151
|
Zhai BT, Sun J, Shi YJ, Zhang XF, Zou JB, Cheng JX, Fan Y, Guo DY, Tian H. Review targeted drug delivery systems for norcantharidin in cancer therapy. J Nanobiotechnology 2022; 20:509. [DOI: 10.1186/s12951-022-01703-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/11/2022] [Indexed: 12/04/2022] Open
Abstract
AbstractNorcantharidin (NCTD) is a demethylated derivative of cantharidin (CTD), the main anticancer active ingredient isolated from traditional Chinese medicine Mylabris. NCTD has been approved by the State Food and Drug Administration for the treatment of various solid tumors, especially liver cancer. Although NCTD greatly reduces the toxicity of CTD, there is still a certain degree of urinary toxicity and organ toxicity, and the poor solubility, short half-life, fast metabolism, as well as high venous irritation and weak tumor targeting ability limit its widespread application in the clinic. To reduce its toxicity and improve its efficacy, design of targeted drug delivery systems based on biomaterials and nanomaterials is one of the most feasible strategies. Therefore, this review focused on the studies of targeted drug delivery systems combined with NCTD in recent years, including passive and active targeted drug delivery systems, and physicochemical targeted drug delivery systems for improving drug bioavailability and enhancing its efficacy, as well as increasing drug targeting ability and reducing its adverse effects.
Graphical Abstract
Collapse
|
152
|
Wang Y, Li S, Weng L, Du H, Wang J, Xu X. LASS2 overexpression enhances early apoptosis of lung cancer cells through the caspase‑dependent pathway. Oncol Rep 2022; 48:220. [PMID: 36300249 DOI: 10.3892/or.2022.8435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 06/27/2022] [Indexed: 11/06/2022] Open
Abstract
In a previous study by the authors, the longevity assurance homolog 2 (LASS2) gene was determined to inhibit activity of vacuolar H+‑ATPase (V‑ATPase) by combining with the C subunit (ATP6L) of V‑ATPase. However, the influence of LASS2 overexpression and silencing on apoptosis of human lung cancer cells 95D or 95C remains unclear. Thus, the effect of LASS2 on apoptosis and its potential mechanisms were investigated in 95D and 95C cells. Using the lentiviral transfection method, lentiviral vectors of LASS2 overexpression and silencing were transfected into 95D and 95C cells, respectively. The apoptotic ability of tumor cells was observed by flow cytometry. The expression levels of LASS2, Bcl‑2, Bax, cytochrome c, caspase‑9, and caspase‑3 were detected by western blotting. CCK‑8 assay was used to detect the growth ability of tumor cells in vitro. Flow cytometric analysis revealed that LASS2 overexpression could promote the early apoptosis of lung cancer cells 95D. CCK‑8 assay demonstrated that LASS2 overexpression inhibited the proliferation of 95D cells. Additionally, LASS2 overexpression decreased the expression of Bcl‑2, induced the release of cytochrome c from mitochondria, and promoted the activation of caspase‑9 and caspase‑3. There was a significant difference in the expression of Bcl‑2, cytochrome c, caspase‑9 and caspase‑3 in the LASS2‑overexpression group compared with the normal and negative control groups. Alternatively, the aforementioned experiments in lung cancer cells 95C following LASS2 silencing produced the opposite effects. LASS2 may induce early apoptosis of lung cancer cells by influencing the caspase‑dependent mitochondrial pathway.
Collapse
Affiliation(s)
- Yamei Wang
- Department of Pathology, College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010059, P.R. China
| | - Shirong Li
- Department of Pathology, College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010059, P.R. China
| | - Lixin Weng
- Department of Pathology, College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010059, P.R. China
| | - Hua Du
- Department of Pathology, College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010059, P.R. China
| | - Jingyuan Wang
- Department of Pathology, College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010059, P.R. China
| | - Xiaoyan Xu
- Department of Pathology, College of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region 010059, P.R. China
| |
Collapse
|
153
|
An J, Nagaki Y, Motoyama S, Kuze Y, Hoshizaki M, Kemuriyama K, Yamaguchi T, Ebihara T, Minamiya Y, Suzuki Y, Imai Y, Kuba K. Identification of Galectin-7 as a crucial metastatic enhancer of squamous cell carcinoma associated with immunosuppression. Oncogene 2022; 41:5319-5330. [PMID: 36335283 DOI: 10.1038/s41388-022-02525-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Metastasis predicts poor prognosis in cancer patients. It has been recognized that specific tumor microenvironment defines cancer cell metastasis, whereas the underlying mechanisms remain elusive. Here we show that Galectin-7 is a crucial mediator of metastasis associated with immunosuppression. In a syngeneic mouse squamous cell carcinoma (SCC) model of NR-S1M cells, we isolated metastasized NR-S1M cells from lymph nodes in tumor-bearing mice and established metastatic NR-S1M cells in in vitro culture. RNA-seq analysis revealed that interferon gene signature was markedly downregulated in metastatic NR-S1M cells compared with parental cells, and in vivo NR-S1M tumors heterogeneously developed focal immunosuppressive areas featured by deficiency of anti-tumor immune cells. Spatial transcriptome analysis (Visium) for the NR-S1M tumors revealed that various pro-metastatic genes were significantly upregulated in immunosuppressive areas when compared to immunocompetent areas. Notably, Galectin-7 was identified as a novel metastasis-driving factor. Galectin-7 expression was induced during tumorigenesis particularly in the microenvironment of immunosuppression, and extracellularly released at later stage of tumor progression. Deletion of Galectin-7 in NR-S1M cells significantly suppressed lymph node and lung metastasis without affecting primary tumor growth. Therefore, Galectin-7 is a crucial mediator of tumor metastasis of SCC, which is educated in the immune-suppressed tumor areas, and may be a potential target of cancer immunotherapy.
Collapse
Affiliation(s)
- Jianbo An
- Department of Biochemistry and Metabolic Science, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Yushi Nagaki
- Department of Biochemistry and Metabolic Science, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan.,Department of Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Satoru Motoyama
- Department of Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Yuta Kuze
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, 277-8562, Japan
| | - Midori Hoshizaki
- Department of Biochemistry and Metabolic Science, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan.,Department of Medical Biology, Akita University Graduate School of Medicine, Akita, Japan
| | - Kohei Kemuriyama
- Department of Biochemistry and Metabolic Science, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan.,Department of Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Tomokazu Yamaguchi
- Department of Biochemistry and Metabolic Science, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Takashi Ebihara
- Department of Medical Biology, Akita University Graduate School of Medicine, Akita, Japan
| | - Yoshihiro Minamiya
- Department of Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Yutaka Suzuki
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, 277-8562, Japan
| | - Yumiko Imai
- Laboratory of Regulation of Intractable Infectious Diseases, National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, 567-0085, Japan
| | - Keiji Kuba
- Department of Biochemistry and Metabolic Science, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan. .,Department of Pharmacology, Kyushu University Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan.
| |
Collapse
|
154
|
Sun Y, Yang J, Li Y, Luo J, Sun J, Li D, Wang Y, Wang K, Yang L, Wu L, Sun X. Single low-dose INC280-loaded theranostic nanoparticles achieve multirooted delivery for MET-targeted primary and liver metastatic NSCLC. Mol Cancer 2022; 21:212. [PMID: 36457016 PMCID: PMC9717478 DOI: 10.1186/s12943-022-01681-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/11/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) patients with primary tumors and liver metastases have substantially reduced survival. Since mesenchymal-epithelial transition factor (MET) plays a significant role in the molecular mechanisms of advanced NSCLC, small molecule MET inhibitor capmatinib (INC280) hold promise for clinically NSCLC treatment. However, the major obstacles of MET-targeted therapy are poor drug solubility and off-tumor effects, even oral high-dosing regimens cannot significantly increase the therapeutic drug concentration in primary and metastatic NSCLC. METHODS We developed a multirooted delivery system INC280-PFCE nanoparticles (NPs) by loading INC280 into perfluoro-15-crown-5-ether for improving MET-targeted therapy. Biodistribution and anti-MET/antimetastatic effects of NPs were validated in orthotopic NSCLC and NSCLC liver metastasis models in a single low-dose. The efficacy of INC280-PFCE NPs was also explored in human NSCLC specimens. RESULTS INC280-PFCE NPs exhibited excellent antitumor ability in vitro. In orthotopic NSCLC models, sustained release and prolonged retention behaviors of INC280-PFCE NPs within tumors could be visualized in real-time by 19F magnetic resonance imaging (19F-MRI), and single pulmonary administration of NPs showed more significant tumor growth inhibition than oral administration of free INC280 at a tenfold higher dose. Furthermore, a single low-dose INC280-PFCE NPs administered intravenously suppressed widespread dissemination of liver metastasis without systemic toxicity. Finally, we verified the clinical translation potential of INC280-PFCE NPs in human NSCLC specimens. CONCLUSIONS These results demonstrated high anti-MET/antimetastatic efficacies, real-time MRI visualization and high biocompatibility of NPs after a single low-dose.
Collapse
Affiliation(s)
- Yige Sun
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Jie Yang
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Yingbo Li
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Jing Luo
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Jiemei Sun
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Daoshuang Li
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Yuchen Wang
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Kai Wang
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Lili Yang
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Lina Wu
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| | - Xilin Sun
- grid.410736.70000 0001 2204 9268 Department of Nuclear Medicine, the Fourth Hospital of Harbin Medical University, Harbin, 150028 Heilongjiang China ,grid.410736.70000 0001 2204 9268NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, 150028 Heilongjiang China
| |
Collapse
|
155
|
Chen P, Li Y, Liu R, Xie Y, Jin Y, Wang M, Yu Z, Wang W, Luo X. Non-small cell lung cancer-derived exosomes promote proliferation, phagocytosis, and secretion of microglia via exosomal microRNA in the metastatic microenvironment. Transl Oncol 2022; 27:101594. [PMID: 36463825 PMCID: PMC9719005 DOI: 10.1016/j.tranon.2022.101594] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/29/2022] [Accepted: 11/23/2022] [Indexed: 12/04/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common tumor that metastasizes to the brain. It is now accepted that the successful colonization and growth of tumor cells are determined by the interaction between tumor cells and the tumor microenvironment (TME). Microglia, brain innate immune cells, have been reported to play a vital role in the establishment of brain metastases. As essential mediators of intercellular communications, tumor-derived exosomes have an important role in the pathogenesis and progression of cancer by transferring their cargos to specific recipient cells. The crosstalk between microglia and tumor-derived exosomes has been extensively described. However, it is still unclear whether metastatic NSCLC cells secret exosomes to microglia and regulate the microglial functions. Here, our results showed that microglia aggregated in the brain metastatic sites. Meanwhile, microglia could take up the exosomes derived from NSCLC cells, leading to alterations of microglial morphology and increased proliferation, phagocytosis, and release of inflammatory cytokines including interleukin-6, interleukin-8, and CXCL1. Further investigation indicated that miR1246 was the most enriched microRNA in NSCLC-derived exosomes and mediated the partial effects of exosomes on microglia. Notably, miR1246 was also upregulated in the plasmatic exosomes of NSCLC patients. These results offer a new insight into the impact of NSCLC-derived exosomes on microglia and provide a new potential biomarker for diagnosing NSCLC.
Collapse
Affiliation(s)
- Peng Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ying Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rui Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yi Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yu Jin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Minghuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhiyuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiang Luo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China,Correspondence author.
| |
Collapse
|
156
|
Nascentes Melo LM, Lesner NP, Sabatier M, Ubellacker JM, Tasdogan A. Emerging metabolomic tools to study cancer metastasis. Trends Cancer 2022; 8:988-1001. [PMID: 35909026 DOI: 10.1016/j.trecan.2022.07.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/24/2022] [Accepted: 07/06/2022] [Indexed: 12/24/2022]
Abstract
Metastasis is responsible for 90% of deaths in patients with cancer. Understanding the role of metabolism during metastasis has been limited by the development of robust and sensitive technologies that capture metabolic processes in metastasizing cancer cells. We discuss the current technologies available to study (i) metabolism in primary and metastatic cancer cells and (ii) metabolic interactions between cancer cells and the tumor microenvironment (TME) at different stages of the metastatic cascade. We identify advantages and disadvantages of each method and discuss how these tools and technologies will further improve our understanding of metastasis. Studies investigating the complex metabolic rewiring of different cells using state-of-the-art metabolomic technologies have the potential to reveal novel biological processes and therapeutic interventions for human cancers.
Collapse
Affiliation(s)
| | - Nicholas P Lesner
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marie Sabatier
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jessalyn M Ubellacker
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Alpaslan Tasdogan
- Department of Dermatology, University Hospital Essen and German Cancer Consortium, Partner Site, Essen, Germany.
| |
Collapse
|
157
|
Shasha T, Gruijs M, van Egmond M. Mechanisms of colorectal liver metastasis development. Cell Mol Life Sci 2022; 79:607. [PMID: 36436127 PMCID: PMC9701652 DOI: 10.1007/s00018-022-04630-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/11/2022] [Accepted: 11/13/2022] [Indexed: 11/28/2022]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related death worldwide, largely due to the development of colorectal liver metastases (CRLM). For the establishment of CRLM, CRC cells must remodel their tumor-microenvironment (TME), avoid the immune system, invade the underlying stroma, survive the hostile environment of the circulation, extravasate into the liver, reprogram the hepatic microenvironment into a permissive pre-metastatic niche, and finally, awake from a dormant state to grow out into clinically detectable CRLM. These steps form part of the invasion-metastasis cascade that relies on reciprocal interactions between the tumor and its ever-changing microenvironment. Such interplay provides a strong rational for therapeutically targeting the TME. In fact, several TME constituents, such as VEGF, TGF-β coreceptor endoglin, and CXCR4, are already targeted in clinical trials. It is, however, of utmost importance to fully understand the complex interactions in the invasion-metastasis cascade to identify novel potential therapeutic targets and prevent the establishment of CRLM, which may ultimately greatly improve patient outcome.
Collapse
Affiliation(s)
- Tal Shasha
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Mandy Gruijs
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Marjolein van Egmond
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands.
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands.
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Surgery, De Boelelaan 1117, Amsterdam, The Netherlands.
| |
Collapse
|
158
|
Recent Advances in Cancer Vaccines: Challenges, Achievements, and Futuristic Prospects. Vaccines (Basel) 2022; 10:vaccines10122011. [PMID: 36560420 PMCID: PMC9788126 DOI: 10.3390/vaccines10122011] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer is a chronic disease, and it can be lethal due to limited therapeutic options. The conventional treatment options for cancer have numerous challenges, such as a low blood circulation time as well as poor solubility of anticancer drugs. Therapeutic cancer vaccines emerged to try to improve anticancer drugs' efficiency and to deliver them to the target site. Cancer vaccines are considered a viable therapeutic technique for most solid tumors. Vaccines boost antitumor immunity by delivering tumor antigens, nucleic acids, entire cells, and peptides. Cancer vaccines are designed to induce long-term antitumor memory, causing tumor regression, eradicate minimal residual illness, and prevent non-specific or unpleasant effects. These vaccines can assist in the elimination of cancer cells from various organs or organ systems in the body, with minimal risk of tumor recurrence or metastasis. Vaccines and antigens for anticancer therapy are discussed in this review, including current vaccine adjuvants and mechanisms of action for various types of vaccines, such as DNA- or mRNA-based cancer vaccines. Potential applications of these vaccines focusing on their clinical use for better therapeutic efficacy are also discussed along with the latest research available in this field.
Collapse
|
159
|
Jansen L, Dubois BFH, Hollmann MW. The Effect of Propofol versus Inhalation Anesthetics on Survival after Oncological Surgery. J Clin Med 2022; 11:jcm11226741. [PMID: 36431218 PMCID: PMC9696269 DOI: 10.3390/jcm11226741] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
Every year, 19.3 million patients worldwide are diagnosed with cancer. Surgical resection represents a major therapeutical option and the vast majority of these patients receive anesthesia. However, despite surgical resection, almost one third of these patients develop local recurrence or distant metastases. Perioperative factors, such as surgical stress and anesthesia technique, have been suggested to play a role to a greater or lesser extent in the development of recurrences, but oncology encompasses a complicated tumor biology of which much is still unknown. The effect of total intravenous anesthesia (TIVA) or volatile anesthesia (VA) on survival after oncological surgery has become a popular topic in recent years. Multiple studies conclude in favor of propofol. Despite the a priori probability that relevant differences in postoperative outcomes are due to the anesthesia technique employed, TIVA or VA, is extremely small. The existing literature includes mainly hypothesis-forming retrospective studies and small randomized trials with many methodological limitations. To date, it is unlikely that use of TIVA or VA affect cancer-free survival days to a clinically relevant extent. This review addresses all relevant studies in the field and provides a substantiated different view on this deeply controversial research topic.
Collapse
|
160
|
Zhu D, Lu Y, Wang Y, Wang Y. PAD4 and Its Inhibitors in Cancer Progression and Prognosis. Pharmaceutics 2022; 14:2414. [PMID: 36365233 PMCID: PMC9699117 DOI: 10.3390/pharmaceutics14112414] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/28/2022] [Accepted: 11/06/2022] [Indexed: 07/24/2023] Open
Abstract
The systemic spread of malignancies and the risk of cancer-associated thrombosis are major clinical challenges in cancer therapy worldwide. As an important post-translational modification enzyme, peptidyl arginine deiminase 4 (PAD4) could mediate the citrullination of protein in different components (including nucleus and cytoplasm, etc.) of a variety of cells (tumor cells, neutrophils, macrophages, etc.), thus participating in gene regulation, neutrophil extracellular trap (NET) and macrophage extracellular trap (MET). Thereby, PAD4 plays an important role in enhancing the growth of primary tumors and facilitating the distant metastasis of cancer cells. In addition, it is related to the formation of cancer-associated thrombosis. Therefore, the development of PAD4-specific inhibitors may be a promising strategy for treating cancer, and it may improve patient prognosis. In this review, we describe PAD4 involvement in gene regulation, protein citrullination, and NET formation. We also discuss its potential role in cancer and cancer-associated thrombosis, and we summarize the development and application of PAD4 inhibitors.
Collapse
Affiliation(s)
- Di Zhu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Yu Lu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Yanming Wang
- School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Yuji Wang
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| |
Collapse
|
161
|
Hashemi M, Hajimazdarany S, Mohan CD, Mohammadi M, Rezaei S, Olyaee Y, Goldoost Y, Ghorbani A, Mirmazloomi SR, Gholinia N, Kakavand A, Salimimoghadam S, Ertas YN, Rangappa KS, Taheriazam A, Entezari M. Long non-coding RNA/epithelial-mesenchymal transition axis in human cancers: Tumorigenesis, chemoresistance, and radioresistance. Pharmacol Res 2022; 186:106535. [DOI: 10.1016/j.phrs.2022.106535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/22/2022] [Accepted: 10/30/2022] [Indexed: 11/07/2022]
|
162
|
Lu DY, Lu TR, Yarla NS, Xu B. Drug Sensitivity Testing for Cancer Therapy, Key Areas. Rev Recent Clin Trials 2022; 17:291-299. [PMID: 35986532 DOI: 10.2174/1574887117666220819094528] [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: 12/04/2021] [Revised: 04/20/2022] [Accepted: 04/20/2022] [Indexed: 01/15/2023]
Abstract
AIMS Cancer is a high-mortality disease (9.6 million deaths in 2018 worldwide). Given various anticancer drugs, drug selection plays a key role in patient survival in clinical trials. METHODS Drug Sensitivity Testing (DST), one of the leading drug selective systems, was widely practiced for therapeutic promotion in the clinic. Notably, DSTs assist in drug selection that benefits drug responses against cancer from 20-22% to 30-35% over the past two decades. The relationship between drug resistance in vitro and drug treatment benefits was associated with different tumor origins and subtypes. Medical theory and underlying DST mechanisms remain poorly understood until now. The study of the clinical scenario, sustainability and financial support for mechanism and technical promotions is indispensable. RESULTS Despite the great technical advance, therapeutic prediction and drug selection by DST needs to be miniature, versatility and cost-effective in the clinic. Multi-parameters and automation of DST should be a future trend. Advanced biomedical knowledge and clinical approaches to translating oncologic profiles into drug selection were the main focuses of DST developments. With a great technical stride, the clinical architecture of the DST platform was entering higher levels (drug response testing at any stage of cancer patients and miniaturization of tumor samples). DISCUSSION The cancer biology and pharmacology for drug selection mutually benefit the clinic. New proposals to reveal more therapeutic information and drug response prediction at genetic, molecular and omics levels should be estimated overall. CONCLUSION By upholding this goal of non-invasive, versatility and automation, DST could save the life of several thousand annually worldwide. In this article, new insights into DST novelty and development are highlighted.
Collapse
Affiliation(s)
- Da-Yong Lu
- School of Life Sciences, Shanghai University, Shanghai 200444, PRC, China
| | - Ting-Ren Lu
- College of Science, Shanghai University, Shanghai 200444, PRC, China
| | | | - Bin Xu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
| |
Collapse
|
163
|
Karthikeyan S, Casey PJ, Wang M. RAB4A GTPase regulates epithelial-to-mesenchymal transition by modulating RAC1 activation. Breast Cancer Res 2022; 24:72. [PMID: 36307864 DOI: 10.1186/s13058-022-01564-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 10/04/2022] [Indexed: 11/10/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a critical underpinning process for cancer progression, recurrence and resistance to drug treatment. Identification of new regulators of EMT could lead to the development of effective therapies to improve the outcome of advanced cancers. In the current study we discovered, using a variety of in vitro and in vivo approaches, that RAB4A function is essential for EMT and related manifestation of stemness and invasive properties. Consistently, RAB4A suppression abolished the cancer cells' self-renewal and tumor forming ability. In terms of downstream signaling, we found that RAB4A regulation of EMT is achieved through its control of activation of the RAC1 GTPase. Introducing activated RAC1 efficiently rescued EMT gene expression, invasion and tumor formation suppressed by RAB4A knockdown in both the in vitro and in vivo cancer models. In summary, this study identifies a RAB4A-RAC1 signaling axis as a key regulatory mechanism for the process of EMT and cancer progression and suggests a potential therapeutic approach to controlling these processes.
Collapse
Affiliation(s)
- Subbulakshmi Karthikeyan
- Program in Cancer Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Patrick J Casey
- Program in Cancer Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore.,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Mei Wang
- Program in Cancer Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore. .,Department of Biochemistry, National University of Singapore, Singapore, 117596, Singapore.
| |
Collapse
|
164
|
Procaine Abrogates the Epithelial-Mesenchymal Transition Process through Modulating c-Met Phosphorylation in Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:cancers14204978. [PMID: 36291760 PMCID: PMC9599628 DOI: 10.3390/cancers14204978] [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] [Received: 08/16/2022] [Revised: 09/28/2022] [Accepted: 10/06/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Epithelial-mesenchymal transition (EMT) is a vital process that leads to the dissemination of tumor cells to distant organs and promotes cancer progression. Aberrant activation of c-Met has been positively correlated with tumor metastasis in hepatocellular carcinoma (HCC). In this report, we have demonstrated the suppressive effect of procaine on the EMT process through the blockade of the c-Met signaling pathway. Procaine downregulated mesenchymal markers and upregulated epithelial markers. Functionally, procaine abrogated cellular migration and invasion. Moreover, procaine suppressed c-Met and its downstream signaling events in HCC models. We report that procaine can function as a novel inhibitor of the EMT process and c-Met-dependent signaling cascades. These results support the consideration of procaine being tested as a potential anti-metastatic agent. Abstract EMT is a critical cellular phenomenon that promotes tumor invasion and metastasis. Procaine is a local anesthetic agent used in oral surgeries and as an inhibitor of DNA methylation in some types of cancers. In this study, we have investigated whether procaine can inhibit the EMT process in HCC cells and the preclinical model. Procaine suppressed the expression of diverse mesenchymal markers but induced the levels of epithelial markers such as E-cadherin and occludin in HGF-stimulated cells. Procaine also significantly reduced the invasion and migration of HCC cells. Moreover, procaine inhibited HGF-induced c-Met and its downstream oncogenic pathways, such as PI3K/Akt/mTOR and MEK/ERK. Additionally, procaine decreased the tumor burden in the HCC mouse model and abrogated lung metastasis. Overall, our study suggests that procaine may inhibit the EMT process through the modulation of a c-Met signaling pathway.
Collapse
|
165
|
Prasad Shenoy G, Pal R, Gurubasavaraja Swamy P, Singh E, Manjunathaiah Raghavendra N, Sanjay Dhiwar P. Discoidin Domain Receptor Inhibitors as Anticancer Agents: A Systematic Review on Recent Development of DDRs Inhibitors, their Resistance and Structure Activity Relationship. Bioorg Chem 2022; 130:106215. [DOI: 10.1016/j.bioorg.2022.106215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/12/2022] [Accepted: 10/15/2022] [Indexed: 11/02/2022]
|
166
|
Chen S, Huang W, Liu Z, Jin M, Li J, Meng L, Li T, Diao Y, Gao H, Hong C, Zheng J, Li F, Zhang Y, Bi D, Teng L, Li X. Identification of nine mutant genes and establishment of three prediction models of organ tropism metastases of non-small cell lung cancer. Cancer Med 2022; 12:3089-3100. [PMID: 36161776 PMCID: PMC9939125 DOI: 10.1002/cam4.5233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Most Non-small cell lung cancer (NSCLC) patients tend to have metastases at the initial diagnosis. However, limited knowledge has been established regarding which factors, are associated with its metastases. This study aims to identify more biomarkers associated with its organ tropism metastasis and to establish models for prediction of its metastatic organs. METHODS We performed targeted next-generation sequencing (NGS) to detect genes related to lung cancer in 272 patients with primary advanced NSCLC from Northeast China. We adopted Fisher test, multivariate logistic regression analysis to identify metastasis-related gene mutations and to establish prediction models. RESULTS Mutations of EGFR (p = 0.0003, OR = 2.554) (especially EGFR L858R [p = 0.02, OR = 2.009]), ATM (p = 0.008, OR = 11.032), and JAK2 (p = 0.009, OR = Inf) were positively and of TP53 exon4mut (p = 0.001, OR = 0.173) was negatively correlated with lung metastasis, and those of CSF1R (p = 0.01, OR = Inf), KIT (p = 0.03, OR = 4.746), MYC (p = 0.05, OR = 7.938), and ERBB2 (p = 0.02, OR = 2.666) were positively correlated with pleural dissemination; those of TP53 (p = 0.01, OR = 0.417) was negatively, while of SMAD4 (p = 0.03, OR = 4.957) was positively correlated with brain metastasis of NSCLC. Additionally, smoking history (p = 0.004, OR = 0.004) was negatively correlated with pleural dissemination of NSCLC. Furthermore, models for prediction of lung metastasis (AUC = 0.706), pleural dissemination (AUC = 0.651), and brane metastasis (AUC = 0.629) were established. CONCLUSION Taken together, this study revealed nine mutant genes and smoking history associated with organ tropism metastases of NSCLC and provided three models for the prediction of metastatic organs. This study enables us to predict the organs to which non-small cell lung cancer metastasizes before it does develop.
Collapse
Affiliation(s)
- Shuchen Chen
- Department of Thoracic Medicine, Cancer Hospital of China Medical UniversityLiaoning Cancer Hospital and InstituteShenyangChina
| | - Wanyi Huang
- School and Hospital of StomatologyChina Medical University, Liaoning Provincial Key Laboratory of Oral DiseasesShenyangChina,Department of Aging Science and Pharmacology, Faculty of Dental ScienceKyushu UniversityFukuokaJapan
| | - Zhenzhen Liu
- Department of Thoracic Medicine, Cancer Hospital of China Medical UniversityLiaoning Cancer Hospital and InstituteShenyangChina
| | - Meizi Jin
- Department of Thoracic Medicine, Cancer Hospital of China Medical UniversityLiaoning Cancer Hospital and InstituteShenyangChina
| | - Jielin Li
- Department of Thoracic Medicine, Cancer Hospital of China Medical UniversityLiaoning Cancer Hospital and InstituteShenyangChina
| | - Lihui Meng
- Department of Thoracic Medicine, Cancer Hospital of China Medical UniversityLiaoning Cancer Hospital and InstituteShenyangChina
| | - Ting Li
- Department of Thoracic Medicine, Cancer Hospital of China Medical UniversityLiaoning Cancer Hospital and InstituteShenyangChina
| | - Yuzhu Diao
- Department of Thoracic Medicine, Cancer Hospital of China Medical UniversityLiaoning Cancer Hospital and InstituteShenyangChina
| | - Hong Gao
- Department of Thoracic Medicine, Cancer Hospital of China Medical UniversityLiaoning Cancer Hospital and InstituteShenyangChina
| | - Chengyu Hong
- Department of Thoracic Medicine, Cancer Hospital of China Medical UniversityLiaoning Cancer Hospital and InstituteShenyangChina
| | - Jian Zheng
- Department of Thoracic Medicine, Cancer Hospital of China Medical UniversityLiaoning Cancer Hospital and InstituteShenyangChina
| | - Fei Li
- Department of Thoracic Medicine, Cancer Hospital of China Medical UniversityLiaoning Cancer Hospital and InstituteShenyangChina
| | - Yue Zhang
- Hangzhou Jichenjunchuang Medical Laboratory Co. Ltd.HangzhouChina
| | - Dan Bi
- Hangzhou Jichenjunchuang Medical Laboratory Co. Ltd.HangzhouChina
| | - Lin Teng
- Hangzhou Jichenjunchuang Medical Laboratory Co. Ltd.HangzhouChina
| | - Xiaoling Li
- Department of Thoracic Medicine, Cancer Hospital of China Medical UniversityLiaoning Cancer Hospital and InstituteShenyangChina
| |
Collapse
|
167
|
Huang D, Wu T, Lan S, Liu C, Guo Z, Zhang W. In situ photothermal nano-vaccine based on tumor cell membrane-coated black phosphorus-Au for photo-immunotherapy of metastatic breast tumors. Biomaterials 2022; 289:121808. [PMID: 36137415 DOI: 10.1016/j.biomaterials.2022.121808] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/01/2022] [Accepted: 09/13/2022] [Indexed: 11/02/2022]
Abstract
Cancer vaccines which can activate antitumor immune response have great potential for metastatic tumors treatment. However, clinical translation of cancer vaccines remained challenging due to weak tumor antigen immunogenicity, inefficient in vivo delivery, and immunosuppressive tumor microenvironment. Nanomaterials-based photothermal treatment (PTT) triggers immunogenic cell death while providing in situ tumor-associated antigens for subsequent anti-tumor immunity. Here, an in situ photothermal nano-vaccine (designated as BCNCCM) based on cancer cell membrane (CCM) was explored by co-encapsulating immune adjuvant CpG oligodeoxynucleotide (ODN) loaded black phosphorus-Au (BP-Au) nanosheets together with an indoleamine 2,3-dioxygenase (IDO) inhibitor (NLG919) by CCM, for the elimination of primary and metastatic breast tumors. The nano-vaccine could be delivered to tumor site selectively by CCM targeting and exhibit vaccine-like functions through the combined effect of in situ generated tumor-associate agents after PTT and immune adjuvant CpG, resulting in trigger of tumor-specific immunity. Furthermore, tumor inhibition was enhanced owing to the reversed immunosuppressive microenvironment mediated by IDO inhibitors. The nano-vaccine not only had good therapeutic effect on primary and metastatic tumors, but also could prevent tumor recurrence by producing systemic immune memory. Therefore, the photothermal nano-vaccine which coordinate in situ vaccine-like function and immune modulation may be a promising stragegy for photo-immunotherapy of metastatic tumors.
Collapse
Affiliation(s)
- Deqiu Huang
- School of Medical Information Engineering, Guangzhou University of Chinese Medicine. Guangzhou. Guangdong. PR China
| | - Tong Wu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine. Guangzhou. Guangdong. PR China
| | - Siyuan Lan
- Research Center for Integrative Medicine of Guangzhou University of Chinese Medicine (Key Laboratory of Chinese Medicine Pathogenesis and Therapy Research), School of Basic Medical Sciences, Guangzhou University of Chinese Medicine. Guangzhou. Guangdong. PR China
| | - Chengkuan Liu
- Research Center for Integrative Medicine of Guangzhou University of Chinese Medicine (Key Laboratory of Chinese Medicine Pathogenesis and Therapy Research), School of Basic Medical Sciences, Guangzhou University of Chinese Medicine. Guangzhou. Guangdong. PR China
| | - Zhouyi Guo
- MOE Key Laboratory of Laser Life Science & SATCM Third Grade Laboratory of Chinese Medicine and Photonics Technology, College of Biophotonics, South China Normal University, Guangzhou, 510631, Guangdong, China.
| | - Wen Zhang
- Research Center for Integrative Medicine of Guangzhou University of Chinese Medicine (Key Laboratory of Chinese Medicine Pathogenesis and Therapy Research), School of Basic Medical Sciences, Guangzhou University of Chinese Medicine. Guangzhou. Guangdong. PR China; Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine. Guangzhou. Guangdong. PR China.
| |
Collapse
|
168
|
Implication of Netrin-1 Gain of Expression in Canine Nodal Lymphoma. Vet Sci 2022; 9:vetsci9090494. [PMID: 36136711 PMCID: PMC9501284 DOI: 10.3390/vetsci9090494] [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] [Received: 06/20/2022] [Revised: 07/22/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Canine lymphomas represent one of the most frequent groups of neoplasia, for which prognosis may be poor. Treatments are based on polychemotherapy, with variable responses. As in human lymphomas, more and more targeted therapies are studied and developed. Therapy to restore apoptosis in neoplastic cells is one of them. Netrin-1 is a ligand of dependence receptors. When bound to its receptor, a positive signaling is triggered. When unbound, apoptosis is induced. In some human cancers, neoplastic cells can lose the ability to induce apoptosis by overexpressing netrin-1, or by decreasing the receptor expression. We hypothesized a similar pathway in canine lymphomas. We observed increased expression of netrin-1, particularly in high-grade nodal lymphomas. In vitro evaluation of an anti-netrin-1 antibody is encouraging as apoptosis is restored in a T-cell lymphoma cell line. Netrin-1 appears thus as a possible survival factor in dog lymphomas. This study suggests it can be a promising tool for a targeted therapy in lymphoma management in dogs. Abstract Netrin-1 is a member of the laminin superfamily, and is known to interact with specific receptors, called dependence receptors. While upon netrin-1 binding these receptors initiate positive signaling, in absence of netrin-1, these receptors trigger apoptosis. Tumor cells can avoid apoptosis by inactivating these receptors or by gaining ligand expression. The aim of the present study was to investigate the expression of netrin-1, the ligand of dependence receptors, in canine healthy lymph nodes (LN), and in lymphomas and to evaluate efficiency of a netrin-1 interfering compound in cell cultures from canine lymphoma. Thirty-two control LN and 169 lymphomas were analyzed through immunohistochemistry. Netrin-1 was expressed in the nucleoli of lymphoid and non-lymphoid cells in controls. Acquisition of a cytoplasmic expression was present in B-cell lymphomas (23.1 % in low-grade and 50.6% in high-grade) and T-cell lymphomas (50.0 % in low-grade and 78.8 % in high-grade), with a significant difference between the high- and low-grade in B-cell lymphomas. Through flow cytometry, we showed a significant increase in netrin-1 expression in either high-grade B-cell and T-cell lymphomas (19 and 5, respectively) compared with healthy LN (5), likewise an RT-qPCR analysis demonstrated a significant increase in netrin-1 expression level in 14 samples of lymphomas compared with eight samples of healthy LN. A T-cell aggressive canine lymphoma cell line and four primary canine nodal lymphomas cell cultures were treated with a netrin-1 interfering antibody. Apoptosis by measuring caspase 3 activity was significantly increased in the cell line and viability was decreased in three of the four primary cell cultures. Together, these data suggest that netrin-1 expression is increased in lymphoma, and more specifically in high-grade lymphomas, and that netrin-1 can act as a survival factor for the neoplastic cells, and so be a therapeutic target.
Collapse
|
169
|
Immunoglobulin superfamily 9 (IGSF9) is trans-activated by p53, inhibits breast cancer metastasis via FAK. Oncogene 2022; 41:4658-4672. [PMID: 36088502 PMCID: PMC9546770 DOI: 10.1038/s41388-022-02459-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 11/08/2022]
Abstract
AbstractMetastasis of breast cancer represents the major reason for its poor prognosis, leading to high mortality. In breast cancer, a tumor suppressor gene TP53 is commonly mutated. TP53 mutation leads to an altered expression of various genes, an event that is associated with aggressive tumor and is a strong independent marker for survival. In this study, we identified a novel p53 target gene, immunoglobulin superfamily 9 (IGSF9). IGSF9 is generally down-regulated in breast cancer tissues. Loss of IGSF9 is associated with frequent metastasis and poor prognosis of breast cancer patients. Wild-type p53, but not R175H mutant, trans-activates the transcription of IGSF9 via binding to its promoter (−137 to −131 bp), inhibits epithelial-mesenchymal transition (EMT), consequently the inhibition of breast cancer cells migration and invasion. IGSF9 interacts with focal adhesion kinase (FAK) and inhibits FAK/AKT signaling activity. PND1186, FAK inhibitor, inhibits breast cancer metastasis induced by IGSF9 knockdown in vitro and in vivo. Taken together, IGSF9 is trans-activated by p53 and inhibits breast cancer metastasis by modulating FAK/AKT signaling pathway. IGSF9 could serve as a prognostic marker and potential therapeutic target for breast cancer.
Collapse
|
170
|
Sun Z, Li P, Wu Z, Li B, Li W, Zhao M, Zhou X, Wang Z, Yu Z, Liu W, Zhu W, Wang H, Wang Y. Circulating CD45+EpCAM+ cells as a diagnostic marker for early-stage primary lung cancer. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:982308. [PMID: 36147748 PMCID: PMC9487715 DOI: 10.3389/fmedt.2022.982308] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Lung cancer is a highly prevalent type of cancer, accounting for 11.6% of all cancer incidences. Early detection and treatment can significantly improve the survival rate and quality of life of patients; however, there is no accurate, effective, and easy-to-use test for early lung cancer screening. In this study, flow cytometry was used to detect the presence of CD45+EpCAM+ cells in tumor tissues and peripheral blood mononuclear cells (PBMCs) in patients with lung cancer. Moreover, the proportion of CD45+EpCAM+ cells in PBMCs of patients with lung cancer was found to be significantly higher than that of healthy volunteers. Tumor-related serum markers level was also measured in the peripheral blood of these patients using an electrochemiluminescence assay. The correlation between CD45+EpCAM+ cells, carcinoembryonic antigen (CEA), and lung cancer was investigated using receiver operating characteristic (ROC) curve analysis, which showed the sensitivity and specificity of the CD45+EpCAM+ cell to be 81.58% and 88.89%, respectively. Further analysis yielded an area under the ROC curve (ROC/area under the curve [AUC]) of 0.845 in patients PBMCs with lung cancer, which was slightly higher than that of CEA (0.732). Therefore, the detection of CD45+EpCAM+ cells in PBMCs may be helpful for the early screening and auxiliary diagnosis of lung cancer.
Collapse
Affiliation(s)
- Zhen Sun
- Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, China
| | - Peng Li
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhaojun Wu
- Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, China
| | - Bin Li
- Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, China
| | - Wenjing Li
- Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, China
| | | | - Xiaobin Zhou
- Department of Epidemiology and Health Statistics, School of Public Health, Qingdao University, Qingdao, China
| | - Zeyao Wang
- Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, China
| | - Zhongjie Yu
- Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, China
| | - Wenna Liu
- Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, China
| | - Wenshu Zhu
- Qingdao Sino-Cell Biomed Co., Ltd., Qingdao, China
| | - Haibo Wang
- Institute of Translational Research for Solid Tumor, Qingdao University, Qingdao, China
- *Correspondence: Haibo Wang
| | - Yongjie Wang
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Yongjie Wang
| |
Collapse
|
171
|
Trivedi S, Bhattacharya M, Starz-Gaiano M. Mind bomb 2 promotes cell migration and epithelial structure by regulating adhesion complexes and the actin cytoskeleton. Dev Biol 2022; 491:94-104. [PMID: 36067835 DOI: 10.1016/j.ydbio.2022.08.009] [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: 10/01/2021] [Revised: 07/29/2022] [Accepted: 08/27/2022] [Indexed: 11/03/2022]
Abstract
Cell migration is essential in animal development and co-opted during metastasis and inflammatory diseases. Some cells migrate collectively, which requires them to balance epithelial characteristics such as stable cell-cell adhesions with features of motility like rapid turnover of adhesions and dynamic cytoskeletal structures. How this is regulated is not entirely clear but important to understand. While investigating Drosophila oogenesis, we found that the putative E3 ubiquitin ligase, Mind bomb 2 (Mib2), is required to promote epithelial stability and the collective cell migration of border cells. Through biochemical analysis, we identified components of Mib2 complexes, which include E-cadherin and α- and β-catenins, as well as actin regulators. We also found that three Mib2 interacting proteins, RhoGAP19D, Supervillin, and Myosin heavy chain-like, affect border cell migration. mib2 mutant main body follicle cells have drastically reduced E-cadherin-based adhesion complexes and diminished actin filaments. We conclude that Mib2 acts to stabilize E-cadherin-based adhesion complexes and promote a robust actin cytoskeletal network, which is important for maintenance of epithelial integrity. The interaction with cadherin adhesion complexes and other cytoskeletal regulators contribute to its role in collective cell migration. Since Mib2 is well conserved, it may have similar functional significance in other organisms.
Collapse
Affiliation(s)
- Sunny Trivedi
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD, 21250, USA
| | - Mallika Bhattacharya
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD, 21250, USA
| | - Michelle Starz-Gaiano
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD, 21250, USA.
| |
Collapse
|
172
|
Elwood P, Protty M, Morgan G, Pickering J, Delon C, Watkins J. Aspirin and cancer: biological mechanisms and clinical outcomes. Open Biol 2022; 12:220124. [PMID: 36099932 PMCID: PMC9470249 DOI: 10.1098/rsob.220124] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Evidence on aspirin and cancer comes from two main sources: (1) the effect of aspirin upon biological mechanisms in cancer, and (2) clinical studies of patients with cancer, some of whom take aspirin. A series of systematic literature searches identified published reports relevant to these two sources. The effects of aspirin upon biological mechanisms involved in cancer initiation and growth appear to generate reasonable expectations of effects upon the progress and mortality of cancer. Clinical evidence on aspirin appears overall to be favourable to the use of aspirin, but evidence from randomized trials is limited, and inconsistent. The main body of evidence comes from meta-analyses of observational studies of patients with a wide range of cancers, about 25% of whom were taking aspirin. Heterogeneity is large but, overall, aspirin is associated with increases in survival and reductions in metastatic spread and vascular complications of different cancers. It is important that evaluations of aspirin used as an adjunct cancer treatment are based upon all the available relevant evidence, and there appears to be a marked harmony between the effects of aspirin upon biological mechanisms and upon the clinical progress of cancer.
Collapse
Affiliation(s)
- Peter Elwood
- Division of Population Medicine, University of Cardiff, Cardiff, Wales CF10 3AT, UK
| | - Majd Protty
- Department of Cardiology, Cardiff Lipidomic Group, University of Cardiff, Cardiff, Wales, UK
| | | | - Janet Pickering
- Division of Population Medicine, University of Cardiff, Cardiff, Wales CF10 3AT, UK
| | | | - John Watkins
- Division of Population Medicine, University of Cardiff, Cardiff, Wales CF10 3AT, UK
| |
Collapse
|
173
|
Pace J, Ivich F, Marple E, Niedre M. Near-infrared diffuse in vivo flow cytometry. JOURNAL OF BIOMEDICAL OPTICS 2022; 27:JBO-220101GR. [PMID: 36114606 PMCID: PMC9478904 DOI: 10.1117/1.jbo.27.9.097002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/07/2022] [Indexed: 06/15/2023]
Abstract
Significance Diffuse in vivo flow cytometry (DiFC) is an emerging technique for enumerating rare fluorescently labeled circulating cells noninvasively in the bloodstream. Thus far, we have reported red and blue-green versions of DiFC. Use of near-infrared (NIR) fluorescent light would in principle allow use of DiFC in deeper tissues and would be compatible with emerging NIR fluorescence molecular contrast agents. Aim We describe the design of a NIR-DiFC instrument and demonstrate its use in optical flow phantoms in vitro and in mice in vivo. Approach We developed an improved optical fiber probe design for efficient collection of fluorescence from individual circulating cells and efficient rejection of instrument autofluorescence. We built a NIR-DiFC instrument. We tested this with NIR fluorescent microspheres and cell lines labeled with OTL38 fluorescence contrast agent in a flow phantom model. We also tested NIR-DiFC in nude mice injected intravenously with OTL38-labeled L1210A cells. Results NIR-DiFC allowed detection of circulating tumor cells (CTCs) in flow phantoms with mean signal-to-noise ratios (SNRs) of 19 to 32 dB. In mice, fluorescently labeled CTCs were detectable with mean SNR of 26 dB. NIR-DiFC also exhibited orders significantly lower autofluorescence and false-alarm rates than blue-green DiFC. Conclusions NIR-DiFC allows use of emerging NIR contrast agents. Our work could pave the way for future use of NIR-DiFC in humans.
Collapse
Affiliation(s)
- Joshua Pace
- Northeastern University, Department of Bioengineering, Boston, Massachusetts, United States
| | - Fernando Ivich
- Northeastern University, Department of Bioengineering, Boston, Massachusetts, United States
| | - Eric Marple
- EmVision LLC, Loxahatchee, Florida, United States
| | - Mark Niedre
- Northeastern University, Department of Bioengineering, Boston, Massachusetts, United States
| |
Collapse
|
174
|
Anti-Tumoral Effect of Chemerin on Ovarian Cancer Cell Lines Mediated by Activation of Interferon Alpha Response. Cancers (Basel) 2022; 14:cancers14174108. [PMID: 36077645 PMCID: PMC9454566 DOI: 10.3390/cancers14174108] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/13/2022] [Accepted: 08/22/2022] [Indexed: 11/26/2022] Open
Abstract
Simple Summary Chemerin is a multifunctional protein with an important role in the immune system. Recent evidence showed that chemerin also regulates the development of cancer. Ovarian cancer is a common type of tumor in women. In this study, we observed that chemerin decreases the growth of ovarian cancer cell lines in vitro when cultivated in standard cell culture or in globular multicellular aggregates. When we examined the mechanisms involved in this process, we found that treatment of ovarian cancer cells with chemerin led to the activation of genes that are known to mediate the effects of interferon alpha (IFNα). The main effect of IFNα is to defend body cells against viral infections, but it is also able to defeat cancer cells. We observed that this activation of IFNα response by chemerin resulted from the increased production of IFNα protein in ovarian cancer cells, which then reduced cancer cells numbers. However, it remains to be investigated how exactly chemerin might be able to activate interferon alpha and its anti-tumoral actions. Abstract The pleiotropic adipokine chemerin affects tumor growth primarily as anti-tumoral chemoattractant inducing immunocyte recruitment. However, little is known about its effect on ovarian adenocarcinoma. In this study, we examined chemerin actions on ovarian cancer cell lines in vitro and intended to elucidate involved cell signaling mechanisms. Employing three ovarian cancer cell lines, we observed differentially pronounced effects of this adipokine. Treatment with chemerin (huChem-157) significantly reduced OVCAR-3 cell numbers (by 40.8% on day 6) and decreased the colony and spheroid growth of these cells by half. The spheroid size of SK-OV-3 ovarian cancer cells was also significantly reduced upon treatment. Transcriptome analyses of chemerin-treated cells revealed the most notably induced genes to be interferon alpha (IFNα)-response genes like IFI27, OAS1 and IFIT1 and their upstream regulator IRF9 in all cell lines tested. Finally, we found this adipokine to elevate IFNα levels about fourfold in culture medium of the employed cell lines. In conclusion, our data for the first time demonstrate IFNα as a mediator of chemerin action in vitro. The observed anti-tumoral effect of chemerin on ovarian cancer cells in vitro was mediated by the notable activation of IFNα response genes, resulting from the chemerin-triggered increase of secreted levels of this cytokine.
Collapse
|
175
|
Greenlee JD, Liu K, Lopez-Cavestany M, King MR. Piezo1 Mechano-Activation Is Augmented by Resveratrol and Differs between Colorectal Cancer Cells of Primary and Metastatic Origin. Molecules 2022; 27:5430. [PMID: 36080197 PMCID: PMC9458129 DOI: 10.3390/molecules27175430] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/09/2022] [Accepted: 08/20/2022] [Indexed: 11/24/2022] Open
Abstract
Cancer cells must survive aberrant fluid shear stress (FSS) in the circulation to metastasize. Herein, we investigate the role that FSS has on colorectal cancer cell apoptosis, proliferation, membrane damage, calcium influx, and therapeutic sensitization. We tested this using SW480 (primary tumor) and SW620 cells (lymph node metastasis) derived from the same patient. The cells were exposed to either shear pulses, modeling millisecond intervals of high FSS seen in regions of turbulent flow, or sustained shear to model average magnitudes experienced by circulating tumor cells. SW480 cells were significantly more sensitive to FSS-induced death than their metastatic counterparts. Shear pulses caused significant cell membrane damage, while constant shear decreased cell proliferation and increased the expression of CD133. To investigate the role of mechanosensitive ion channels, we treated cells with the Piezo1 agonist Yoda1, which increased intracellular calcium. Pretreatment with resveratrol further increased the calcium influx via the lipid-raft colocalization of Piezo1. However, minimal changes in apoptosis were observed due to calcium saturation, as predicted via a computational model of apoptosis. Furthermore, SW480 cells had increased levels of Piezo1, calcium influx, and TRAIL-mediated apoptosis compared to SW620 cells, highlighting differences in the mechano-activation of metastatic cells, which may be a necessary element for successful dissemination in vivo.
Collapse
Affiliation(s)
| | | | | | - Michael R. King
- Department of Biomedical Engineering, Vanderbilt University, PMB 351631, 2301 Vanderbilt Place, Nashville, TN 37235-1631, USA
| |
Collapse
|
176
|
Jinesh GG, Brohl AS. Classical epithelial-mesenchymal transition (EMT) and alternative cell death process-driven blebbishield metastatic-witch (BMW) pathways to cancer metastasis. Signal Transduct Target Ther 2022; 7:296. [PMID: 35999218 PMCID: PMC9399134 DOI: 10.1038/s41392-022-01132-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 07/14/2022] [Accepted: 07/24/2022] [Indexed: 12/13/2022] Open
Abstract
Metastasis is a pivotal event that accelerates the prognosis of cancer patients towards mortality. Therapies that aim to induce cell death in metastatic cells require a more detailed understanding of the metastasis for better mitigation. Towards this goal, we discuss the details of two distinct but overlapping pathways of metastasis: a classical reversible epithelial-to-mesenchymal transition (hybrid-EMT)-driven transport pathway and an alternative cell death process-driven blebbishield metastatic-witch (BMW) transport pathway involving reversible cell death process. The knowledge about the EMT and BMW pathways is important for the therapy of metastatic cancers as these pathways confer drug resistance coupled to immune evasion/suppression. We initially discuss the EMT pathway and compare it with the BMW pathway in the contexts of coordinated oncogenic, metabolic, immunologic, and cell biological events that drive metastasis. In particular, we discuss how the cell death environment involving apoptosis, ferroptosis, necroptosis, and NETosis in BMW or EMT pathways recruits immune cells, fuses with it, migrates, permeabilizes vasculature, and settles at distant sites to establish metastasis. Finally, we discuss the therapeutic targets that are common to both EMT and BMW pathways.
Collapse
Affiliation(s)
- Goodwin G Jinesh
- Department of Molecular Oncology, 12902 USF Magnolia Drive, H. Lee Moffitt Cancer Center & Research Institute, Tampa, 33612, FL, USA. .,Sarcoma Department, 12902 USF Magnolia Drive, H. Lee Moffitt Cancer Center & Research Institute, Tampa, 33612, FL, USA.
| | - Andrew S Brohl
- Department of Molecular Oncology, 12902 USF Magnolia Drive, H. Lee Moffitt Cancer Center & Research Institute, Tampa, 33612, FL, USA. .,Sarcoma Department, 12902 USF Magnolia Drive, H. Lee Moffitt Cancer Center & Research Institute, Tampa, 33612, FL, USA.
| |
Collapse
|
177
|
Sarvari P, Sarvari P, Ramírez-Díaz I, Mahjoubi F, Rubio K. Advances of Epigenetic Biomarkers and Epigenome Editing for Early Diagnosis in Breast Cancer. Int J Mol Sci 2022; 23:ijms23179521. [PMID: 36076918 PMCID: PMC9455804 DOI: 10.3390/ijms23179521] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 12/02/2022] Open
Abstract
Epigenetic modifications are known to regulate cell phenotype during cancer progression, including breast cancer. Unlike genetic alterations, changes in the epigenome are reversible, thus potentially reversed by epi-drugs. Breast cancer, the most common cause of cancer death worldwide in women, encompasses multiple histopathological and molecular subtypes. Several lines of evidence demonstrated distortion of the epigenetic landscape in breast cancer. Interestingly, mammary cells isolated from breast cancer patients and cultured ex vivo maintained the tumorigenic phenotype and exhibited aberrant epigenetic modifications. Recent studies indicated that the therapeutic efficiency for breast cancer regimens has increased over time, resulting in reduced mortality. Future medical treatment for breast cancer patients, however, will likely depend upon a better understanding of epigenetic modifications. The present review aims to outline different epigenetic mechanisms including DNA methylation, histone modifications, and ncRNAs with their impact on breast cancer, as well as to discuss studies highlighting the central role of epigenetic mechanisms in breast cancer pathogenesis. We propose new research areas that may facilitate locus-specific epigenome editing as breast cancer therapeutics.
Collapse
Affiliation(s)
- Pourya Sarvari
- Department of Clinical Genetics, National Institute of Genetic Engineering and Biotechnology, Tehran P.O. Box 14965/161, Iran
| | - Pouya Sarvari
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Puebla 72160, Mexico
| | - Ivonne Ramírez-Díaz
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Puebla 72160, Mexico
- Facultad de Biotecnología, Campus Puebla, Universidad Popular Autónoma del Estado de Puebla (UPAEP), Puebla 72410, Mexico
| | - Frouzandeh Mahjoubi
- Department of Clinical Genetics, National Institute of Genetic Engineering and Biotechnology, Tehran P.O. Box 14965/161, Iran
| | - Karla Rubio
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Puebla 72160, Mexico
- Licenciatura en Médico Cirujano, Universidad de la Salud del Estado de Puebla (USEP), Puebla 72000, Mexico
- Correspondence:
| |
Collapse
|
178
|
Ryu WK, Oh S, Lim JH, Lee SJ, Shin HT, Ryu JS. Monitoring Circulating Tumor DNA in Untreated Non-Small-Cell Lung Cancer Patients. Int J Mol Sci 2022; 23:ijms23179527. [PMID: 36076922 PMCID: PMC9455735 DOI: 10.3390/ijms23179527] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/14/2022] [Accepted: 08/20/2022] [Indexed: 11/29/2022] Open
Abstract
Circulating tumor DNA (ctDNA) has been utilized to monitor the clinical course of patients of non-small-cell lung cancer (NSCLC) who receive therapies targeting druggable mutations. However, despite providing valuable information on how NSCLC would naturally progress, the clinical utility of ctDNA for clinical-course monitoring and prediction of treatment-naïve NSCLC patients without druggable mutations remain unknown. We longitudinally followed a total of 12 treatment-naïve NSCLC patients, who did not harbor EGFR and ALK mutations, by collecting clinical information, radiological data, and plasma samples. Changes in ctDNA levels and tumor burden (TB) were compared with each other. New metastasis development, volume doubling time (VDT), and overall survival (OS) were analyzed regarding ctDNA detection at diagnosis. ctDNA was detected in the plasma of seven (58.3%) patients. Changes in ctDNA levels correlated with those in TB in a substantial fraction (57.1%) of patients and was also associated with brain metastasis, tumor necrosis, or pneumonia in other patients. All patients with ctDNA detection developed new metastasis during follow-ups in the organs that had been devoid of metastasis at diagnosis. The patients without ctDNA detection did not develop new metastasis (median duration of follow-ups: 9.8 months). In addition, patients with ctDNA detection had shorter VDT (p = 0.039) and worse OS (p = 0.019) than those without ctDNA detection. The natural course of NSCLC progression can be monitored by measuring ctDNA levels. Detection of ctDNA at diagnosis can predict development of new metastasis, rapid tumor growth and poor survival of NSCLC patients.
Collapse
Affiliation(s)
- Woo Kyung Ryu
- Department of Internal Medicine, Inha University Hospital, Incheon 22332, Korea
| | - Sekyung Oh
- Department of Medical Sciences, Catholic Kwandong University College of Medicine, Incheon 22711, Korea
| | - Jun Hyeok Lim
- Department of Internal Medicine, Inha University Hospital, Incheon 22332, Korea
| | | | - Hyun-Tae Shin
- Department of Dermatology, Inha University Hospital, Incheon 22332, Korea
- Research Center for Controlling Intercellular Communication (RCIC), College of Medicine, Inha University, Incheon 22212, Korea
- Correspondence: (H.-T.S.); (J.-S.R.)
| | - Jeong-Seon Ryu
- Department of Internal Medicine, Inha University Hospital, Incheon 22332, Korea
- Correspondence: (H.-T.S.); (J.-S.R.)
| |
Collapse
|
179
|
Mokini Z, Cama A, Forget P. Anesthetics and Long Term Cancer Outcomes: May Epigenetics Be the Key for Pancreatic Cancer? MEDICINA (KAUNAS, LITHUANIA) 2022; 58:1102. [PMID: 36013569 PMCID: PMC9414834 DOI: 10.3390/medicina58081102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 06/15/2023]
Abstract
Knowledge shows a divergence of results between preclinical and clinical studies regarding anesthesia and postoperative progression of cancer. While laboratory and animal data from then 2000s onwards raised much enthusiasm in this field of research leading to several clinical investigations worldwide, data from randomized trials seem to have killed off hope for many scientists. However several aspects of the actual knowledge should be reevaluated and there is space for new strategies of investigation. In this paper, we perform a critical review of actual knowledge and propose new research strategies with a special focus on anesthetic management and repurposed anesthetic adjuvants for pancreatic cancer.
Collapse
Affiliation(s)
- Zhirajr Mokini
- ESAIC Mentorship Program, BE-1000 Brussels, Belgium
- The European Platform for Research Outcomes after PerIoperative Interventions in Surgery for Cancer Research Group (Euro-Periscope): The Onco-Anaesthesiology Research Group (RG), BE-1000 Brussels, Belgium
| | - Alessandro Cama
- The European Platform for Research Outcomes after PerIoperative Interventions in Surgery for Cancer Research Group (Euro-Periscope): The Onco-Anaesthesiology Research Group (RG), BE-1000 Brussels, Belgium
- Department of Pharmacy, G. d’Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
| | - Patrice Forget
- The European Platform for Research Outcomes after PerIoperative Interventions in Surgery for Cancer Research Group (Euro-Periscope): The Onco-Anaesthesiology Research Group (RG), BE-1000 Brussels, Belgium
- Epidemiology Group, Institute of Applied Health Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
- Department of Anaesthesia, National Health Service (NHS) Grampian, Aberdeen AB25 2ZD, UK
| |
Collapse
|
180
|
Li F, Liang H, You H, Xiao J, Xia H, Chen X, Huang M, Cheng Z, Yang C, Liu W, Zhang H, Zeng L, Wu Y, Ge F, Li Z, Zhou W, Wen Y, Zhou Z, Liu R, Jiang D, Xie N, Liang B, Liu Z, Kong Y, Chen C. Targeting HECTD3-IKKα axis inhibits inflammation-related metastasis. Signal Transduct Target Ther 2022; 7:264. [PMID: 35918322 PMCID: PMC9345961 DOI: 10.1038/s41392-022-01057-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 02/07/2023] Open
Abstract
Metastasis is the leading cause of cancer-related death. The interactions between circulating tumor cells and endothelial adhesion molecules in distant organs is a key step during extravasation in hematogenous metastasis. Surgery is a common intervention for most primary solid tumors. However, surgical trauma-related systemic inflammation facilitates distant tumor metastasis by increasing the spread and adhesion of tumor cells to vascular endothelial cells (ECs). Currently, there are no effective interventions to prevent distant metastasis. Here, we show that HECTD3 deficiency in ECs significantly reduces tumor metastasis in multiple mouse models. HECTD3 depletion downregulates expression of adhesion molecules, such as VCAM-1, ICAM-1 and E-selectin, in mouse primary ECs and HUVECs stimulated by inflammatory factors and inhibits adhesion of tumor cells to ECs both in vitro and in vivo. We demonstrate that HECTD3 promotes stabilization, nuclear localization and kinase activity of IKKα by ubiquitinating IKKα with K27- and K63-linked polyubiquitin chains at K296, increasing phosphorylation of histone H3 to promote NF-κB target gene transcription. Knockout of HECTD3 in endothelium significantly inhibits tumor cells lung colonization, while conditional knockin promotes that. IKKα kinase inhibitors prevented LPS-induced pulmonary metastasis. These findings reveal the promotional role of the HECTD3-IKKα axis in tumor hematogenous metastasis and provide a potential strategy for tumor metastasis prevention.
Collapse
Affiliation(s)
- Fubing Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Huichun Liang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.,Department of Pathology, School of Basic Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Hua You
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Ji Xiao
- College of Life Science and Technology, Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, Guangzhou, 510632, China
| | - Houjun Xia
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Maobo Huang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Zhuo Cheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Chuanyu Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Wenjing Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Hailin Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Li Zeng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Yingying Wu
- First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Fei Ge
- First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Zhen Li
- Department of the Third Breast Surgery, the Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650118, China
| | - Wenhui Zhou
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Yi Wen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Zhongmei Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Rong Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Dewei Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Ni Xie
- Biobank, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen, 518035, China
| | - Bin Liang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, 650091, China
| | - Zhenzhen Liu
- Department of Breast disease, Henan Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
| | - Yanjie Kong
- Biobank, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen, 518035, China.
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
| |
Collapse
|
181
|
Palladium-Doped Single-Walled Carbon Nanotubes as a New Adsorbent for Detecting and Trapping Volatile Organic Compounds: A First Principle Study. NANOMATERIALS 2022; 12:nano12152572. [PMID: 35957007 PMCID: PMC9370723 DOI: 10.3390/nano12152572] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/23/2022] [Accepted: 07/23/2022] [Indexed: 01/27/2023]
Abstract
Volatile organic compounds (VOCs) are in the vapor state in the atmosphere and are considered pollutants. Density functional theory (DFT) calculations with the wb97xd exchange correlation functional and the 6-311+G(d,p) basis set are carried out to explore the potential possibility of palladium-doped single-walled carbon nanotubes (Pd/SWCNT-V), serving as the resource for detecting and/or adsorbing acetonitrile (ACN), styrene (STY), and perchloroethylene (PCE) molecules as VOCs. The suggested adsorbent in this study is discussed with structural parameters, frontier molecular orbital theory, molecular electrical potential surfaces (MEPSs), natural bond orbital (NBO) analyses, and the density of states. Furthermore, following the Bader theory of atoms in molecules (AIM), the topological properties of the electron density contributions for intermolecular interactions are analyzed. The obtained results show efficient VOC loading via a strong chemisorption process with a mean adsorption energy of −0.94, −1.27, and −0.54 eV for ACN, STY, and PCE, respectively. Our results show that the Pd/SWCNT-V can be considered a good candidate for VOC removal from the environment.
Collapse
|
182
|
Jiang B, Zhao X, Chen W, Diao W, Ding M, Qin H, Li B, Cao W, Chen W, Fu Y, He K, Gao J, Chen M, Lin T, Deng Y, Yan C, Guo H. Lysosomal protein transmembrane 5 promotes lung-specific metastasis by regulating BMPR1A lysosomal degradation. Nat Commun 2022; 13:4141. [PMID: 35842443 PMCID: PMC9288479 DOI: 10.1038/s41467-022-31783-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 06/30/2022] [Indexed: 12/03/2022] Open
Abstract
Organotropism during cancer metastasis occurs frequently but the underlying mechanism remains poorly understood. Here, we show that lysosomal protein transmembrane 5 (LAPTM5) promotes lung-specific metastasis in renal cancer. LAPTM5 sustains self-renewal and cancer stem cell-like traits of renal cancer cells by blocking the function of lung-derived bone morphogenetic proteins (BMPs). Mechanistic investigations showed that LAPTM5 recruits WWP2, which binds to the BMP receptor BMPR1A and mediates its lysosomal sorting, ubiquitination and ultimate degradation. BMPR1A expression was restored by the lysosomal inhibitor chloroquine. LAPTM5 expression could also serve as an independent predictor of lung metastasis in renal cancer. Lastly, elevation of LAPTM5 expression in lung metastases is a common phenomenon in multiple cancer types. Our results reveal a molecular mechanism underlying lung-specific metastasis and identify LAPTM5 as a potential therapeutic target for cancers with lung metastasis. The mechanisms that confer lung-specific metastasis in renal cell carcinomas (RCC) remain to be detailed. Here the authors show that LAPTM5 contributes to lung-specific metastasis of RCCs by suppressing BMP signalling and thus, enhancing self-renewal and cancer stem cell-like traits of RCCs.
Collapse
Affiliation(s)
- Bo Jiang
- Department of Urology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Xiaozhi Zhao
- Department of Urology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Wei Chen
- Department of Urology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Wenli Diao
- Department of Urology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Meng Ding
- Department of Urology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Haixiang Qin
- Department of Urology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Binghua Li
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Wenmin Cao
- Department of Urology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Wei Chen
- Department of Urology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Yao Fu
- Department of Pathology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Kuiqiang He
- Department of Urology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Jie Gao
- Department of Urology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Mengxia Chen
- Department of Urology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Tingsheng Lin
- Department of Urology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Yongming Deng
- Department of Urology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Chao Yan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
| | - Hongqian Guo
- Department of Urology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, Jiangsu, 210008, China.
| |
Collapse
|
183
|
A dielectrophoresis-based microfluidic system having double-sided optimized 3D electrodes for label-free cancer cell separation with preserving cell viability. Sci Rep 2022; 12:12100. [PMID: 35840699 PMCID: PMC9287561 DOI: 10.1038/s41598-022-16286-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 07/07/2022] [Indexed: 11/25/2022] Open
Abstract
Early detection of circulating tumor cells (CTCs) in a patient's blood is essential to accurate prognosis and effective cancer treatment monitoring. The methods used to detect and separate CTCs should have a high recovery rate and ensure cells viability for post-processing operations, such as cell culture and genetic analysis. In this paper, a novel dielectrophoresis (DEP)-based microfluidic system is presented for separating MDA-MB-231 cancer cells from various subtypes of WBCs with the practical cell viability approach. Three configurations for the sidewall electrodes are investigated to evaluate the separation performance. The simulation results based on the finite-element method show that semi-circular electrodes have the best performance with a recovery rate of nearly 95% under the same operational and geometric conditions. In this configuration, the maximum applied electric field (1.11 × 105 V/m) to separate MDA-MB-231 is lower than the threshold value for cell electroporation. Also, the Joule heating study in this configuration shows that the cells are not damaged in the fluid temperature gradient (equal to 1 K). We hope that such a complete and step-by-step design is suitable to achieve DEP-based applicable cell separation biochips.
Collapse
|
184
|
Mitchell MI, Ma J, Carter CL, Loudig O. Circulating Exosome Cargoes Contain Functionally Diverse Cancer Biomarkers: From Biogenesis and Function to Purification and Potential Translational Utility. Cancers (Basel) 2022; 14:3350. [PMID: 35884411 PMCID: PMC9318395 DOI: 10.3390/cancers14143350] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/01/2022] [Accepted: 07/07/2022] [Indexed: 12/12/2022] Open
Abstract
Although diagnostic and therapeutic treatments of cancer have tremendously improved over the past two decades, the indolent nature of its symptoms has made early detection challenging. Thus, inter-disciplinary (genomic, transcriptomic, proteomic, and lipidomic) research efforts have been focused on the non-invasive identification of unique "silver bullet" cancer biomarkers for the design of ultra-sensitive molecular diagnostic assays. Circulating tumor biomarkers, such as CTCs and ctDNAs, which are released by tumors in the circulation, have already demonstrated their clinical utility for the non-invasive detection of certain solid tumors. Considering that exosomes are actively produced by all cells, including tumor cells, and can be found in the circulation, they have been extensively assessed for their potential as a source of circulating cell-specific biomarkers. Exosomes are particularly appealing because they represent a stable and encapsulated reservoir of active biological compounds that may be useful for the non-invasive detection of cancer. T biogenesis of these extracellular vesicles is profoundly altered during carcinogenesis, but because they harbor unique or uniquely combined surface proteins, cancer biomarker studies have been focused on their purification from biofluids, for the analysis of their RNA, DNA, protein, and lipid cargoes. In this review, we evaluate the biogenesis of normal and cancer exosomes, provide extensive information on the state of the art, the current purification methods, and the technologies employed for genomic, transcriptomic, proteomic, and lipidomic evaluation of their cargoes. Our thorough examination of the literature highlights the current limitations and promising future of exosomes as a liquid biopsy for the identification of circulating tumor biomarkers.
Collapse
Affiliation(s)
- Megan I Mitchell
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Junfeng Ma
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Claire L Carter
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Olivier Loudig
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| |
Collapse
|
185
|
Yun H, Im HJ, Choe C, Roh S. Effect of LOXL2 on metastasis through remodeling of the cell surface matrix in non-small cell lung cancer cells. Gene 2022; 830:146504. [PMID: 35483499 DOI: 10.1016/j.gene.2022.146504] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/08/2022] [Accepted: 04/14/2022] [Indexed: 12/17/2022]
Abstract
Lung cancer is the prominent cause of cancer-associated death primarily because of distant metastatic disease. The metastatic potential of non-small cell lung cancer (NSCLC) is associated with tumor cell aggregation. However, the systemic mechanotransduction mechanism by which tumor cells dynamically aggregate and disseminate is poorly understood, especially in NSCLC. In this study, we examine whether the cell surface matrix plays an important role in metastasis. We used poly-2-hydroxyethyl methacrylate-based 3D spheroid formation methods to mimic in vivo metastatic lesions. Supra-structural analysis of human NSCLC A549 cells stained with ruthenium red for transmission electron microscopy (TEM) showed that glycocalyx surrounding the cell surface in 2D culture decreases in 3D culture. Comprehensive gene expression analysis revealed that the genes associated with cell adhesion were distinctly enriched in A549 cell spheroids. Of these, downregulation of the tumor metastatic microenvironment facilitator LOXL2, a copper-dependent enzyme catalyzing posttranslational oxidative deamination of peptidyl lysine, was of special interest. Knockdown of LOXL2 thickened the cell surface matrix in 2D culture and impaired compact aggregate formation in 3D culture. Moreover, A549 cell spheroids with endogenous overexpression of LOXL2 increased their dissemination on basement extracellular matrix Matrigel. Overall, these data imply that cell detachment-downregulated LOXL2 contributes to cell surface matrix remodeling, leading to collective dissemination of free-floating aggregates.
Collapse
Affiliation(s)
- Heesu Yun
- Cellular Reprogramming and Embryo Biotechnology Lab, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Hee-Jeong Im
- Department of Biomedical Engineering, University of Illinois, Chicago, IL, USA; Jesse Brown Veterans Affairs Medical Center (JBVAMC), Chicago, IL, USA
| | - Chungyoul Choe
- Samsung Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University, School of Medicine, Seoul, Republic of Korea.
| | - Sangho Roh
- Cellular Reprogramming and Embryo Biotechnology Lab, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea.
| |
Collapse
|
186
|
Hu J, Wang M, Yang Y, Xing Y, Li S. LncRNA DLEU2 silencing impedes the migration, invasion and EMT in gastric cancer cell by suppressing PI3K/AKT signaling pathway. Immunopharmacol Immunotoxicol 2022; 44:719-731. [PMID: 35736813 DOI: 10.1080/08923973.2022.2078727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Context: The high expression of long non-coding RNA deleted in lymphocytic leukaemia 2 (lncRNA DLEU2) has been confirmed in gastric cancer (GC).Objective: However, the detailed mechanism concerning its involvement in GC remained unclear, which we aimed to explore in this study.Materials and methods: LncRNA DLEU2 expression in GC was estimated by bioinformatic analysis, and the relationship between the expression of DLEU2 and the clinicopathological characteristics of patients with GC was performed. qRT-PCR was employed to detect the expression of lncRNA DLEU2 and confirm the transfection efficiency following the knockdown or overexpression of DLEU2. Functional assays, including CCK-8, flow cytometry, scratching test and Transwell assays, were used to determine the role of DLEU2 in tumor phenotypes. The effects of DLEU2 on the PI3K/Akt pathway were detected by western blot. For elucidating the functions of DLEU2/PI3K/Akt axis in GC, we inhibited the PI3K/Akt pathway in rescue experiments, and evaluated the expression levels of epithelial-mesenchymal transition (EMT)-related proteins by western blot.Results: The expression of DLEU2 was aberrantly up-regulated in GC tissues and cells, which was correlated with the degree of tumor differentiation, cancer antigen 19-9 (CA19-9) and Lauren histologic classification of patients with GC. Silencing of DLEU2 induced apoptosis, attenuated viability, migration and invasion as well as inhibited the PI3K/Akt signaling pathway in GC cells. Mechanistically, the DLEU2/PI3K/Akt axis promoted the progression of GC and the EMT by down-regulating the expression of E-Cadherin and up-regulating those of N-Cadherin and Vimentin.Discussion and conclusions: LncRNA DLEU2 promoted the migration, invasion and EMT in GC by activating the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Jun Hu
- Oncology Department, Gaochun People's Hospital, Nanjing City, China
| | - Mingyun Wang
- Oncology Department, Gaochun People's Hospital, Nanjing City, China
| | - Yang Yang
- Oncology Department, Nanjing Drum Tower Hospital (Gaochun Branch), Nanjing City, China
| | - Yajun Xing
- Oncology Department, Gaochun People's Hospital, Nanjing City, China
| | - Shuanggen Li
- Oncology Department, Gaochun People's Hospital, Nanjing City, China
| |
Collapse
|
187
|
Jung YY, Chinnathambi A, Alahmadi TA, Alharbi SA, Kumar AP, Sethi G, Ahn KS. Fangchinoline targets epithelial-mesenchymal transition process by modulating activation of multiple cell-signaling pathways. J Cell Biochem 2022; 123:1222-1236. [PMID: 35621239 DOI: 10.1002/jcb.30279] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 12/15/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is a key process, which can promote the transition of tumor cells into other organs by weakening the cell-cell junctions. Tumor cell invasion and metastasis arising because of EMT can determine the prognosis of cancer. EMT can be induced by several growth factors including transforming growth factor-β (TGF-β), which can exert their effects by affecting several cell-signaling pathways. Fangchinoline (FCN), a kind of bisbenzylisoquinoline, belongs to the family Menispermaceae. FCN can display substantial antitumor effects against various malignant cell lines but its possible impact on EMT has not been explored. We examined the potential impact of FCN in affecting the activation of EMT in human colon cancer cells. We evaluated the influence of FCN on EMT in colon cancer cells by using Western blot analysis and reverse transcription-polymerase chain reaction assays. The cellular invasion and migration were observed by Boyden chamber and wound healing assays. Thereafter, the effect of the drug on proliferation and invasion was also evaluated by real-time cell analysis. FCN suppressed the levels of TGF-β-induced mesenchymal markers, such as fibronectin, vimentin, MMP-9, MMP-2, N-cadherin, Twist, and Snail. However, FCN markedly enhanced the expression of epithelial markers such as occludin and E-cadherin. These results imply that FCN can potentially inhibit tumor metastasis through abrogating EMT. In addition, FCN downregulated c-Met/PI3K/Akt/mTOR and Wnt/β-catenin cell signaling pathways and mitigated tumor migration as well as invasion. Overall, our study suggests a potential novel role of FCN as an antimetastatic agent against human colon cancer cells.
Collapse
Affiliation(s)
- Young Y Jung
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Tahani A Alahmadi
- Department of Pediatrics, King Khalid University Hospital [Medical City], King Saud University, Riyadh, Saudi Arabia
| | - Sulaiman A Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Alan P Kumar
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kwang S Ahn
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
188
|
Bhome R, Emaduddin M, James V, House LM, Thirdborough SM, Mellone M, Tulkens J, Primrose JN, Thomas GJ, De Wever O, Mirnezami AH, Sayan AE. Epithelial to mesenchymal transition influences fibroblast phenotype in colorectal cancer by altering miR-200 levels in extracellular vesicles. J Extracell Vesicles 2022; 11:e12226. [PMID: 35595718 PMCID: PMC9122835 DOI: 10.1002/jev2.12226] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 02/14/2022] [Accepted: 04/26/2022] [Indexed: 12/28/2022] Open
Abstract
Colorectal cancer (CRC) with a mesenchymal gene expression signature has the greatest propensity for distant metastasis and is characterised by the accumulation of cancer‐associated fibroblasts in the stroma. We investigated whether the epithelial to mesenchymal transition status of CRC cells influences fibroblast phenotype, with a focus on the transfer of extracellular vesicles (EVs), as a controlled means of cell–cell communication. Epithelial CRC EVs suppressed TGF‐β‐driven myofibroblast differentiation, whereas mesenchymal CRC EVs did not. This was driven by miR‐200 (miR‐200a/b/c, ‐141), which was enriched in epithelial CRC EVs and transferred to recipient fibroblasts. Ectopic miR‐200 expression or ZEB1 knockdown, in fibroblasts, similarly suppressed myofibroblast differentiation. Supporting these findings, there was a strong negative correlation between miR‐200 and myofibroblastic markers in a cohort of CRC patients in the TCGA dataset. This was replicated in mice, by co‐injecting epithelial or mesenchymal CRC cells with fibroblasts and analysing stromal markers of myofibroblastic phenotype. Fibroblasts from epithelial tumours contained more miR‐200 and expressed less ACTA2 and FN1 than those from mesenchymal tumours. As such, these data provide a new mechanism for the development of fibroblast heterogeneity in CRC, through EV‐mediated transfer of miRNAs, and provide an explanation as to why CRC tumours with greater metastatic potential are CAF rich.
Collapse
Affiliation(s)
- Rahul Bhome
- Cancer Sciences Unit, University of Southampton, Southampton, UK.,University Surgery, University of Southampton, Southampton, UK
| | | | - Victoria James
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
| | - Louise M House
- Cancer Sciences Unit, University of Southampton, Southampton, UK
| | | | | | - Joeri Tulkens
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - John N Primrose
- University Surgery, University of Southampton, Southampton, UK
| | - Gareth J Thomas
- Cancer Sciences Unit, University of Southampton, Southampton, UK
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Alex H Mirnezami
- Cancer Sciences Unit, University of Southampton, Southampton, UK.,University Surgery, University of Southampton, Southampton, UK
| | - A Emre Sayan
- Cancer Sciences Unit, University of Southampton, Southampton, UK
| |
Collapse
|
189
|
Gül D, Schweitzer A, Khamis A, Knauer SK, Ding GB, Freudelsperger L, Karampinis I, Strieth S, Hagemann J, Stauber RH. Impact of Secretion-Active Osteoblast-Specific Factor 2 in Promoting Progression and Metastasis of Head and Neck Cancer. Cancers (Basel) 2022; 14:2337. [PMID: 35565465 PMCID: PMC9106029 DOI: 10.3390/cancers14092337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/28/2022] [Accepted: 05/05/2022] [Indexed: 11/16/2022] Open
Abstract
Treatment success of head and neck cancer (HNC) is still hampered by tumor relapse due to metastases. Our study aimed to identify biomarkers by exploiting transcriptomics profiles of patient-matched metastases, primary tumors, and normal tissue mucosa as well as the TCGA HNC cohort data sets. Analyses identified osteoblast-specific factor 2 (OSF-2) as significantly overexpressed in lymph node metastases and primary tumors compared to normal tissue. High OSF-2 levels correlate with metastatic disease and reduced overall survival of predominantly HPV-negative HNC patients. No significant correlation was observed with tumor localization or therapy response. These findings were supported by the fact that OSF-2 expression was not elevated in cisplatin-resistant HNC cell lines. OSF-2 was strongly expressed in tumor-associated fibroblasts, suggesting a tumor microenvironment-promoting function. Molecular cloning and expression studies of OSF-2 variants from patients identified an evolutionary conserved bona fide protein secretion signal (1MIPFLPMFSLLLLLIVNPINA21). OSF-2 enhanced cell migration and cellular survival under stress conditions, which could be mimicked by the extracellular administration of recombinant protein. Here, OSF-2 executes its functions via ß1 integrin, resulting in the phosphorylation of PI3K and activation of the Akt/PKB signaling pathway. Collectively, we suggest OSF-2 as a potential prognostic biomarker and drug target, promoting metastases by supporting the tumor microenvironment and lymph node metastases survival rather than by enhancing primary tumor proliferation or therapy resistance.
Collapse
Affiliation(s)
- Désirée Gül
- Department of Otorhinolaryngology, Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (A.S.); (A.K.); (L.F.); (J.H.)
| | - Andrea Schweitzer
- Department of Otorhinolaryngology, Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (A.S.); (A.K.); (L.F.); (J.H.)
| | - Aya Khamis
- Department of Otorhinolaryngology, Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (A.S.); (A.K.); (L.F.); (J.H.)
- Oral Pathology Department, Faculty of Dentistry, Alexandria University, El Azareta, Alexandria, Egypt
| | - Shirley K. Knauer
- Institute for Molecular Biology, Centre for Medical Biotechnology (ZMB), University Duisburg-Essen, Universitätsstraße, 45117 Essen, Germany;
| | - Guo-Bin Ding
- Institute of Biotechnology, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China;
| | - Laura Freudelsperger
- Department of Otorhinolaryngology, Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (A.S.); (A.K.); (L.F.); (J.H.)
| | - Ioannis Karampinis
- Academic Thoracic Center, University Medical Center Mainz, Johannes Gutenberg University Mainz, 55131 Mainz, Germany;
| | - Sebastian Strieth
- Department of Otorhinolaryngology, University Medical Center Bonn, 53127 Bonn, Germany;
| | - Jan Hagemann
- Department of Otorhinolaryngology, Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (A.S.); (A.K.); (L.F.); (J.H.)
| | - Roland H. Stauber
- Department of Otorhinolaryngology, Head and Neck Surgery, Molecular and Cellular Oncology, University Medical Center, 55131 Mainz, Germany; (A.S.); (A.K.); (L.F.); (J.H.)
- Institute of Biotechnology, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China;
| |
Collapse
|
190
|
Autophagy and EMT in cancer and metastasis: Who controls whom? Biochim Biophys Acta Mol Basis Dis 2022; 1868:166431. [PMID: 35533903 DOI: 10.1016/j.bbadis.2022.166431] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/10/2022] [Accepted: 05/02/2022] [Indexed: 02/06/2023]
Abstract
Metastasis consists of hallmark events, including Epithelial-Mesenchymal Transition (EMT), angiogenesis, initiation of inflammatory tumor microenvironment, and malfunctions in apoptosis. Autophagy is known to play a pivotal role in the metastatic process. Autophagy has pulled researchers towards it in recent times because of its dual role in the maintenance of cancer cells. Evidence states that cells undergoing EMT need autophagy in order to survive during migration and dissemination. Additionally, it orchestrates EMT markers in certain cancers. On the other side of the coin, autophagy plays an oncosuppressive role in impeding early metastasis. This review aims to project the interrelationship between autophagy and EMT. Targeting EMT via autophagy as a useful strategy is discussed in this review. Furthermore, for the first time, we have covered the possible reciprocating roles of EMT and autophagy and its consequences in cancer metastasis.
Collapse
|
191
|
Ring finger protein 6 enhances chemo-resistance by transcriptionally activating proliferating cell nuclear antigen expression and attenuating DNA damage in lung adenocarcinoma. Cancer Lett 2022; 534:215609. [DOI: 10.1016/j.canlet.2022.215609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/15/2022] [Accepted: 02/24/2022] [Indexed: 11/20/2022]
|
192
|
Dang TT, Lerner M, Saunders D, Smith N, Gulej R, Zalles M, Towner RA, Morales JC. XRN2 Is Required for Cell Motility and Invasion in Glioblastomas. Cells 2022; 11:1481. [PMID: 35563787 PMCID: PMC9100175 DOI: 10.3390/cells11091481] [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: 02/21/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 02/01/2023] Open
Abstract
One of the major obstacles in treating brain cancers, particularly glioblastoma multiforme, is the occurrence of secondary tumor lesions that arise in areas of the brain and are inoperable while obtaining resistance to current therapeutic agents. Thus, gaining a better understanding of the cellular factors that regulate glioblastoma multiforme cellular movement is imperative. In our study, we demonstrate that the 5'-3' exoribonuclease XRN2 is important to the invasive nature of glioblastoma. A loss of XRN2 decreases cellular speed, displacement, and movement through a matrix of established glioblastoma multiforme cell lines. Additionally, a loss of XRN2 abolishes tumor formation in orthotopic mouse xenograft implanted with G55 glioblastoma multiforme cells. One reason for these observations is that loss of XRN2 disrupts the expression profile of several cellular factors that are important for tumor invasion in glioblastoma multiforme cells. Importantly, XRN2 mRNA and protein levels are elevated in glioblastoma multiforme patient samples. Elevation in XRN2 mRNA also correlates with poor overall patient survival. These data demonstrate that XRN2 is an important cellular factor regulating one of the major obstacles in treating glioblastomas and is a potential molecular target that can greatly enhance patient survival.
Collapse
Affiliation(s)
- Tuyen T. Dang
- Department of Neurosurgery, Sttephenson Cancer Center University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA;
| | - Megan Lerner
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA;
| | - Debra Saunders
- Department of Pathology, University of Oklahoma Health Science Center, Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (D.S.); (N.S.); (R.G.); (M.Z.); (R.A.T.)
| | - Nataliya Smith
- Department of Pathology, University of Oklahoma Health Science Center, Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (D.S.); (N.S.); (R.G.); (M.Z.); (R.A.T.)
| | - Rafal Gulej
- Department of Pathology, University of Oklahoma Health Science Center, Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (D.S.); (N.S.); (R.G.); (M.Z.); (R.A.T.)
| | - Michelle Zalles
- Department of Pathology, University of Oklahoma Health Science Center, Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (D.S.); (N.S.); (R.G.); (M.Z.); (R.A.T.)
| | - Rheal A. Towner
- Department of Pathology, University of Oklahoma Health Science Center, Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (D.S.); (N.S.); (R.G.); (M.Z.); (R.A.T.)
| | - Julio C. Morales
- Department of Neurosurgery, Sttephenson Cancer Center University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA;
| |
Collapse
|
193
|
Choi HY, Ahn JH, Kwon H, Yim JH, Lee D, Choi JH. Citromycin Isolated from the Antarctic Marine-Derived Fungi, Sporothrix sp., Inhibits Ovarian Cancer Cell Invasion via Suppression of ERK Signaling. Mar Drugs 2022; 20:md20050275. [PMID: 35621926 PMCID: PMC9143255 DOI: 10.3390/md20050275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/18/2022] [Accepted: 04/18/2022] [Indexed: 02/01/2023] Open
Abstract
Recently, microorganisms and their metabolites in the Antarctic marine environment have attracted attention as useful sources for novel therapeutics, including anticancer drugs. Here, we investigated the effects of citromycin, isolated from the Antarctic marine-derived fungus, Sporothrix sp., on human ovarian cancer cells. Citromycin inhibited the migration and invasion of human ovarian cancer SKOV3 and A2780 cells, but had no cytotoxic activity against them. Additionally, it inhibited the expression of epithelial–mesenchymal transition (EMT) markers and the activation of matrix metalloproteinase (MMP)-2 and MMP9. Moreover, extracellular signal-regulated kinase (ERK)-1/2 signaling was inhibited after citromycin treatment, and the ectopic expression of ERK negated the anti-invasive activity of citromycin. Our findings suggest that citromycin inhibits the migration and invasion of human ovarian cancer cells by downregulating the expression levels of EMT markers and MMP-2/9 via inhibition of the ERK1/2 pathway.
Collapse
Affiliation(s)
- He Yun Choi
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul 02447, Korea;
| | - Ji-Hye Ahn
- Department of Oriental Pharmacy, Woosuk University, Jeonju 55338, Korea;
| | - Haeun Kwon
- Department of Plant Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea; (H.K.); (D.L.)
| | - Joung Han Yim
- Korea Polar Research Institute, Korea Ocean Research and Development Institute, Incheon 21990, Korea;
| | - Dongho Lee
- Department of Plant Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea; (H.K.); (D.L.)
| | - Jung-Hye Choi
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul 02447, Korea;
- College of Pharmacy, Kyung Hee University, Seoul 02447, Korea
- Correspondence:
| |
Collapse
|
194
|
Nam MK, Moon JM, Kim GY, Kim SM, Rhim H. The novel human HtrA2 ortholog in zebrafish: New molecular insight and challenges into the imbalance of homeostasis. Gene 2022; 819:146263. [PMID: 35121025 DOI: 10.1016/j.gene.2022.146263] [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: 08/27/2021] [Revised: 01/17/2022] [Accepted: 01/27/2022] [Indexed: 11/28/2022]
Abstract
High temperature requirement A2 (HtrA2) contributes to regulating mitochondrial quality control and maintaining the balance between the death and survival of cells and living organisms. However, the molecular mechanism of HtrA2 in physiological and pathophysiological processes remains unclear. HtrA2 exhibits multifaceted characteristics according to the expression levels and acts opposite functions depending on its subcellular localization. Thus, innovative technologies and systems that can be freely manipulated at the quantitative, biochemical, molecular and cellular levels are needed to address not only the challenges faced by HtrA2 research but also the general obstacles to protein research. Here, we are the first to identify zebrafish HtrA2 (zHtrA2) as the true ortholog of human HtrA2 (hHtrA2), by in silico sequence analysis of genomic DNA and molecular biological techniques, which is highly conserved structurally and functionally as a serine protease and cell death regulator. The zHtrA2 protein is primarily localized in the mitochondria, where alanine-exposed mature zHtrA2 ((A)-zHtrA2) is generated by removing 111 residues at the N-terminus of pro-zHtrA2. The (A)-zHtrA2 released from the mitochondria into the cytosol induces the caspase cascade by binding to and inhibiting hXIAP, a cognate partner of hHtrA2. Notably, zHtrA2 has well conserved properties of serine protease that specifically cleaves hParkin, a cognate substrate of hHtrA2. Interestingly, cytosolic (M)-zHtrA2, which does not bind hXIAP, induces atypical cell death in a serine protease-dependent manner, as occurs in hHtrA2. Thus, the zebrafish-zHtrA2 system can be used to clarify the crucial role of HtrA2 in maintaining the survival of living organisms and provide an opportunity to develop novel therapeutics for HtrA2-associated diseases, such as neurodegenerative diseases and cancer, which are caused by dysregulation of HtrA2.
Collapse
Affiliation(s)
- Min-Kyung Nam
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seocho-gu, Seoul 06591, Republic of Korea.
| | - Jeong-Mi Moon
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seocho-gu, Seoul 06591, Republic of Korea
| | - Goo-Young Kim
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seocho-gu, Seoul 06591, Republic of Korea
| | - Sung Min Kim
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seocho-gu, Seoul 06591, Republic of Korea
| | - Hyangshuk Rhim
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seocho-gu, Seoul 06591, Republic of Korea; Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seocho-gu, Seoul 06591, Republic of Korea.
| |
Collapse
|
195
|
Ma L. From Photon Beam to Accelerated Particle Beam: Antimetastasis Effect of Combining Radiotherapy With Immunotherapy. Front Public Health 2022; 10:847119. [PMID: 35425754 PMCID: PMC9002008 DOI: 10.3389/fpubh.2022.847119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/25/2022] [Indexed: 12/18/2022] Open
Abstract
Cancer is one of the major diseases that seriously threaten the human health. Radiotherapy is a common treatment for cancer. It is noninvasive and retains the functions of the organ where the tumor is located. Radiotherapy includes photon beam radiotherapy, which uses X-rays or gamma rays, and particle beam radiotherapy, using beams of protons and heavy ions. Compared with photon beam radiotherapy, particle beam radiotherapy has excellent dose distribution, which enables it to kill the primary tumor cells more effectively and simultaneously minimize the radiation-induced damage to normal tissues and organs surrounding the tumor. Despite the excellent therapeutic effect of particle beam radiotherapy on the irradiated tumors, it is not an effective treatment for metastatic cancers. Therefore, developing novel and effective treatment strategies for cancer is urgently needed to save patients with distant cancer metastasis. Immunotherapy enhances the body's own immune system to fight cancer by activating the immune cells, and consequently, to achieve the systemic anticancer effects, and it is considered to be an adjuvant therapy that can enhance the efficacy of particle beam radiotherapy. This review highlights the research progress of the antimetastasis effect and the mechanism of the photon beam or particle beam radiotherapy combined with immunotherapy and predicts the development prospects of this research area.
Collapse
Affiliation(s)
- Liqiu Ma
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing, China.,National Innovation Center of Radiation Application, Beijing, China
| |
Collapse
|
196
|
Hanalis-Miller T, Nudelman G, Ben-Eliyahu S, Jacoby R. The Effect of Pre-operative Psychological Interventions on Psychological, Physiological, and Immunological Indices in Oncology Patients: A Scoping Review. Front Psychol 2022; 13:839065. [PMID: 35572335 PMCID: PMC9094613 DOI: 10.3389/fpsyg.2022.839065] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/22/2022] [Indexed: 11/19/2022] Open
Abstract
Introduction The stressful pre-operative period exerts a profound impact on psychological, physiological and immunological outcomes. Oncological surgeries, in particular, elicit significantly higher stress responses than most other surgeries. Managing these responses through psychological interventions may improve long-term outcomes. The purpose of the current research was to review studies that have explored pre-operative psychological interventions in cancer patients in order to map the types of current interventions and provide an initial assessment of whether these interventions improved psychological, physiological, and/or immunological indices as well as long-term cancer outcomes. Methods A systematic literature search for studies that included pre-operative psychological interventions in oncology patients was conducted, using the databases PubMed and Web of Science. Inclusion criteria included studies pertaining to oncological surgery in adults, study designs that included a clearly defined pre-operative psychological intervention and control group. Results We found 44 studies, each using one of the following interventions: psychoeducation, cognitive interventions, relaxation techniques, integrated approaches. All the studies reported improved immediate post-operative psychological, physiological, and/or immunological outcomes. Only a few studies addressed long-term cancer outcomes, and only one reported improved survival. Conclusions Research on pre-operative interventions with cancer patients is missing systematic methods. Studies provide varying results, which makes it difficult to compare them and reach reliable conclusions. There is considerable heterogeneity in the literature regarding the specific intervention used, the timing of intervention, the characteristics of the patients studied and the outcome measures. In order to improve research in this field, including the measurement of long-term outcomes, we suggest some steps that should be taken in further research.
Collapse
Affiliation(s)
| | - Gabriel Nudelman
- School of Behavioral Sciences, The Academic College of Tel Aviv-Yaffo, Tel Aviv-Yafo, Israel
| | - Shamgar Ben-Eliyahu
- Sagol School of Neuroscience and School of Psychological Sciences, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Rebecca Jacoby
- Stress, Hope and Cope Laboratory, School of Behavioral Sciences, The Academic College of Tel Aviv-Yaffo, Tel Aviv-Yafo, Israel
| |
Collapse
|
197
|
In Vitro Angiogenesis Inhibition and Endothelial Cell Growth and Morphology. Int J Mol Sci 2022; 23:ijms23084277. [PMID: 35457095 PMCID: PMC9025250 DOI: 10.3390/ijms23084277] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/04/2022] [Accepted: 04/08/2022] [Indexed: 02/05/2023] Open
Abstract
A co-culture assay with human umbilical vein endothelial cells (HUVECs) and normal human dermal fibroblasts (NHDFs) was used to study whether selected angiogenesis inhibitors were able to inhibit differentiation and network formation of HUVECs in vitro. The effect of the inhibitors was determined by the morphology and the calculated percentage area covered by HUVECs. Neutralizing VEGF with avastin and polyclonal goat anti-VEGF antibody and inhibiting VEGFR2 with sorafenib and vatalanib resulted in the formation of HUVEC clusters of variable sizes as a result of inhibited EC differentiation. Furthermore, numerous inhibitors of the VEGF signaling pathways were tested for their effect on the growth and differentiation of HUVECs. The effects of these inhibitors did not reveal a cluster morphology, either individually or when combined to block VEGFR2 downstream pathways. Only the addition of N-methyl-p-bromolevamisole revealed a similar morphology as when targeting VEGF and VEGFR2, meaning it may have an inhibitory influence directly on VEGFR signaling. Additionally, several nuclear receptor ligands and miscellaneous compounds that might affect EC growth and differentiation were tested, but only dexamethasone gave rise to cluster formation similarly to VEGF-neutralizing compounds. These results point to a link between angiogenesis, HUVEC differentiation and glucocorticoid receptor activation.
Collapse
|
198
|
Wu Q, Tsai HI, Zhu H, Wang D. The Entanglement between Mitochondrial DNA and Tumor Metastasis. Cancers (Basel) 2022; 14:cancers14081862. [PMID: 35454769 PMCID: PMC9028275 DOI: 10.3390/cancers14081862] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Mitochondrial dysfunction is one of the main features of cancer cells. As genetic material in mitochondria, mitochondrial DNA (mtDNA) variations and dysregulation of mitochondria-encoded genes have been shown to correlate with survival outcomes in cancer patients. Cancer metastasis is often a major cause of treatment failure, which is a multi-step cascade process. With the development of gene sequencing and in vivo modeling technology, the role of mtDNA in cancer metastasis has been continuously explored. Our review systematically provides a summary of the multiple roles of mtDNA in cancer metastasis and presents the broad prospects for mtDNA in cancer prediction and therapy. Abstract Mitochondrial DNA, the genetic material in mitochondria, encodes essential oxidative phosphorylation proteins and plays an important role in mitochondrial respiration and energy transfer. With the development of genome sequencing and the emergence of novel in vivo modeling techniques, the role of mtDNA in cancer biology is gaining more attention. Abnormalities of mtDNA result in not only mitochondrial dysfunction of the the cancer cells and malignant behaviors, but regulation of the tumor microenvironment, which becomes more aggressive. Here, we review the recent progress in the regulation of cancer metastasis using mtDNA and the underlying mechanisms, which may identify opportunities for finding novel cancer prediction and therapeutic targets.
Collapse
Affiliation(s)
- Qiwei Wu
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
| | - Hsiang-i Tsai
- Laboratory of Radiology, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
| | - Haitao Zhu
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
- Laboratory of Radiology, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
- Correspondence: (H.Z.); (D.W.); Tel.: +86-138-6139-0259 (D.W.)
| | - Dongqing Wang
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
- Correspondence: (H.Z.); (D.W.); Tel.: +86-138-6139-0259 (D.W.)
| |
Collapse
|
199
|
Luo Z, Lu L, Xu W, Meng N, Wu S, Zhou J, Xu Q, Xie C, Liu Y, Lu W. In vivo self-assembled drug nanocrystals for metastatic breast cancer all-stage targeted therapy. J Control Release 2022; 346:32-42. [PMID: 35378211 DOI: 10.1016/j.jconrel.2022.03.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/11/2022] [Accepted: 03/29/2022] [Indexed: 12/22/2022]
Abstract
Chemotherapy is still the mainstay treatment for metastatic triple-negative breast cancers (TNBC) currently in clinical practice. The unmet needs of chemotherapy for metastatic TNBC are mainly from the insufficient drug delivery and unavailable targeting strategy that thwart the whole progression of metastatic TNBC. The in vivo ligands-mediated active targeting efficiency is usually affected by protein corona. While, the protein corona-bridged natural targeting, in turn, provides a new way for specific drug delivery. Herein, we develop a novel metastatic progression-oriented in vivo self-assembled Cabazitaxel nanocrystals (CNC) delivery system (PC/CNC) through the CNC automatically absorbing functional plasma proteins (transferrin, apolipoprotein A-IV and apolipoprotein E) in vivo, aiming to achieve the simultaneously targeted delivery to primary tumors, circulating tumor cells and metastatic lesions. With the unique advantages of superhigh drug-loading and protein corona empowered active targeting properties to tumor cells, HUVECs, active-platelets and blood-brain barrier/blood-tumor barrier, the PC/CNC exhibits a significantly improved therapeutic effect in metastatic TNBC therapy compared with free drug and CNC-loaded liposomes.
Collapse
Affiliation(s)
- Zimiao Luo
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education and PLA), State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Institutes of Integrative Medicine of Fudan University, Shanghai 200041, China
| | - Weixia Xu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education and PLA), State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Nana Meng
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education and PLA), State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Sunyi Wu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education and PLA), State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Jianfen Zhou
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education and PLA), State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Qianzhu Xu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education and PLA), State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Department of Integrative Medicine, Huashan Hospital, Institutes of Integrative Medicine of Fudan University, Shanghai 200041, China
| | - Cao Xie
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education and PLA), State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yu Liu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education and PLA), State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education and PLA), State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Department of Integrative Medicine, Huashan Hospital, Institutes of Integrative Medicine of Fudan University, Shanghai 200041, China; Minhang Branch, Zhongshan Hospital and Institute of Fudan-Minghang Academic Health System, Minghang Hospital, Fudan University, Shanghai 201199, China; Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Center for Druggability of Cardiovascular non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| |
Collapse
|
200
|
PLUS: Predicting cancer metastasis potential based on positive and unlabeled learning. PLoS Comput Biol 2022; 18:e1009956. [PMID: 35349572 PMCID: PMC8992993 DOI: 10.1371/journal.pcbi.1009956] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 04/08/2022] [Accepted: 02/23/2022] [Indexed: 11/19/2022] Open
Abstract
Metastatic cancer accounts for over 90% of all cancer deaths, and evaluations of metastasis potential are vital for minimizing the metastasis-associated mortality and achieving optimal clinical decision-making. Computational assessment of metastasis potential based on large-scale transcriptomic cancer data is challenging because metastasis events are not always clinically detectable. The under-diagnosis of metastasis events results in biased classification labels, and classification tools using biased labels may lead to inaccurate estimations of metastasis potential. This issue is further complicated by the unknown metastasis prevalence at the population level, the small number of confirmed metastasis cases, and the high dimensionality of the candidate molecular features. Our proposed algorithm, called Positive and unlabeled Learning from Unbalanced cases and Sparse structures (PLUS), is the first to use a positive and unlabeled learning framework to account for the under-detection of metastasis events in building a classifier. PLUS is specifically tailored for studying metastasis that deals with the unbalanced instance allocation as well as unknown metastasis prevalence, which are not considered by other methods. PLUS achieves superior performance on synthetic datasets compared with other state-of-the-art methods. Application of PLUS to The Cancer Genome Atlas Pan-Cancer gene expression data generated metastasis potential predictions that show good agreement with the clinical follow-up data, in addition to predictive genes that have been validated by independent single-cell RNA-sequencing datasets. Metastasis is the major cause of cancer-related deaths, and evaluations of metastasis risk are essential for tailored treatment of cancer patients. Existing methods often build a classifier using the clinical metastasis diagnoses as binary responses or detect genomic features significantly associated with metastasis-related survival outcomes. However, these methods tend to identify genomic predictors that have little consistency across different cancer types. Thus, there is an urgent need for a powerful tool to characterize the cancer metastasis potential applicable across a wide span of cancer types. Computational assessment of metastasis potential based on large-scale transcriptomic cancer data is challenging because metastasis events are not always clinically detectable, which results in biased estimations of metastasis potential. Our proposed algorithm, called PLUS, considers patients with metastasis diagnosis as positive instances and the remainder as unlabeled instances, meaning they are either metastatic or non-metastatic. Such a classifier given by PLUS rendered concordance between the predicted cancer metastasis and observed metastasis survival outcomes in the follow-up data for almost all cancer types considered. The selected genes were found to perform functions consistent with experimental research findings and are capable of clustering the single cells based on their levels of metastasis potential.
Collapse
|