1
|
Mun SK, Sim HB, Han JY, Kim H, Park DH, Chang DJ, Yee ST, Chang YT, Kim JJ. Visualization of Metastatic Lung Cancer with TiNIR. Tomography 2023; 9:1187-1195. [PMID: 37489464 PMCID: PMC10366764 DOI: 10.3390/tomography9040096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 07/26/2023] Open
Abstract
The development of efficient biomarkers and probes for monitoring and treating cancer, specifically metastatic cancer, is a critical research area that can have a significant impact on both patient outcomes and drug discovery. In this context, TiNIR has been developed to detect tumor-initiating cells (TICs), with heme oxygenase 2 (HO2) as a promising therapeutic biomarker for tumor-initiating cells. In this study, TiNIR has demonstrated its effectiveness as an in vivo metastatic lung cancer tracker, highlighting its potential as a valuable tool in cancer research and therapy. The development of innovative approaches that selectively target metastatic cancers represents a promising avenue for improving survival rates and enhancing the quality of life of cancer patients.
Collapse
Affiliation(s)
- Seul-Ki Mun
- Department of Biomedical Science, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Republic of Korea
- Department of Pharmacy, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Republic of Korea
| | - Hyun Bo Sim
- Department of Biomedical Science, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Republic of Korea
| | - Ji Yeon Han
- Department of Biomedical Science, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Republic of Korea
| | - Hyeongyeong Kim
- Department of Biomedical Science, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Republic of Korea
| | - Dae-Han Park
- Department of Biomedical Science, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Republic of Korea
| | - Dong-Jo Chang
- Department of Pharmacy, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Republic of Korea
| | - Sung-Tae Yee
- Department of Pharmacy, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Republic of Korea
| | - Young-Tae Chang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Jong-Jin Kim
- Department of Biomedical Science, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Republic of Korea
| |
Collapse
|
2
|
MicroRNA expression is deregulated by aberrant methylation in B-cell acute lymphoblastic leukemia mouse model. Mol Biol Rep 2022; 49:1731-1739. [PMID: 35001247 DOI: 10.1007/s11033-021-06982-x] [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: 07/28/2021] [Accepted: 11/17/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND The expression of microRNAs (miRNAs) in the serum of B-cell acute lymphoblastic leukemia (B-ALL) patients is abnormal. Nevertheless, the underlying mechanism remains unclear. Recent studies indicate that the methylation state of circulating cell-free DNA (cfDNA) is different between cancer patients and healthy individuals. Therefore, we speculate that abnormal expression of miRNA may be associated with cfDNA methylation. METHODS A green fluorescent protein (GFP) labeled B-ALL transplantation animal model was established to explore the relationship between the miRNA expression and cfDNA methylation of the related gene. Quantitative real-time PCR (qRT-PCR) was used to detect the expression levels of miRNAs. Further, cfDNA methylation levels of the related genes were evaluated through bisulfite sequencing polymerase chain reaction (BSP). RESULTS The expression levels of miR-196b, miR-203, miR-34a-5p, miR-335-3p, miR-34b-5p, miR-615, miR-375-3p and miR-193b-5p in the serum of the model mice were significantly lower than those of the control group (P < 0.05). The methylation level of miR-196b promoter in cfDNA of the model group was significantly lower than that of the control group (P < 0.05), whereas no significant difference was noted in miR-203 promoter. The methylation levels of miR-196b and miR-203 coding region in cfDNA of the model group were significantly higher than those of the control group (P < 0.05). CONCLUSIONS These results showed that CpG island hypermethylation in the miRNA coding region of cfDNA is related to the low expression of miR-196b and miR-203.
Collapse
|
3
|
Jug-PLGA-NPs, a New Form of Juglone with Enhanced Efficiency and Reduced Toxicity on Melanoma. Chin J Integr Med 2021; 28:909-917. [PMID: 34913148 DOI: 10.1007/s11655-021-3461-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To verrify the anti-tumor efficacy and toxicity between juglone (Jug) and Jug-loaded PLGA nanoparticles (Jug-PLGA-NPs). METHODS Jug-PLGA-NPs were prepared by ultrasonic emulsification. The anti-tumor activity of Jug (2, 3, 4 µg/mL) and Jug-PLGA-NPs (Jug: 2, 3, 4 µg/mL) in vitro was measured by MTT assay and cell apoptosis analysis. The distribution, anti-tumor effect and biological safety in vivo was evaluated on A375 nude mice. RESULTS With the advantage of good penetration and targeting properties, Jug-PLGA-NPs significantly inhibited proliferation and migration of melanoma cells both in vitro and in vivo (P<0.05 or P<0.01) with acceptable biocompatibility. CONCLUSIONS Jug can inhibit the growth of melanoma but is highly toxic. With the advantage of sustained release, tumor targeting, anti-tumor activity and acceptable biological safety, Jug-PLGA-NPs provide a new pharmaceutical form for future application of Jug.
Collapse
|
4
|
Nikovics K, Favier AL. Macrophage Identification In Situ. Biomedicines 2021; 9:1393. [PMID: 34680510 PMCID: PMC8533306 DOI: 10.3390/biomedicines9101393] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022] Open
Abstract
Understanding the processes of inflammation and tissue regeneration after injury is of great importance. For a long time, macrophages have been known to play a central role during different stages of inflammation and tissue regeneration. However, the molecular and cellular mechanisms by which they exert their effects are as yet mostly unknown. While in vitro macrophages have been characterized, recent progress in macrophage biology studies revealed that macrophages in vivo exhibited distinctive features. Actually, the precise characterization of the macrophages in vivo is essential to develop new healing treatments and can be approached via in situ analyses. Nowadays, the characterization of macrophages in situ has improved significantly using antigen surface markers and cytokine secretion identification resulting in specific patterns. This review aims for a comprehensive overview of different tools used for in situ macrophage identification, reporter genes, immunolabeling and in situ hybridization, discussing their advantages and limitations.
Collapse
Affiliation(s)
- Krisztina Nikovics
- Imagery Unit, Department of Platforms and Technology Research, French Armed Forces Biomedical Research Institute, 91223 Brétigny-sur-Orge, France;
| | | |
Collapse
|
5
|
Wang R, Lewis MS, Lyu J, Zhau HE, Pandol SJ, Chung LWK. Cancer-stromal cell fusion as revealed by fluorescence protein tracking. Prostate 2020; 80:274-283. [PMID: 31846114 PMCID: PMC6949378 DOI: 10.1002/pros.23941] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 12/06/2019] [Indexed: 12/29/2022]
Abstract
PURPOSE We previously determined that cancer-stromal interaction was a direct route to tumor cell heterogeneity progression, since cancer-stromal cell fusion in coculture resulted in the creation of heterogeneous clones of fusion hybrid progeny. In this report, we modified the cancer-stromal coculture system to establish optimal experimental conditions for investigating cell fusion machinery and the mechanism of heterogeneity progression. EXPERIMENTAL DESIGN Red fluorescence protein-tagged LNCaP cells were cocultured with green fluorescence protein-labeled prostate stromal cells for cancer-stromal cell fusion, which was tracked as dual fluorescent cells by fluorescence microscopy. RESULTS We identified the most efficient strategy to isolate clones of fusion hybrid progenies. From the coculture, mixed cells including fusion hybrids were subjected to low-density replating for colony formation by fusion hybrid progeny. These colonies could propagate into derivative cell populations. Compared to the parental LNCaP cells, clones of the fusion hybrid progeny displayed divergent behaviors and exhibited permanent genomic hybridization. CONCLUSIONS Cancer-stromal cell fusion leads to cancer cell heterogeneity. The cancer-stromal coculture system characterized in this study can be used as a model for molecular characterization of cancer cell fusion as the mechanism behind the progression of heterogeneity observed in clinical prostate cancers.
Collapse
Affiliation(s)
- Ruoxiang Wang
- Uro-Oncology Research, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Pathology, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Michael S. Lewis
- Uro-Oncology Research, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Pathology, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Ji Lyu
- Uro-Oncology Research, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Haiyen E. Zhau
- Uro-Oncology Research, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Stephen J. Pandol
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Pathology, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | | |
Collapse
|
6
|
Okada S, Vaeteewoottacharn K, Kariya R. Application of Highly Immunocompromised Mice for the Establishment of Patient-Derived Xenograft (PDX) Models. Cells 2019; 8:889. [PMID: 31412684 PMCID: PMC6721637 DOI: 10.3390/cells8080889] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/09/2019] [Accepted: 08/09/2019] [Indexed: 12/11/2022] Open
Abstract
Patient-derived xenograft (PDX) models are created by engraftment of patient tumor tissues into immunocompetent mice. Since a PDX model retains the characteristics of the primary patient tumor including gene expression profiles and drug responses, it has become the most reliable in vivo human cancer model. The engraftment rate increases with the introduction of Non-obese diabetic Severe combined immunodeficiency (NOD/SCID)-based immunocompromised mice, especially the NK-deficient NOD strains NOD/SCID/interleukin-2 receptor gamma chain(IL2Rγ)null (NOG/NSG) and NOD/SCID/Jak3(Janus kinase 3)null (NOJ). Success rates differ with tumor origin: gastrointestinal tumors acquire a higher engraftment rate, while the rate is lower for breast cancers. Subcutaneous transplantation is the most popular method to establish PDX, but some tumors require specific environments, e.g., orthotropic or renal capsule transplantation. Human hormone treatment is necessary to establish hormone-dependent cancers such as prostate and breast cancers. PDX mice with human hematopoietic and immune systems (humanized PDX) are powerful tools for the analysis of tumor-immune system interaction and evaluation of immunotherapy response. A PDX biobank equipped with patients' clinical data, gene-expression patterns, mutational statuses, tumor tissue architects, and drug responsiveness will be an authoritative resource for developing specific tumor biomarkers for chemotherapeutic predictions, creating individualized therapy, and establishing precise cancer medicine.
Collapse
Affiliation(s)
- Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-0811, Japan.
- Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan.
| | - Kulthida Vaeteewoottacharn
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-0811, Japan
- Department of Biochemistry, Khon Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Ryusho Kariya
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
7
|
Mouse-Derived Allografts: A Complementary Model to the KPC Mice on Researching Pancreatic Cancer In Vivo. Comput Struct Biotechnol J 2019; 17:498-506. [PMID: 31011408 PMCID: PMC6462783 DOI: 10.1016/j.csbj.2019.03.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 03/29/2019] [Accepted: 03/30/2019] [Indexed: 12/31/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant cancers and has an extremely undesirable prognosis because little is known about the initiation and progression mechanisms of pancreatic cancer. The lack of an appropriate research model may have hindered this process. Using LSL-KrasG12D/+; Trp53fl/+; Pdx1-Cre (KPC) mice and the tumor tissue fragment transplantation technique, we constructed the mouse-derived subcutaneous/orthotopic allograft tumor models (MDAs-ST/OT). H&E staining, Masson staining and immunohistochemical staining were adopted to describe the histopathology and biomarkers of the MDAs and the recruitment of immune cells. The intervention of gemcitabine was applied to measure the chemotherapeutic response of MDAs tumors. MDAs could mimic the pathological histology and the high proliferation characteristics of PDAC. Indeed, the fibrosis, epithelial-mesenchyme transition (EMT) and invasion/metastasis related markers of MDAs were similar to those observed in pancreatic cancer. Further, the recruitment of immune cells in PDAC was precisely simulated by MDAs. In addition, gemcitabine suppressed the tumor growth of MDAs-ST significantly. MDAs are an effective model for investigating the progression and treatment of pancreatic cancer.
Collapse
Key Words
- ADM, Acinar to ductal metaplasia (ADM)
- CAFs, Cancer-associated fibroblasts
- EMT, Epithelial-mesenchyme transition
- GEMMs, Genetically engineered mouse models
- Gemcitabine
- KC, LSL-KrasG12D/+, Pdx1-Cre
- KPC, LSL-KrasG12D/+, Trp53fl/+, Pdx1-Cre
- MDAs, Mouse-derived allografts
- MDAs-OT, Mouse-derived orthotopic allograft tumor models
- MDAs-ST, Mouse-derived subcutaneous allograft tumor models
- MDSCs, Myeloid-derived suppressor cells
- Mouse-derived allografts
- PDAC, Pancreatic ductal adenocarcinoma
- PDXs, Patient-derived xenografts
- PanINs, Pancreatic intraepithelial neoplasias
- Pancreatic ductal adenocarcinoma
- Tregs, T regulatory cells
- Tumor tissue fragment
Collapse
|
8
|
Zhang W, Qian S, Yang G, Zhu L, Zhou B, Qu X, Yan Z, Liu R, Wang J. Establishment and characterization of McA-RH7777 cells using virus-mediated stable overexpression of enhanced green fluorescent protein. Exp Ther Med 2018; 16:3149-3154. [PMID: 30250518 DOI: 10.3892/etm.2018.6580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/29/2018] [Indexed: 12/31/2022] Open
Abstract
Hepatocellular carcinoma (HCC), the most common primary tumor of the liver, has a poor prognosis, rapid progression. The aim of the current study was to establish a stable lentiviral expression vector for enhanced green fluorescent protein (EGFP) and to evaluate biological characteristics on HCC growth and migration following transfection of HCC cells with EGFP. McA-RH7777 cells were transfected with EGFP overexpression lentiviral vector. Cell activity and mobility were monitored with a Cell-IQ Analyzer. Transwell assays were performed to detect invasiveness and flow cytometry was performed for cell cycle analysis. A subcutaneous tumor rat model was established to analyze the stability of fluorescent protein expression. The result suggested no significant differences between wild-type and EGFP-overexpressing McA-RH7777 cells with regards to cell proliferation, activity, mobility, invasiveness and cell cycle. Green fluorescence was detected over 108 days of culturing. The subcutaneous tumor rat model demonstrated that EGFP expression had no influence on tumor growth and long-term expression was stable. The stable EGFP expression of the HCC transplanted tumor rat model may share biological characteristics with human liver cancer. The model established in the current study may be suitable for various applications, including research focusing on liver cancer metastasis and recurrence, interventional therapy, imaging diagnosis and drug screenings.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Intervention Radiology, Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Sheng Qian
- Department of Intervention Radiology, Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Guowei Yang
- Department of Intervention Radiology, Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Liang Zhu
- Department of Intervention Radiology, Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Bo Zhou
- Department of Intervention Radiology, Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Xudong Qu
- Department of Intervention Radiology, Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Zhiping Yan
- Department of Intervention Radiology, Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Rong Liu
- Department of Intervention Radiology, Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| | - Jianhua Wang
- Department of Intervention Radiology, Zhongshan Hospital of Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
9
|
Ye X, Luke BT, Wei BR, Kaczmarczyk JA, Loncarek J, Dwyer JE, Johann DJ, Saul RG, Nissley DV, McCormick F, Whiteley GR, Blonder J. Direct molecular dissection of tumor parenchyma from tumor stroma in tumor xenograft using mass spectrometry-based glycoproteomics. Oncotarget 2018; 9:26431-26452. [PMID: 29899869 PMCID: PMC5995176 DOI: 10.18632/oncotarget.25449] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 05/02/2018] [Indexed: 12/18/2022] Open
Abstract
The most widely used cancer animal model is the human-murine tumor xenograft. Unbiased molecular dissection of tumor parenchyma versus stroma in human-murine xenografts is critical for elucidating dysregulated protein networks/pathways and developing therapeutics that may target these two functionally codependent compartments. Although antibody-reliant technologies (e.g., immunohistochemistry, imaging mass cytometry) are capable of distinguishing tumor-proper versus stromal proteins, the breadth or extent of targets is limited. Here, we report an antibody-free targeted cross-species glycoproteomic (TCSG) approach that enables direct dissection of human tumor parenchyma from murine tumor stroma at the molecular/protein level in tumor xenografts at a selectivity rate presently unattainable by other means. This approach was used to segment/dissect and obtain the protein complement phenotype of the tumor stroma and parenchyma of the metastatic human lung adenocarcinoma A549 xenograft, with no need for tissue microdissection prior to mass-spectrometry analysis. An extensive molecular map of the tumor proper and the associated microenvironment was generated along with the top functional N-glycosylated protein networks enriched in each compartment. Importantly, immunohistochemistry-based cross-validation of selected parenchymal and stromal targets applied on human tissue samples of lung adenocarcinoma and normal adjacent tissue is indicative of a noteworthy translational capacity for this unique approach that may facilitate identifications of novel targets for next generation antibody therapies and development of real time preclinical tumor models.
Collapse
Affiliation(s)
- Xiaoying Ye
- National Cancer Institute RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Brian T. Luke
- Advanced Biomedical Computing Center, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Bih-Rong Wei
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Jan A. Kaczmarczyk
- Cancer Research Technology Program, Antibody Characterization Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Jadranka Loncarek
- Laboratory of Protein Dynamics and Signaling, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Jennifer E. Dwyer
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Donald J. Johann
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72209, USA
| | - Richard G. Saul
- Cancer Research Technology Program, Antibody Characterization Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Dwight V. Nissley
- National Cancer Institute RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Frank McCormick
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA
| | - Gordon R. Whiteley
- National Cancer Institute RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Josip Blonder
- National Cancer Institute RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| |
Collapse
|
10
|
Yu M, Ohmiya Y, Naumov P, Liu YJ. Theoretical Insight into the Emission Properties of the Luciferin and Oxyluciferin of Latia. Photochem Photobiol 2018; 94:540-544. [PMID: 29253310 DOI: 10.1111/php.12876] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 11/27/2017] [Indexed: 11/27/2022]
Abstract
Latia neritoides is a small limpet-like snail that produces a bright green bioluminescence (BL) via a unique light-emitting system. The process, mechanism, and even light emitter of its light emission remain unknown, although this BL has been known for decades. Unlike the other BL systems, neither the luciferin (Luc) nor the oxyluciferin (OxyLuc) of Latia is fluorescent according to the previous experiments. To help to identify its bioluminophore, we studied the geometrical and electronic structures and absorption and fluorescence spectra of Latia Luc and its six analogs as well as its OxyLuc in the gas phase and in water. The calculated results provide clear evidence of the lack of fluorescence in the Luc and OxyLuc of Latia. For the analogs of Latia Luc, the electron-withdrawing or electron-donating ability of the substituted group affects the fluorescence. The results shed new light on the BL mechanism and will likely aid the understanding of Latia BL.
Collapse
Affiliation(s)
- Mohan Yu
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, China
| | - Yoshihiro Ohmiya
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | | | - Ya-Jun Liu
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, China
| |
Collapse
|
11
|
Murata T, Mekada E, Hoffman RM. Reconstitution of a metastatic-resistant tumor microenvironment with cancer-associated fibroblasts enables metastasis. Cell Cycle 2017; 16:533-535. [PMID: 28103135 PMCID: PMC5384587 DOI: 10.1080/15384101.2017.1281486] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The tumor microenvironment is critical for metastasis to occur. Subcutaneous xenografts of tumors in immunodeficient mice are usually encapsulated and rarely metastasize as opposed to orthotopic tumors which metastasize if the original tumor was metastatic. In the present report, we were able to reconstitute a metastatic tumor microenvironment by subcutaneously co-transplanting a human cervical cancer cell line and human cervical cancer-associated fibroblasts (CAFs), in athymic mice, which resulted in lymph node metastasis in 40% of the animals. In contrast, no metastasis occurred from the cervical cancer without CAFs. These results suggest that CAFs can overcome an anti-metastatic tumor environment and are a potential target to prevent metastasis.
Collapse
Affiliation(s)
- Takuya Murata
- a Dept. of Obstetrics and Gynecology Kawasaki Medical School , Okayama , Japan
| | - Eisuke Mekada
- b Dept. of Cell Biology , Research Institute for Microbial Diseases, Osaka University , Osaka , Japan
| | - Robert M Hoffman
- c AntiCancer, Inc. , San Diego , CA , USA.,d Department of Surgery , University of California San Diego , CA , USA
| |
Collapse
|