
- Correspondence:
| |
Collapse
|