601
|
Hui D, Chen J, Jiang Y, Pan Y, Zhang Z, Dong M, Shao C. CD44 +CD24 -/low sphere-forming cells of EBV-associated gastric carcinomas show immunosuppressive effects and induce Tregs partially through production of PGE2. Exp Cell Res 2020; 390:111968. [PMID: 32197932 DOI: 10.1016/j.yexcr.2020.111968] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 03/14/2020] [Accepted: 03/16/2020] [Indexed: 02/06/2023]
Abstract
EBV-associated gastric carcinoma (EBVaGC) is accompanied by massive lymphocyte infiltration, but therapy resistance and tumor progression still occur in patients with EBVaGC. Cancer stem cells (CSCs) are reported to possess immunomodulatory ability that allows them to resist immune-mediated rejection for many tumor types. However, whether and how CSCs in EBVaGC exhibit immunosuppression has not yet been elucidated. We isolated CSC-like sphere-forming cells (SFCs) from EBVaGC cell line SNU-719 using the cancer sphere method. We validated their CSC-associated properties in the expression of the epithelial-mesenchymal transition (EMT)-related genes, the ability to form colonies, and resistance to chemotherapy drug-induced apoptosis and explored their immunomodulatory ability using the coculture system with PBMC (peripheral blood mononuclear cell). These CSC-like SFCs were CD44+CD24-/low and were more tumorigenic than the parental SNU-719 cells in the xenograft mouse model. Remarkably, in the tumor-PBMC co-culturing experiments, these EBVaGC SFCs demonstrated profound immunosuppression by inhibiting the proliferation of PBMCs and T cell activation as well as inducing the generation of regulatory T cells (Tregs). Furthermore, the induction of Tregs was partially dependent on prostaglandin E2 (PGE2) produced from SFCs. Moreover, the presence of high CD44+CD24-/low cells in tumor tissues predicted a decreased disease-free survival in patients with EBVaGC. Our study collectively confirmed the existence and immune resistance of CSCs in EBVaGC and offers new insights into the development of novel anti-EBVaGC strategies by targeting CSCs.
Collapse
Affiliation(s)
- Dayang Hui
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Rd, Guangzhou, 510630, China
| | - Jianning Chen
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Rd, Guangzhou, 510630, China
| | - Ye Jiang
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Rd, Guangzhou, 510630, China
| | - Yuhang Pan
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Rd, Guangzhou, 510630, China
| | - Zhigang Zhang
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Rd, Guangzhou, 510630, China
| | - Min Dong
- Department of Medical Oncology, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Rd, Guangzhou, 510630, China
| | - Chunkui Shao
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Rd, Guangzhou, 510630, China.
| |
Collapse
|
602
|
Rabellino A, Khanna KK. The implication of the SUMOylation pathway in breast cancer pathogenesis and treatment. Crit Rev Biochem Mol Biol 2020; 55:54-70. [PMID: 32183544 DOI: 10.1080/10409238.2020.1738332] [Citation(s) in RCA: 5] [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
Breast cancer is the most commonly diagnosed malignancy in woman worldwide, and is the second most common cause of death in developed countries. The transformation of a normal cell into a malignant derivate requires the acquisition of diverse genomic and proteomic changes, including enzymatic post-translational modifications (PTMs) on key proteins encompassing critical cell signaling events. PTMs occur on proteins after translation, and regulate several aspects of proteins activity, including their localization, activation and turnover. Deregulation of PTMs can potentially lead to tumorigenesis, and several de-regulated PTM pathways contribute to abnormal cell proliferation during breast tumorigenesis. SUMOylation is a PTM that plays a pivotal role in numerous aspects of cell physiology, including cell cycle regulation, protein trafficking and turnover, and DNA damage repair. Consistently with this, the deregulation of the SUMO pathway is observed in different human pathologies, including breast cancer. In this review we will describe the role of SUMOylation in breast tumorigenesis and its implication for breast cancer therapy.
Collapse
Affiliation(s)
- Andrea Rabellino
- QIMR Berghofer Medical Research Institute, Brisbane City, Australia
| | - Kum Kum Khanna
- QIMR Berghofer Medical Research Institute, Brisbane City, Australia
| |
Collapse
|
603
|
On-Chip Synthesis of Hyaluronic Acid-Based Nanoparticles for Selective Inhibition of CD44+ Human Mesenchymal Stem Cell Proliferation. Pharmaceutics 2020; 12:pharmaceutics12030260. [PMID: 32183027 PMCID: PMC7151101 DOI: 10.3390/pharmaceutics12030260] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/05/2020] [Accepted: 03/11/2020] [Indexed: 12/13/2022] Open
Abstract
In this study, an innovative microfluidics-based method was developed for one-step synthesis of hyaluronic acid (HA)-based nanoparticles (NPs), by exploiting polyelectrolytic interactions between HA and chitosan (CS), in order to improve reliability, reproducibility and possible scale-up of the NPs preparation. The on-chip synthesis, using a staggered herringbone micromixer, allowed to produce HA/CS NPs with tailored-made size and suitable for both parenteral (117.50 ± 4.51 nm) and loco-regional (349.15 ± 38.09 nm) administration, mainly composed by HA (more than 85% wt) with high negative surface charge (< −20 mV). HA/CS NPs were successfully loaded with a challenging water-insoluble molecule, Everolimus (EVE), an FDA- and EMA-approved anticancer drug able to lead to cell cycle arrest, reduced angiogenesis and promotion of apoptosis. HA/CS NPs resulted to be massively internalized in CD44+ human mesenchymal stem cells via CD44 receptor-mediated endocytosis. HA/CS NPs selectiveness towards CD44 was highlighted by blocking CD44 receptor by anti-CD44 primary antibody and by comparison to CS-based NPs cellular uptake. Eventually, high effectiveness in inhibiting cell proliferation was demonstrated on-chip synthetized EVE loaded HA/CS NPs by tracking in vitro DNA synthesis.
Collapse
|
604
|
Koyama S, Tsuchiya H, Amisaki M, Sakaguchi H, Honjo S, Fujiwara Y, Shiota G. NEAT1 is Required for the Expression of the Liver Cancer Stem Cell Marker CD44. Int J Mol Sci 2020; 21:ijms21061927. [PMID: 32168951 PMCID: PMC7139689 DOI: 10.3390/ijms21061927] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/14/2020] [Accepted: 03/10/2020] [Indexed: 12/18/2022] Open
Abstract
CD44, a cancer stem cell (CSC) marker, is required for maintaining CSC properties in hepatocellular carcinoma (HCC). Nuclear enriched abundant transcript 1 (NEAT1), a long noncoding RNA (lncRNA), is an oncogenic driver in HCC. In the present study, we investigated the significance of the NEAT1 gene in association with CD44 expression in liver CSCs of human HCC cell lines. The CSC properties were evaluated by spheroid culture, CSC marker expression, and sensitivity to anti-cancer drugs. The expression of both NEAT1 variant 1 (NEAT1v1) and variant 2 (NEAT1v2) as well as CD44 was significantly increased in the spheroid culture, compared with that in monolayer culture. Overexpression of Neat1v1, but not Neat1v2, enhanced the CSC properties, while knockout of the NEAT1 gene suppressed them. CD44 expression was increased by the overexpression of Neat1v1 and abrogated by NEAT1 knockout. The overexpression of NEAT1v1 restored the CSC properties and CD44 expression in NEAT1-knockout cells. NEAT1v1 expression in HCC tissues was correlated with poor prognosis and CD44 expression. These results suggest that NEAT1v1 is required for CD44 expression. To our surprise, NEAT1v1 also restored the CSC properties even in CD44-deficient cells, suggesting that NEAT1v1 maintains the properties of CSCs in a CD44-independent manner.
Collapse
Affiliation(s)
- Shigemi Koyama
- Division of Molecular and Genetic Medicine, Graduate School of Medicine, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Hiroyuki Tsuchiya
- Division of Molecular and Genetic Medicine, Graduate School of Medicine, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
- Correspondence: ; Tel./Fax: +81-859-38-6435
| | - Masataka Amisaki
- Division of Surgical Oncology, Department of Surgery, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Hiromi Sakaguchi
- Division of Radiology, Department of Pathophysiological and Therapeutic Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Soichiro Honjo
- Division of Surgical Oncology, Department of Surgery, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Yoshiyuki Fujiwara
- Division of Surgical Oncology, Department of Surgery, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Goshi Shiota
- Division of Molecular and Genetic Medicine, Graduate School of Medicine, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| |
Collapse
|
605
|
APC/C ubiquitin ligase: Functions and mechanisms in tumorigenesis. Semin Cancer Biol 2020; 67:80-91. [PMID: 32165320 DOI: 10.1016/j.semcancer.2020.03.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/24/2020] [Accepted: 03/02/2020] [Indexed: 12/18/2022]
Abstract
The anaphase promoting complex/ cyclosome (APC/C), is an evolutionarily conserved protein complex essential for cellular division due to its role in regulating the mitotic transition from metaphase to anaphase. In this review, we highlight recent work that has shed light on our understanding of the role of APC/C coactivators, Cdh1 and Cdc20, in cancer initiation and development. We summarize the current state of knowledge regarding APC/C structure and function, as well as the distinct ways Cdh1 and Cdc20 are dysregulated in human cancer. We also discuss APC/C inhibitors, novel approaches for targeting the APC/C as a cancer therapy, and areas for future work.
Collapse
|
606
|
Wang J, Sun J, Wang Y, Chou T, Zhang Q, Zhang B, Ren L, Wang H. Gold Nanoframeworks with Mesopores for Raman-Photoacoustic Imaging and Photo-Chemo Tumor Therapy in the Second Near-Infrared Biowindow. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1908825. [PMID: 34163312 PMCID: PMC8218930 DOI: 10.1002/adfm.201908825] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Indexed: 05/20/2023]
Abstract
Gold-based nanostructures with tunable wavelength of localized surface plasmon resonance (LSPR) in the second near-infrared (NIR-II) biowindow receive increasing attention in phototheranostics. In view of limited progress on NIR-II gold nanostructures, a particular liposome template-guided route is explored to synthesize novel gold nanoframeworks (AuNFs) with large mesopores (≈40 nm) for multimodal imaging along with therapeutic robustness. The synthesized AuNFs exhibit strong absorbance in NIR-II region, affording their capacity of NIR-II photothermal therapy (PTT) and photoacoustic (PA) imaging for deep tumors. Functionalization of AuNFs with hyaluronic acid (HA) endows the targeting capacity for CD44-overexpressed tumor cells while gatekeeping doxorubicin (DOX) loaded into mesopores. Conjugation of Raman reporter 4-aminothiophenol (4-ATP) onto AuNFs yields a surface-enhanced Raman scattering (SERS) fingerprint for Raman spectroscopy/imaging. In vivo evaluation of HA-4-ATP-AuNFs-DOX on tumor-bearing xenografts demonstrates its high efficacy in eradication of solid tumors in NIR-II under PA-Raman dual image-guided photo-chemotherapy. Thus, current AuNFs offer versatile capabilities for phototheranostics.
Collapse
Affiliation(s)
- Jinping Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Jingyu Sun
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Yuhao Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Tsengming Chou
- Laboratory for Multiscale Imaging, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Qiang Zhang
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, State Key Lab of Physical Chemistry of Solid Surface, College of Materials, Xiamen University, Xiamen, Fujian 361005, P. R. China
| | - Beilu Zhang
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Lei Ren
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, State Key Lab of Physical Chemistry of Solid Surface, College of Materials, Xiamen University, Xiamen, Fujian 361005, P. R. China
| | - Hongjun Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| |
Collapse
|
607
|
MAOA-mediated reprogramming of stromal fibroblasts promotes prostate tumorigenesis and cancer stemness. Oncogene 2020; 39:3305-3321. [PMID: 32066880 DOI: 10.1038/s41388-020-1217-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 01/29/2020] [Accepted: 02/07/2020] [Indexed: 02/05/2023]
Abstract
The tumor microenvironment plays a critical role in prostate cancer (PC) development and progression. Inappropriate activation of the stroma potentiates the growth and transformation of epithelial tumor cells. Here, we show that upregulation of monoamine oxidase A (MAOA), a mitochondrial enzyme that degrades monoamine neurotransmitters and dietary amines, in stromal cells elevates production of reactive oxygen species, triggers an inflammatory response including activation of IL-6, and promotes tumorigenesis in vitro and in vivo. Mechanistically, MAOA enhances IL-6 transcription through direct Twist1 binding to a conserved E-box element at the IL-6 promoter. MAOA in stromal fibroblasts provides tumor cell growth advantages through paracrine IL-6/STAT3 signaling. Tissue microarray analysis revealed co-expression correlations between individual pairs of proteins of the stromal MAOA-induced Twist1/IL-6/STAT3 pathway in clinical specimens. Downstream of stromal MAOA, STAT3 also promotes cell stemness and transcriptionally activates expression of cancer stem cell marker CD44 in PC cells. MAOA inhibitor treatment effectively suppressed prostate tumor growth in mice in a stroma-specific targeted manner. Collectively, these findings characterize the contribution of MAOA to stromal activation in PC pathogenesis and provide a rationale for targeting MAOA in stromal cells to treat PC.
Collapse
|
608
|
Anuja K, Kar M, Chowdhury AR, Shankar G, Padhi S, Roy S, Akhter Y, Rath AK, Banerjee B. Role of telomeric RAP1 in radiation sensitivity modulation and its interaction with CSC marker KLF4 in colorectal cancer. Int J Radiat Biol 2020; 96:790-802. [PMID: 31985344 DOI: 10.1080/09553002.2020.1721609] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Aims: Radiotherapy is predominantly used as one of the treatment modalities to treat local tumor in colorectal cancer (CRC). Hindrance in disease treatment can be attributed to radio-tolerance of cancer stem cells (CSCs) subsistence in the tumor. Understanding the radio-resistant property of CSCs might help in the accomplishment of targeted radiotherapy treatment and increased disease-free survival. Telomeric RAP1 contributes in modulation of various transcription factors leading to aberrant cell proliferation and tumor cell migration. Therefore, we investigated the role of RAP1 in maintaining resistance phenotype and acquired stemness in radio-resistant cells.Main methods: Characterization of HCT116 derived radio-resistant cell (HCT116RR) was performed by cell survival and DNA damage profiling. RAP1 silenced cells were investigated for DNA damage and expression of CSC markers through western blotting and Real-time PCR post-irradiation. Molecular docking and co-immunoprecipitation study were performed to investigate RAP1 and KLF4 interaction followed by RAP1 protein status profiling in CRC patient.Key findings: We established radio-resistant cells, which showed tolerance to radiotherapy and elevated expression of CSC markers along with RAP1. RAP1 silencing showed enhanced DNA damage and reduced expression of CSC markers post-irradiation. We observed strong physical interaction between RAP1 and KLF4 protein. Furthermore, higher RAP1 expression was observed in the tumor of CRC patients. Dataset analysis also revealed that high expression of RAP1 expression is associated with poor prognosis.Significance: We conclude that higher expression of RAP1 implicates its possible role in promoting radio-resistance in CRC cells by modulating DNA damage and CSC phenotype.
Collapse
Affiliation(s)
- Kumari Anuja
- Molecular Stress and Stem Cell Biology Group, School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Madhabananda Kar
- Department of Surgical Oncology, All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India
| | - Amit Roy Chowdhury
- Molecular Stress and Stem Cell Biology Group, School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Gauri Shankar
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Swatishree Padhi
- Molecular Stress and Stem Cell Biology Group, School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Souvick Roy
- Molecular Stress and Stem Cell Biology Group, School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Yusuf Akhter
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | | | - Birendranath Banerjee
- Molecular Stress and Stem Cell Biology Group, School of Biotechnology, KIIT University, Bhubaneswar, India
| |
Collapse
|
609
|
Kakkad S, Krishnamachary B, Jacob D, Pacheco-Torres J, Goggins E, Bharti SK, Penet MF, Bhujwalla ZM. Molecular and functional imaging insights into the role of hypoxia in cancer aggression. Cancer Metastasis Rev 2020; 38:51-64. [PMID: 30840168 DOI: 10.1007/s10555-019-09788-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hypoxia in cancers has evoked significant interest since 1955 when Thomlinson and Gray postulated the presence of hypoxia in human lung cancers, based on the observation of necrosis occurring at the diffusion limit of oxygen from the nearest blood vessel, and identified the implication of these observations for radiation therapy. Coupled with discoveries in 1953 by Gray and others that anoxic cells were resistant to radiation damage, these observations have led to an entire field of research focused on exploiting oxygenation and hypoxia to improve the outcome of radiation therapy. Almost 65 years later, tumor heterogeneity of nearly every parameter measured including tumor oxygenation, and the dynamic landscape of cancers and their microenvironments are clearly evident, providing a strong rationale for cancer personalized medicine. Since hypoxia is a major cause of extracellular acidosis in tumors, here, we have focused on the applications of imaging to understand the effects of hypoxia in tumors and to target hypoxia in theranostic strategies. Molecular and functional imaging have critically important roles to play in personalized medicine through the detection of hypoxia, both spatially and temporally, and by providing new understanding of the role of hypoxia in cancer aggressiveness. With the discovery of the hypoxia-inducible factor (HIF), the intervening years have also seen significant progress in understanding the transcriptional regulation of hypoxia-induced genes. These advances have provided the ability to silence HIF and understand the associated molecular and functional consequences to expand our understanding of hypoxia and its role in cancer aggressiveness. Most recently, the development of hypoxia-based theranostic strategies that combine detection and therapy are further establishing imaging-based treatment strategies for precision medicine of cancer.
Collapse
Affiliation(s)
- Samata Kakkad
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Rm 208C Traylor Building, Baltimore, MD, 21205, USA
| | - Balaji Krishnamachary
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Rm 208C Traylor Building, Baltimore, MD, 21205, USA
| | - Desmond Jacob
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Rm 208C Traylor Building, Baltimore, MD, 21205, USA
| | - Jesus Pacheco-Torres
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Rm 208C Traylor Building, Baltimore, MD, 21205, USA
| | - Eibhlin Goggins
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Rm 208C Traylor Building, Baltimore, MD, 21205, USA
| | - Santosh Kumar Bharti
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Rm 208C Traylor Building, Baltimore, MD, 21205, USA
| | - Marie-France Penet
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Rm 208C Traylor Building, Baltimore, MD, 21205, USA
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Rm 208C Traylor Building, Baltimore, MD, 21205, USA.
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
610
|
Cole AJ, Fayomi AP, Anyaeche VI, Bai S, Buckanovich RJ. An evolving paradigm of cancer stem cell hierarchies: therapeutic implications. Theranostics 2020; 10:3083-3098. [PMID: 32194856 PMCID: PMC7053211 DOI: 10.7150/thno.41647] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/22/2020] [Indexed: 02/07/2023] Open
Abstract
Over a decade of research has confirmed the critical role of cancer stem-like cells (CSCs) in tumor initiation, chemoresistance, and metastasis. Increasingly, CSC hierarchies have begun to be defined with some recurring themes. This includes evidence that these hierarchies are 'flexible,' with both cell state transitions and dedifferentiation events possible. These findings pose therapeutic hurdles and opportunities. Here, we review cancer stem cell hierarchies and their interactions with the tumor microenvironment. We also discuss the current therapeutic approaches designed to target CSC hierarchies and initial clinical trial results for CSC targeting agents. While cancer stem cell targeted therapies are still in their infancy, we are beginning to see encouraging results that suggest a positive outlook for CSC-targeting approaches.
Collapse
Affiliation(s)
- Alexander J Cole
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adetunji P Fayomi
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Shoumei Bai
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ronald J Buckanovich
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
611
|
Multifunctional, CD44v6-Targeted ORMOSIL Nanoparticles Enhance Drugs Toxicity in Cancer Cells. NANOMATERIALS 2020; 10:nano10020298. [PMID: 32050605 PMCID: PMC7075197 DOI: 10.3390/nano10020298] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/24/2020] [Accepted: 01/31/2020] [Indexed: 12/14/2022]
Abstract
Drug-loaded, PEGylated, organic-modified silica (ORMOSIL) nanoparticles prepared by microemulsion condensation of vinyltriethoxysilane (VTES) were investigated as potential nanovectors for cancer therapy. To target cancer stem cells, anti-CD44v6 antibody and hyaluronic acid (HA) were conjugated to amine-functionalized PEGylated ORMOSIL nanoparticles through thiol-maleimide and amide coupling chemistries, respectively. Specific binding and uptake of conjugated nanoparticles were studied on cells overexpressing the CD44v6 receptor. Cytotoxicity was subsequently evaluated in the same cells after the uptake of the nanoparticles. Internalization of nanocarriers loaded with the anticancer drug 3N-cyclopropylmethyl-7-phenyl-pyrrolo- quinolinone (MG2477) into cells resulted in a substantial increase of the cytotoxicity with respect to the free formulation. Targeting with anti-CD44v6 antibodies or HA yielded nanoparticles with similar effectiveness, in their optimized formulation.
Collapse
|
612
|
Zhao R, Liu Z, Xu W, Song L, Ren H, Ou Y, Liu Y, Wang S. Helicobacter pylori infection leads to KLF4 inactivation in gastric cancer through a TET1-mediated DNA methylation mechanism. Cancer Med 2020; 9:2551-2563. [PMID: 32017451 PMCID: PMC7131848 DOI: 10.1002/cam4.2892] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/04/2020] [Accepted: 01/14/2020] [Indexed: 12/15/2022] Open
Abstract
Krüppel-like factor 4 (KLF4) has a tumor suppressor role in the progression of gastric cancer (GC), and inhibition or loss of KLF4 expression was identified in GC. The aim of this study was to explore the new molecular mechanism of KLF4 inactivation in gastric cancer. Herein, we report that Helicobacter pylori infection or Cag pathogenicity island protein A (CagA) gene transduction resulted in KLF4 expression downregulation and promoted gastric epithelial cell and gastric cancel cell proliferation, migration, and colony formation. Mechanistically, we found that CagA gene transduction led to DNA methylation of the KLF4 promoter, an effect that was relevant to the significant downregulation of TET1 expression. Causally, knockdown of TET1 expression decreased KLF4 expression, whereas overexpression of TET1 had the opposite effect. Clinically, we found that KLF4 expression and the 5-hmC levels were lower in GC cells with H pylori infection than in GC cells without H pylori infection. Thus, our study not only sheds new light on how H pylori infection promotes the progression of GC but also elucidates a novel mechanism of KLF4 inactivation in GC pathogenesis. During pathogenesis, an alteration in the H pylori/CagA-TET1-KLF4 signaling pathway plays a critical role, suggesting that this pathway may be a prospective target for gastric carcinoma intervention and therapy.
Collapse
Affiliation(s)
- Rongrong Zhao
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China.,Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhengxia Liu
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Wenting Xu
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Le Song
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Haifeng Ren
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Yang Ou
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Yakun Liu
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Siying Wang
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
613
|
Si D, Yin F, Peng J, Zhang G. High Expression of CD44 Predicts a Poor Prognosis in Glioblastomas. Cancer Manag Res 2020; 12:769-775. [PMID: 32099472 PMCID: PMC7006859 DOI: 10.2147/cmar.s233423] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/15/2020] [Indexed: 12/15/2022] Open
Abstract
Purpose Glioblastoma multiforme (GBM) is the most common of the malignant and invasive gliomas. High grade glioma is prone to relapse and has a poor prognosis. However, there is a big difference in terms of survival time with the same grade glioma. Cluster of differentiation 44 (CD44) is an indicator of cancer stem cells with abnormal expression in many malignant tumors, however the expression in GBM is unknown. Methods Tissue specimens were collected from 62 GBM patients to investigate CD44 expression and their prognosis was followed-up. Chi-square test was used to identify the association between CD44 staining and clinical characteristics of the patients. Kaplan-Meier analysis was performed to draw survival curves and Cox regression analysis to confirm the independent prognostic factors of GBM patients. Results In total, 38.7% (24/62) of the patients had high CD44 staining. The median survival times were 3.5 months and 18.5 months for high and low expressions of CD44, respectively. Kaplan-Meier analysis revealed that tumor location, the extent of tumor resection, adjuvant chemotherapy, and CD44 expression were related to overall survival time of GBM patients (P<0.05). Multivariate analysis showed that non-usage of adjuvant chemotherapy (HR=4.097, 95% CI=1.489-11.277, P=0.006) and CD44 overexpression (HR=3.216, 95% CI=1.452-7.125, P=0.004) were independent unfavorable prognostic factors for GBM patients. Conclusion The results demonstrate that high expression of CD44 acts as a poor prognosis indicator in GBM patients.
Collapse
Affiliation(s)
- Daolin Si
- Department of Pediatric Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, People's Republic of China
| | - Fei Yin
- Department of Pediatric Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, People's Republic of China
| | - Jing Peng
- Department of Pediatric Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, People's Republic of China
| | - Guangying Zhang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, People's Republic of China
| |
Collapse
|
614
|
Terada D, Genjo T, Segawa TF, Igarashi R, Shirakawa M. Nanodiamonds for bioapplications–specific targeting strategies. Biochim Biophys Acta Gen Subj 2020; 1864:129354. [DOI: 10.1016/j.bbagen.2019.04.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/25/2019] [Indexed: 12/21/2022]
|
615
|
Zhou S, Zhu X, Liu W, Cheng F, Zou P, You Y, Xiao Y, Guo A, Zhu X. Comparison of chronic myeloid leukemia stem cells and hematopoietic stem cells by global proteomic analysis. Biochem Biophys Res Commun 2020; 522:362-367. [DOI: 10.1016/j.bbrc.2019.11.092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 11/14/2019] [Accepted: 11/14/2019] [Indexed: 01/01/2023]
|
616
|
Shen S, Lu H, Liu L, Wang Y, Zhang C, Yang W, Xu W. Role of CD44 in tumor-initiating cells of salivary gland pleomorphic adenoma: More than a surface biomarker. Oral Dis 2020; 26:547-557. [PMID: 31926047 DOI: 10.1111/odi.13279] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 12/24/2019] [Accepted: 01/05/2020] [Indexed: 02/06/2023]
Abstract
CD44, a cell-surface glycoprotein, functions as a receptor for hyaluronic acid. Our research group has previously shown that CD44 is a biomarker for the CD44hi cells (tumor-initiating cells; TICs) in murine salivary gland tumors. However, little is known concerning the biological roles of CD44 in the tumorigenesis of pleomorphic adenoma. The present study is aimed to investigate the effects of CD44 on the proliferation, invasive capability, and apoptosis of TICs in vitro, as well as the tumorigenicity of TICs in vivo. The results demonstrated that knockdown of CD44 attenuated the malignant phenotype of TICs. Furthermore, in vivo xenograft studies indicated that CD44 knockdown inhibited tumorigenesis of pleomorphic adenoma. In addition, neither the CD44low cells nor the CD44-modified CD44low cells developed neo-tumors, which indicated that overexpression of CD44 did not enable the CD44low cells to be transformed into TICs. Taken together, these data demonstrate that CD44 not only acts as a biomarker, but also functions as a key player in the tumor-initiating capacity of TICs. These results shed light on the pathogenesis of salivary gland tumors and provide a potential therapeutic target for treating pleomorphic adenoma.
Collapse
Affiliation(s)
- Shukun Shen
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Lu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Limin Liu
- Department of Oral pathology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Wang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chenping Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenjun Yang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wanlin Xu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
617
|
Jang JH, Kim DH, Lim JM, Lee JW, Jeong SJ, Kim KP, Surh YJ. Breast Cancer Cell-Derived Soluble CD44 Promotes Tumor Progression by Triggering Macrophage IL1β Production. Cancer Res 2020; 80:1342-1356. [PMID: 31969374 DOI: 10.1158/0008-5472.can-19-2288] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/27/2019] [Accepted: 01/13/2020] [Indexed: 11/16/2022]
Abstract
IL1β is a central regulator of systemic inflammatory response in breast cancer, but the precise regulatory mechanisms that dictate the overproduction of IL1β are largely unsolved. Here, we show that IL1β secretion is increased by the coculture of human monocyte-like cells and triple-negative breast cancer (TNBC) cells. In addition, macrophages robustly produced IL1β when exposed to the conditioned media of TNBC cells. Consistent with these observations, macrophage depletion decreased serum IL1β and reduced breast cancer progression in an orthotopic breast cancer mouse model. Profiling the secretome of human breast cancer cells revealed that the CD44 antigen was the most differentially released protein in basal conditions of TNBC cells. Antibody-mediated neutralization of CD44 abrogated IL1β production in macrophages and inhibited the growth of primary tumors. These results suggest IL1β-mediated oncogenic signaling is triggered by breast cancer cell membrane-derived soluble CD44 (sCD44) antigen, and targeting sCD44 antigen may provide an alternative therapeutic strategy for breast cancer treatment by modulating inflammatory tumor microenvironment. SIGNIFICANCE: A novel positive feedback loop between IL1β and CD44 promotes TNBC malignant progression.
Collapse
Affiliation(s)
- Jeong-Hoon Jang
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Do-Hee Kim
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea.,Cancer Research Institute, Seoul National University, Seoul, South Korea
| | - Jae Min Lim
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Youngin, South Korea.,Department of Biomedical Science and Technology, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, South Korea
| | - Joon Won Lee
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Youngin, South Korea.,Department of Biomedical Science and Technology, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, South Korea
| | - Su Jin Jeong
- Statistics Support Department, Medical Science Research Institute, Kyung Hee University Hospital, Seoul, South Korea
| | - Kwang Pyo Kim
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Youngin, South Korea.,Department of Biomedical Science and Technology, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, South Korea
| | - Young-Joon Surh
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea. .,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea.,Cancer Research Institute, Seoul National University, Seoul, South Korea
| |
Collapse
|
618
|
Targeting strategies for superparamagnetic iron oxide nanoparticles in cancer therapy. Acta Biomater 2020; 102:13-34. [PMID: 31759124 DOI: 10.1016/j.actbio.2019.11.027] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/01/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022]
Abstract
Among various nanoparticles, superparamagnetic iron oxide nanoparticles (SPIONs) have been increasingly studied for their excellent superparamagnetism, magnetic heating properties, and enhanced magnetic resonance imaging (MRI). The conjugation of SPIONs with drugs to obtain delivery nanosystems has several advantages including magnetic targeted functionalization, in vivo imaging, magnetic thermotherapy, and combined delivery of anticancer agents. To further increase the targeting efficiency of drugs through a delivery nanosystem based on SPIONs, additional targeting moieties including transferrin, antibodies, aptamers, hyaluronic acid, folate, and targeting peptides are coated onto the surface of SPIONs. Therefore, this review summarizes the latest progresses in the conjugation of targeting molecules and drug delivery nanosystems based on SPIONs, especially focusing on their performances to develop efficient targeted drug delivery systems for tumor therapy. STATEMENT OF SIGNIFICANCE: Some magnetic nanoparticle-based nanocarriers loaded with drugs were evaluated in patients and did not produce convincing results, leading to termination of clinical development in phase II/III. An alternative strategy for drug delivery systems based on SPIONs is the conjugation of these systems with targeting segments such as transferrin, antibodies, aptamers, hyaluronic acid, folate, and targeting peptides. These targeting moieties can be recognized by specific integrin/receptors that are overexpressed specifically on the tumor cell surface, resulting in minimizing dosage and reducing off-target effects. This review focuses on magnetic nanoparticle-based nonviral drug delivery systems with targeting moieties to deliver anticancer drugs, with an aim to provide suggestions on the development of SPIONs through discussion.
Collapse
|
619
|
Crocetin Extracted from Saffron Shows Antitumor Effects in Models of Human Glioblastoma. Int J Mol Sci 2020; 21:ijms21020423. [PMID: 31936544 PMCID: PMC7013996 DOI: 10.3390/ijms21020423] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/23/2019] [Accepted: 01/04/2020] [Indexed: 12/14/2022] Open
Abstract
Over recent years, many authors discussed the effects of different natural compounds on glioblastoma (GBM). Due to its capacity to impair survival and progression of different cancer types, saffron extract (SE), named crocetin (CCT), is particularly noteworthy. In this work, we elucidated the antitumor properties of crocetin in glioma in vivo and in vitro models for the first time. The in vitro results showed that the four tumor cell lines observed in this study (U251, U87, U138, and U373), which were treated with increasing doses of crocetin, showed antiproliferative and pro-differentiative effects as demonstrated by a significant reduction in the number of viable cells, deep changes in cell morphology, and the modulation of mesenchymal and neuronal markers. Indeed, crocetin decreased the expression of Cluster of Differentiation CD44, CD90, CXCR4, and OCT3/4 mesenchymal markers, but increased the expression of βIII-Tubulin and neurofilaments (NFH) neuronal linage-related markers. Epigenetic mechanisms may modulate these changes, since Histone Deacetylase, HDAC1 and HDAC3 were downmodulated in U251 and U87 cells, whereas HDAC1 expression was downmodulated in U138 and U373 cells. Western blotting analyses of Fatty Acid Synthase, FASN, and CD44 resulted in effective inhibition of these markers after CCT treatment, which was associated with important activation of the apoptosis program and reduced glioma cell movement and wound repair. The in vivo studies aligned with the results obtained in vitro. Indeed, crocetin was demonstrated to inhibit the growth of U251 and U87 cells that were subcutaneously injected into animal models. In particular, the Tumor To Progression or TTP values and Kaplan-Meier curves indicated that crocetin had more major effects than radiotherapy alone, but similar effects to temozolomide (TMZ). An intra-brain cell inoculation of a small number of luciferase-transfected U251 cells provided a model that was able to recapitulate recurrence after surgical tumor removal. The results obtained from the orthotopic intra-brain model indicated that CCT treatment increased the disease-free survival (DFS) and overall survival (OS) rates, inducing a delay in appearance of a detectable bioluminescent lesion. CCT showed greater efficacy than Radio Therapy (RT) but comparable efficacy to temozolomide in xenograft models. Therefore, we aimed to continue the study of crocetin's effects in glioma disease, focusing our attention on the radiosensitizing properties of the natural compound and highlighting the ways in which this was realized.
Collapse
|
620
|
Gastric Cancer Stem Cells: Current Insights into the Immune Microenvironment and Therapeutic Targets. Biomedicines 2020; 8:biomedicines8010007. [PMID: 31935894 PMCID: PMC7168269 DOI: 10.3390/biomedicines8010007] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/27/2019] [Accepted: 01/04/2020] [Indexed: 02/07/2023] Open
Abstract
Gastric cancer (GC) is a leading cause of cancer-related death worldwide. Cancer stem cells (CSCs) are known to be involved in chemotherapy resistance and the development of metastases. Although CSCs harbor self-renewal and tumorigenic abilities, the immune microenvironment surrounding CSCs provides various factors and supports the maintenance of CSC properties. The current review summarizes the accumulating findings regarding the relationship between the immune microenvironment and gastric CSCs (GCSCs), which will support the possibility of developing novel therapeutic strategies for targeting GCSCs.
Collapse
|
621
|
Ghanei Z, Jamshidizad A, Joupari MD, Shamsara M. Isolation and characterization of breast cancer stem cell-like phenotype by Oct4 promoter-mediated activity. J Cell Physiol 2020; 235:7840-7848. [PMID: 31904128 DOI: 10.1002/jcp.29437] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 12/23/2019] [Indexed: 12/18/2022]
Abstract
Cancer stem cells (CSCs) are a small subset of cancer cells responsible for self-renewal activity, drug resistance, and tumor recurrence. CSCs have been derived from diverse tumors and cell lines. The expression of stemness markers has been identified in CSCs. Oct4 is a well-established transcription factor expressed in stem cells and CSCs. In this study, we isolated and characterized breast CSC-like cells from murine MC4-L2 cells by Oct4 promoter-mediated activity. The MC4-L2 cells were electroporated by a plasmid expressing puromycin resistance (PuroR ) gene from the Oct4 promoter and then selected by puromycin. The isolated cells were named as the MC4-L2puro cells and characterized for CSCs properties. Immunostaining indicated CD44high and CD24high phenotype for the MC4-L2 and MC4-L2puro cells. The enhanced expression of stem cell markers was detected in the puromycin-selected cells compared with the parental cells. Moreover, the isolated cells only grew up in sphere-formed shape in low attachment plates. Serial dilution transplantation in syngeneic mouse models showed increased tumorigenicity of the MC4-L2puro cells, as they induced new tumors when injected into the mammary fat pad as few as 104 cells. In conclusion, we designed a novel genetic construct, which allows the isolation of Oct4-positive cells in a cancer population by a simple selection step in a puromycin-containing medium. Transfection of this construct into the MC4-L2 cells resulted in growing a subpopulation of cells having tumor-initiating cell characteristics. To the best of our knowledge, this is the first report on the isolation of CSC-like cells from the mouse breast cancer MC4-L2 cells.
Collapse
Affiliation(s)
- Zahra Ghanei
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Abbas Jamshidizad
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Morteza Daliri Joupari
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mehdi Shamsara
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
622
|
Chen KL, Li D, Lu TX, Chang SW. Structural Characterization of the CD44 Stem Region for Standard and Cancer-Associated Isoforms. Int J Mol Sci 2020; 21:E336. [PMID: 31947887 PMCID: PMC6982006 DOI: 10.3390/ijms21010336] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/26/2019] [Accepted: 12/31/2019] [Indexed: 12/18/2022] Open
Abstract
CD44 is widely expressed in most vertebrate cells, whereas the expression of CD44v6 is restricted to only a few tissues and has been considered to be associated with tumor progression and metastasis. Thus, CD44v6 has been recognized as a promising prognostic biomarker and therapeutic target for various cancers for more than a decade. However, despite many experimental studies, the structural dynamics and differences between CD44s and CD44v6, particularly in their stem region, still remain elusive. Here, a computational study was conducted to address these problems. We found that the stem of CD44s adopted predominantly two conformations, one featuring antiparallel β-sheets and the other featuring parallel β-sheets, whereas the stem of CD44v6 adopted mainly one conformation with relatively highly suppressed β-sheet contents. Moreover, Phe215 was found to be essential in the β-sheets of both CD44s and CD44v6. We finally found intramolecular Phe215-Trp224 hydrogen-bonding interactions and hydrophobic interactions with Phe215 that cooperatively drove conformational differences upon the addition of the v6 region to CD44. Our study elucidated the structural differences between the stem regions of CD44s and CD44v6 and thus can offer useful structural information for drug design to specifically target CD44v6 in promising clinical applications.
Collapse
Affiliation(s)
- Kun-Lin Chen
- Department of Engineering Science and Ocean Engineering, National Taiwan University, Taipei 10617, Taiwan;
| | - Deng Li
- Department of Civil Engineering, National Taiwan University, Taipei 10617, Taiwan;
| | - Ting-Xuan Lu
- Department and Graduate Institute of Pharmacology, National Taiwan University, Taipei 10617, Taiwan;
| | - Shu-Wei Chang
- Department of Civil Engineering, National Taiwan University, Taipei 10617, Taiwan;
| |
Collapse
|
623
|
Abstract
The extracellular matrix is part of the microenvironment and its functions are associated with the physical and chemical properties of the tissue. Among the extracellular components, the glycosaminoglycan hyaluronan is a key component, defining both the physical and biochemical characteristics of the healthy matrices. The hyaluronan metabolism is strictly regulated in physiological conditions, but in the tumoral tissues, its expression, size and binding proteins interaction are dysregulated. Hyaluronan from the tumor microenvironment promotes tumor cell proliferation, invasion, immune evasion, stemness alterations as well as drug resistance. This chapter describes data regarding novel concepts of hyaluronan functions in the tumor. Additionally, we discuss potential clinical applications of targeting HA metabolism in cancer therapy.
Collapse
|
624
|
Abstract
Imaging of cancer cells and cancer stem cells (CSCs) plays an important role in studying cell biology and tracking cancer development and metastasis. There is a wide interest in targeting cancer cells using fluorescent nanoclusters (NCs) capped in protein due to excellent cell viability and photostability, one-step synthesis route, large Stokes shift, good aqueous stability, and easy functionalization capability with long lifetime. Since CD44 is a CSC marker as well as a transmembrane receptor for hyaluronic acid (HA) and many other extracellular matrix (ECM) components, in this protocol, a biocompatible platform was synthesized by conjugation of HA onto luminescent platinum nanoclusters (Pt NCs) in human hemoglobin (Hb) (Hb/Pt NCs). This bioplatform could be used for specific imaging via an efficient targeting of CD44-overexpressing cancer cells and cancer stem cells.
Collapse
|
625
|
Martin-Orozco E, Sanchez-Fernandez A, Ortiz-Parra I, Ayala-San Nicolas M. WNT Signaling in Tumors: The Way to Evade Drugs and Immunity. Front Immunol 2019; 10:2854. [PMID: 31921125 PMCID: PMC6934036 DOI: 10.3389/fimmu.2019.02854] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 11/20/2019] [Indexed: 12/19/2022] Open
Abstract
WNT/β-catenin signaling is involved in many physiological processes. Its implication in embryonic development, cell migration, and polarization has been shown. Nevertheless, alterations in this signaling have also been related with pathological events such as sustaining and proliferating the cancer stem cell (CSC) subset present in the tumor bulk. Related with this, WNT signaling has been associated with the maintenance, expansion, and epithelial-mesenchymal transition of stem cells, and furthermore with two distinctive features of this tumor population: therapeutic resistance (MDR, multidrug resistance) and immune escape. These mechanisms are developed and maintained by WNT activation through the transcriptional control of the genes involved in such processes. This review focuses on the description of the best known WNT pathways and the molecules involved in them. Special attention is given to the WNT cascade proteins deregulated in tumors, which have a decisive role in tumor survival. Some of these proteins function as extrusion pumps that, in the course of chemotherapy, expel the drugs from the cells; others help the tumoral cells hide from the immune effector mechanisms. Among the WNT targets involved in drug resistance, the drug extrusion pump MDR-1 (P-GP, ABCB1) and the cell adhesion molecules from the CD44 family are highlighted. The chemokine CCL4 and the immune checkpoint proteins CD47 and PD-L1 are included in the list of WNT target molecules with a role in immunity escape. This pathway should be a main target in cancer therapy as WNT signaling activation is essential for tumor progression and survival, even in the presence of the anti-tumoral immune response and/or antineoplastic drugs. The appropriate design and combination of anti-tumoral strategies, based on the modulation of WNT mediators and/or protein targets, could negatively affect the growth of tumoral cells, improving the efficacy of these types of therapies.
Collapse
Affiliation(s)
- Elena Martin-Orozco
- Department of Biochemistry and Molecular Biology (B) and Immunology, School of Medicine, University of Murcia, Murcia, Spain.,Biomedical Research Institute of Murcia (IMIB), ARADyAL, Murcia, Spain
| | - Ana Sanchez-Fernandez
- Department of Biochemistry and Molecular Biology (B) and Immunology, School of Medicine, University of Murcia, Murcia, Spain
| | - Irene Ortiz-Parra
- Department of Biochemistry and Molecular Biology (B) and Immunology, School of Medicine, University of Murcia, Murcia, Spain
| | - Maria Ayala-San Nicolas
- Department of Biochemistry and Molecular Biology (B) and Immunology, School of Medicine, University of Murcia, Murcia, Spain
| |
Collapse
|
626
|
Masciale V, Grisendi G, Banchelli F, D'Amico R, Maiorana A, Sighinolfi P, Stefani A, Morandi U, Dominici M, Aramini B. Isolation and Identification of Cancer Stem-Like Cells in Adenocarcinoma and Squamous Cell Carcinoma of the Lung: A Pilot Study. Front Oncol 2019; 9:1394. [PMID: 31921651 PMCID: PMC6930193 DOI: 10.3389/fonc.2019.01394] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/26/2019] [Indexed: 12/17/2022] Open
Abstract
Background: Lung cancer stem cells (CSCs) share many characteristics with normal stem cells, such as self-renewal and multipotentiality. High expression of aldehyde dehydrogenase (ALDH) has been detected in many tumors, particularly in the CSC compartment, and it plays an important role in tumor proliferation, metastasis, and drug resistance. CD44 is commonly used as a cell surface marker of cancer stem-like cells in epithelial tumors. The aim of this study was to isolate and analyze cancer stem-like cells from surgically removed specimens to compare lung adenocarcinoma (ADENO) and squamous (SQUAMO) cell carcinoma. Methods: The ALDEFLUOR assay was used to identify and sort ALDHhigh and ALDHlow human lung cancer cells following tissue digestion. Fluorescence-activated cell sorting analysis for CD44 was performed with tumor cells. Quantitative real-time PCR was performed to assess the expression of SOX2 and NANOG as stemness markers. ALDH1A1 expression was additionally determined by immunohistochemistry. Anchorage-independent ALDHhigh cell growth was also evaluated. ALDHhigh ADENO and SQUAMO cells were cultured to analyze spheroid formation. Results: All specimens contained 0.5-12.5% ALDHhigh cells with 3.8-18.9% CD44-positive cells. SOX2 and NANOG relative expression in ALDHhigh compared to ALDHlow cells in ADENO and SQUAMO was analyzed and compared between the histotypes. Immunohistochemistry confirmed the presence of ALDH1A1 in the sections. SOX2 and NANOG were expressed at higher levels in the ALDHhigh subpopulation than in the ALDHlow subpopulation only in ADENO cells, and the opposite result was seen in SQUAMO cells. In vitro functional assays demonstrated that ALDHhigh cells exhibited migration capacity with distinct behaviors between ALDHhigh spheres in ADENO vs. SQUAMO samples. Conclusions: Our results highlight the importance of a better characterization of cancer stem-like cells in ADENO and SQUAMO histotypes. This may suggest new differential approaches for prognostic and therapeutic purposes in patients with non-small-cell lung cancer.
Collapse
Affiliation(s)
- Valentina Masciale
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Grisendi
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy.,Rigenerand SRL, Modena, Italy
| | - Federico Banchelli
- Department of Medical and Surgical Sciences for Children & Adults, Center of Medical Statistic, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberto D'Amico
- Department of Medical and Surgical Sciences for Children & Adults, Center of Medical Statistic, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonino Maiorana
- Department of Medical and Surgical Sciences for Children & Adults, Institute of Pathology, University of Modena and Reggio Emilia, Modena, Italy
| | - Pamela Sighinolfi
- Department of Medical and Surgical Sciences for Children & Adults, Institute of Pathology, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandro Stefani
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Uliano Morandi
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Beatrice Aramini
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
627
|
Cheng C, Easton J, Rosencrance C, Li Y, Ju B, Williams J, Mulder HL, Pang Y, Chen W, Chen X. Latent cellular analysis robustly reveals subtle diversity in large-scale single-cell RNA-seq data. Nucleic Acids Res 2019; 47:e143. [PMID: 31566233 PMCID: PMC6902034 DOI: 10.1093/nar/gkz826] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/30/2019] [Accepted: 09/26/2019] [Indexed: 12/21/2022] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) is a powerful tool for characterizing the cell-to-cell variation and cellular dynamics in populations which appear homogeneous otherwise in basic and translational biological research. However, significant challenges arise in the analysis of scRNA-seq data, including the low signal-to-noise ratio with high data sparsity, potential batch effects, scalability problems when hundreds of thousands of cells are to be analyzed among others. The inherent complexities of scRNA-seq data and dynamic nature of cellular processes lead to suboptimal performance of many currently available algorithms, even for basic tasks such as identifying biologically meaningful heterogeneous subpopulations. In this study, we developed the Latent Cellular Analysis (LCA), a machine learning-based analytical pipeline that combines cosine-similarity measurement by latent cellular states with a graph-based clustering algorithm. LCA provides heuristic solutions for population number inference, dimension reduction, feature selection, and control of technical variations without explicit gene filtering. We show that LCA is robust, accurate, and powerful by comparison with multiple state-of-the-art computational methods when applied to large-scale real and simulated scRNA-seq data. Importantly, the ability of LCA to learn from representative subsets of the data provides scalability, thereby addressing a significant challenge posed by growing sample sizes in scRNA-seq data analysis.
Collapse
Affiliation(s)
- Changde Cheng
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - John Easton
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Celeste Rosencrance
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yan Li
- The University of Texas MD Anderson Cancer Center UTHealthGraduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Bensheng Ju
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Justin Williams
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Heather L Mulder
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yakun Pang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Wenan Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xiang Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
628
|
Evaluating nuclear translocation of surface receptors: recommendations arising from analysis of CD44. Histochem Cell Biol 2019; 153:77-87. [DOI: 10.1007/s00418-019-01835-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2019] [Indexed: 12/16/2022]
|
629
|
Carbonetti G, Wilpshaar T, Kroonen J, Studholme K, Converso C, d'Oelsnitz S, Kaczocha M. FABP5 coordinates lipid signaling that promotes prostate cancer metastasis. Sci Rep 2019; 9:18944. [PMID: 31831821 PMCID: PMC6908725 DOI: 10.1038/s41598-019-55418-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/15/2019] [Indexed: 01/11/2023] Open
Abstract
Prostate cancer (PCa) is defined by dysregulated lipid signaling and is characterized by upregulation of lipid metabolism-related genes including fatty acid binding protein 5 (FABP5), fatty acid synthase (FASN), and monoacylglycerol lipase (MAGL). FASN and MAGL are enzymes that generate cellular fatty acid pools while FABP5 is an intracellular chaperone that delivers fatty acids to nuclear receptors to enhance PCa metastasis. Since FABP5, FASN, and MAGL have been independently implicated in PCa progression, we hypothesized that FABP5 represents a central mechanism linking cytosolic lipid metabolism to pro-metastatic nuclear receptor signaling. Here, we show that the abilities of FASN and MAGL to promote nuclear receptor activation and PCa metastasis are critically dependent upon co-expression of FABP5 in vitro and in vivo. Our findings position FABP5 as a key driver of lipid-mediated metastasis and suggest that disruption of lipid signaling via FABP5 inhibition may constitute a new avenue to treat metastatic PCa.
Collapse
Affiliation(s)
- Gregory Carbonetti
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794, USA.,Department of Anesthesiology, Stony Brook University, Stony Brook, NY, 11794, USA.,Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Tessa Wilpshaar
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794, USA.,Department of Anesthesiology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Jessie Kroonen
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794, USA.,Department of Anesthesiology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Keith Studholme
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Cynthia Converso
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794, USA.,Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Simon d'Oelsnitz
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Martin Kaczocha
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794, USA. .,Department of Anesthesiology, Stony Brook University, Stony Brook, NY, 11794, USA. .,Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
630
|
Xiong X, Schober M, Tassone E, Khodadadi-Jamayran A, Sastre-Perona A, Zhou H, Tsirigos A, Shen S, Chang M, Melamed J, Ossowski L, Wilson EL. KLF4, A Gene Regulating Prostate Stem Cell Homeostasis, Is a Barrier to Malignant Progression and Predictor of Good Prognosis in Prostate Cancer. Cell Rep 2019; 25:3006-3020.e7. [PMID: 30540935 PMCID: PMC6405286 DOI: 10.1016/j.celrep.2018.11.065] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 11/07/2018] [Accepted: 11/15/2018] [Indexed: 12/18/2022] Open
Abstract
There is a considerable need to identify those individuals with prostate cancer who have indolent disease. We propose that
genes that control adult stem cell homeostasis in organs with slow turnover, such as the prostate, control cancer fate. One such
gene, KLF4, overexpressed in murine prostate stem cells, regulates their homeostasis, blocks malignant transformation, and
controls the self-renewal of tumor-initiating cells. KLF4 loss induces the molecular features of aggressive cancer and converts
PIN lesions to invasive sarcomatoid carcinomas; its re-expression in vivo reverses this process. Bioinformatic
analysis links these changes to human cancer. KLF4 and its downstream targets make up a gene signature that identifies indolent
tumors and predicts recurrence-free survival. This approach may improve prognosis and identify therapeutic targets for advanced
cancer. Available criteria for segregating prostate cancer patients into those requiring therapeutic intervention and those who can
be followed are inadequate. Xiong et al. show that KLF4 and its downstream targets make up a gene signature that identifies
indolent tumors. This approach may improve prognosis and identify therapeutic targets for advanced cancer.
Collapse
Affiliation(s)
- Xiaozhong Xiong
- Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA.
| | - Markus Schober
- Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA; Department of Dermatology, NYU School of Medicine, New York, NY 10016, USA
| | - Evelyne Tassone
- Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Alireza Khodadadi-Jamayran
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA; Applied Bioinformatics Laboratories, NYU School of Medicine, New York, NY 10016, USA
| | - Ana Sastre-Perona
- Department of Dermatology, NYU School of Medicine, New York, NY 10016, USA
| | - Hua Zhou
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA; Applied Bioinformatics Laboratories, NYU School of Medicine, New York, NY 10016, USA
| | - Aristotelis Tsirigos
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA; Applied Bioinformatics Laboratories, NYU School of Medicine, New York, NY 10016, USA
| | - Steven Shen
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Miao Chang
- Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Jonathan Melamed
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
| | - Liliana Ossowski
- Department of Medicine, Mt. Sinai School of Medicine, New York, NY 10029, USA
| | - Elaine L Wilson
- Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA; Department of Urology, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
631
|
The PPARδ Agonist GW501516 Improves Lipolytic/Lipogenic Balance through CPT1 and PEPCK during the Development of Pre-Implantation Bovine Embryos. Int J Mol Sci 2019; 20:ijms20236066. [PMID: 31810173 PMCID: PMC6928732 DOI: 10.3390/ijms20236066] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/20/2019] [Accepted: 11/29/2019] [Indexed: 12/15/2022] Open
Abstract
The PPARs (peroxisome proliferator-activated receptors) play critical roles in the regulation of lipid and glucose metabolism. PPARδ, a member of the PPARs family, is associated with decreased susceptibility to ectopic lipid deposition and is implicated in the regulation of mitochondrial processes. The current study aimed to determine the role of PPARδ in fatty acid β-oxidation and its influence on PEPCK for the lipogenic/lipolytic balance during in vitro bovine oocyte maturation and embryo development. Activation of PPARδ by GW501516, but not 2-BP, was indicated by intact embryonic PEPCK (cytosolic) and CPT1 expression and the balance between free fatty acids and mitochondrial β-oxidation that reduced ROS and inhibited p-NF-κB nuclear localization. Genes involved in lipolysis, fatty acid oxidation, and apoptosis showed significant differences after the GW501516 treatment relative to the control- and 2-BP-treated embryos. GSK3787 reversed the PPARδ-induced effects by reducing PEPCK and CPT1 expression and the mitochondrial membrane potential, revealing the importance of PPARδ/PEPCK and PPARδ/CPT1 for controlling lipolysis during embryo development. In conclusion, GW501516-activated PPARδ maintained the correlation between lipolysis and lipogenesis by enhancing PEPCK and CPT1 to improve bovine embryo quality.
Collapse
|
632
|
Chen YC, Sahoo S, Brien R, Jung S, Humphries B, Lee W, Cheng YH, Zhang Z, Luker KE, Wicha MS, Luker GD, Yoon E. Single-cell RNA-sequencing of migratory breast cancer cells: discovering genes associated with cancer metastasis. Analyst 2019; 144:7296-7309. [PMID: 31710321 PMCID: PMC8942075 DOI: 10.1039/c9an01358j] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Considerable evidence suggests breast cancer metastasis arises from cells undergoing epithelial-to-mesenchymal-transition (EMT) and cancer stem-like cells (CSCs). Using a microfluidic device that enriches migratory breast cancer cells with enhanced capacity for tumor formation and metastasis, we identified genes differentially expressed in migratory cells by high-throughput single-cell RNA-sequencing. Migratory cells exhibited overall signatures of EMT and CSCs with variable expression of marker genes, and they retained expression profiles of EMT over time. With single-cell resolution, we discovered intermediate EMT states and distinct epithelial and mesenchymal sub-populations of migratory cells, indicating breast cancer cells can migrate rapidly while retaining an epithelial state. Migratory cells showed differential profiles for regulators of oxidative stress, mitochondrial morphology, and the proteasome, revealing potential vulnerabilities and unexpected consequences of drugs. We also identified novel genes correlated with cell migration and outcomes in breast cancer as potential prognostic biomarkers and therapeutic targets to block migratory cells in metastasis.
Collapse
Affiliation(s)
- Yu-Chih Chen
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI 48109-2122
- Forbes Institute for Cancer Discovery, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI 48109, USA
| | - Saswat Sahoo
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd. Ann Arbor, MI 48109-2099, USA
| | - Riley Brien
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI 48109-2122
| | - Seungwon Jung
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI 48109-2122
| | - Brock Humphries
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA
| | - Woncheol Lee
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI 48109-2122
| | - Yu-Heng Cheng
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI 48109-2122
| | - Zhixiong Zhang
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI 48109-2122
| | - Kathryn E. Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA
| | - Max S. Wicha
- Forbes Institute for Cancer Discovery, University of Michigan, 2800 Plymouth Rd., Ann Arbor, MI 48109, USA
| | - Gary D. Luker
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd. Ann Arbor, MI 48109-2099, USA
- Center for Molecular Imaging, Department of Radiology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA
- Department of Microbiology and Immunology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA
| | - Euisik Yoon
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI 48109-2122
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd. Ann Arbor, MI 48109-2099, USA
- Center for Nanomedicine, Institute for Basic Science (IBS) and Graduate Program of Nano Biomedical Engineering (Nano BME), Yonsei University, Seoul 03722, Korea
| |
Collapse
|
633
|
Windmöller BA, Greiner JF, Förster C, Wilkens L, Mertzlufft F, Esch JSA, Kaltschmidt B, Kaltschmidt C, Beshay M. A typical carcinoid of the lung - a case report with pathological correlation and propagation of the cancer stem cell line BKZ1 with synaptophysin expression. Medicine (Baltimore) 2019; 98:e18174. [PMID: 31804333 PMCID: PMC6919531 DOI: 10.1097/md.0000000000018174] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
RATIONALE Neuroendocrine tumors (NETs) of the lung account for 5% of all cases of lung cancer, which itself is the leading cause of cancer-related death worldwide. In accordance to its rarity, only few cell lines of NETs exist, which even often lack key characteristics of the primary tumor, making it difficult to study underlying molecular mechanisms. PATIENT CONCERNS The patient reported in this case is a 71-year old woman, which never smoked but suffered under dry cough. DIAGNOSES Chest CT-scan showed a paracardiac nodule of the lingula with 2 × 1.8 cm in diameter. INTERVENTIONS The detected paracardiac nodule of the lingula was anatomically resected using video assisted thoracic surgery. OUTCOMES Histopathological diagnostic of the removed tissue identified the tumor as a well-differentiated typical carcinoid (TC), which represents one of the four subgroups of pulmonary NETs. Next to the successful treatment of the patient, we were able to propagate cancer stem cells (CSCs) out of the resected tumor tissue. To the best of our knowledge, we firstly isolated CSCs of a typical carcinoid, which were positive for the prominent CSC markers CD44, CD133 and nestin, confirming their stem cell properties. Additionally, CSCs, further referred as BKZ1, expressed the neuroendocrine marker synaptophysin, verifying their neuroendocrine origin. However, nuclear synaptophysin protein was also present in other stem cell populations, suggesting a role as general stem cell marker. LESSON In line with the importance of CSCs in cancer treatment and the lack of CSC-models for neuroendocrine neoplasms, the here described BKZ1 cancer stem cell line of a typical carcinoid represents a promising new model to study pulmonary carcinoids and particular NETs.
Collapse
Affiliation(s)
- Beatrice Ariane Windmöller
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, Bielefeld
- Forschungsverbund BioMedizin Bielefeld, FBMB, Maraweg 21, Bielefeld, Germany
| | - Johannes F.W. Greiner
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, Bielefeld
- Forschungsverbund BioMedizin Bielefeld, FBMB, Maraweg 21, Bielefeld, Germany
| | - Christine Förster
- Institute of Pathology, KRH Hospital Nordstadt, Haltenhoffstrasse 41, Hannover
- Forschungsverbund BioMedizin Bielefeld, FBMB, Maraweg 21, Bielefeld, Germany
| | - Ludwig Wilkens
- Institute of Pathology, KRH Hospital Nordstadt, Haltenhoffstrasse 41, Hannover
- Forschungsverbund BioMedizin Bielefeld, FBMB, Maraweg 21, Bielefeld, Germany
| | - Fritz Mertzlufft
- Protestant Hospital of Bethel Foundation, Maraweg 21
- Forschungsverbund BioMedizin Bielefeld, FBMB, Maraweg 21, Bielefeld, Germany
| | - Jan Schulte am Esch
- Department of General and Visceral Surgery, Protestant Hospital of Bethel Foundation, Schildescher Strasse 99
- Forschungsverbund BioMedizin Bielefeld, FBMB, Maraweg 21, Bielefeld, Germany
| | - Barbara Kaltschmidt
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, Bielefeld
- Molecular Neurobiology, University of Bielefeld, Universitätsstrasse 25
- Forschungsverbund BioMedizin Bielefeld, FBMB, Maraweg 21, Bielefeld, Germany
| | - Christian Kaltschmidt
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, Bielefeld
- Forschungsverbund BioMedizin Bielefeld, FBMB, Maraweg 21, Bielefeld, Germany
| | - Morris Beshay
- Department of General Thoracic Surgery, Protestant Hospital of Bethel Foundation, Burgsteig 13
- Forschungsverbund BioMedizin Bielefeld, FBMB, Maraweg 21, Bielefeld, Germany
| |
Collapse
|
634
|
Zhou J, Cheng K, Chen X, Yang R, Lu M, Ming L, Chen Y, Lin Z, Chen D. Determination of soluble CD44 in serum by using a label-free aptamer based electrochemical impedance biosensor. Analyst 2019; 145:460-465. [PMID: 31781712 DOI: 10.1039/c9an01764j] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CD44 is a promising biomarker in the diagnosis and prognosis of malignancies. The serum CD44 level is closely related to disease progression and metastasis of malignancies. It is of great clinical significance for the detection of serum soluble CD44. In this study, a facile, label-free aptamer based electrochemical impedance sensor for serum CD44 has been proposed. The aptamer showing high affinity to CD44 was immobilized on the gold electrodes through Au-S interaction. The interaction between target CD44 and the immobilized aptamer will cause a complex structure change of the aptamer, which makes the diffusion of [Fe(CN)6]3-/4- toward the electrode surface easy, thus resulting in the decrease of the impedance of the system. The decreased degree of the impedance had a good linear relationship with the logarithm of the CD44 concentration in the range of 0.1-1000 ng mL-1 with a detection limit of 0.087 ng mL-1 (S/N = 3). The developed biosensor has been applied to detect CD44 in serum samples with satisfactory results.
Collapse
Affiliation(s)
- Jie Zhou
- Department of central laboratory, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, Jiangsu 214002, China.
| | - Kai Cheng
- Department of central laboratory, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, Jiangsu 214002, China.
| | - Xuan Chen
- Kangda college of Nanjing medical university, Lianyungang, Jiangsu 222000, China
| | - Rui Yang
- Department of central laboratory, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, Jiangsu 214002, China.
| | - Mudan Lu
- Department of central laboratory, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, Jiangsu 214002, China.
| | - Lan Ming
- Department of central laboratory, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, Jiangsu 214002, China.
| | - Yu Chen
- Department of central laboratory, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, Jiangsu 214002, China.
| | - Zhenyu Lin
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China.
| | - Daozhen Chen
- Department of central laboratory, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, Jiangsu 214002, China.
| |
Collapse
|
635
|
Vincent A, Ouelkdite-Oumouchal A, Souidi M, Leclerc J, Neve B, Van Seuningen I. Colon cancer stemness as a reversible epigenetic state: Implications for anticancer therapies. World J Stem Cells 2019; 11:920-936. [PMID: 31768220 PMCID: PMC6851010 DOI: 10.4252/wjsc.v11.i11.920] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 08/29/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023] Open
Abstract
The recent discovery of cancer cell plasticity, i.e. their ability to reprogram into cancer stem cells (CSCs) either naturally or under chemotherapy and/or radiotherapy, has changed, once again, the way we consider cancer treatment. If cancer stemness is a reversible epigenetic state rather than a genetic identity, opportunities will arise for therapeutic strategies that remodel epigenetic landscapes of CSCs. However, the systematic use of DNA methyltransferase and histone deacetylase inhibitors, alone or in combination, in advanced solid tumors including colorectal cancers, regardless of their molecular subtypes, does not seem to be the best strategy. In this review, we first summarize the knowledge researchers have gathered on the epigenetic signatures of CSCs with the difficulty of isolating rare populations of cells. We raise questions about the relevant use of currently available epigenetic inhibitors (epidrugs) while the expression of numerous cancer stem cell markers are often repressed by epigenetic mechanisms. These markers include the three cluster of differentiation CD133, CD44 and CD166 that have been extensively used for the isolation of colon CSCs.and . Finally, we describe current treatment strategies using epidrugs, and we hypothesize that, using correlation tools comparing associations of relevant CSC markers with chromatin modifier expression, we could identify better candidates for epienzyme targeting.
Collapse
Affiliation(s)
- Audrey Vincent
- Lille University, Institut National de la Santé et de la Recherche Médicale, CHU Lille, UMR-S 1172-Jean-Pierre Aubert Research Center, Lille F-59000, France
| | - Aïcha Ouelkdite-Oumouchal
- Lille University, Institut National de la Santé et de la Recherche Médicale, CHU Lille, UMR-S 1172-Jean-Pierre Aubert Research Center, Lille F-59000, France
| | - Mouloud Souidi
- Lille University, Institut National de la Santé et de la Recherche Médicale, CHU Lille, UMR-S 1172-Jean-Pierre Aubert Research Center, Lille F-59000, France
| | - Julie Leclerc
- Lille University, Institut National de la Santé et de la Recherche Médicale, CHU Lille, UMR-S 1172-Jean-Pierre Aubert Research Center, Lille F-59000, France
- Department of Biochemistry and Molecular Biology, Lille University Hospital, Lille F-59000, France
| | - Bernadette Neve
- Lille University, Institut National de la Santé et de la Recherche Médicale, CHU Lille, UMR-S 1172-Jean-Pierre Aubert Research Center, Lille F-59000, France
| | - Isabelle Van Seuningen
- Lille University, Institut National de la Santé et de la Recherche Médicale, CHU Lille, UMR-S 1172-Jean-Pierre Aubert Research Center, Lille F-59000, France
| |
Collapse
|
636
|
Lu X, Yang R, Zhang L, Xi Y, Zhao J, Wang F, Zhang H, Li Z. Macrophage Colony-stimulating Factor Mediates the Recruitment of Macrophages in Triple negative Breast Cancer. Int J Biol Sci 2019; 15:2859-2871. [PMID: 31853223 PMCID: PMC6909971 DOI: 10.7150/ijbs.39063] [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: 08/06/2019] [Accepted: 09/17/2019] [Indexed: 12/31/2022] Open
Abstract
Triple negative breast cancer (TNBC) is characterized by aggressive malignant tumor, poor prognosis and lack of targeted treatment. Several studies have established that macrophages are closely associated with the progression of TNBC. Through immunohistochemical analysis, we found that the infiltration of macrophage in TNBC tissue was more than that in non-triple negative breast cancer (nTNBC) tissue. Furthermore, the conditioned medium (CM) of MDA-MB-231 and HCC1937, the TNBC cell lines, had significant migration-promoted effect on macrophages. However, the macrophages migration-promoted ability of nTNBC cell line MCF-7 was weaker than that of MDA-MB-231 and HCC1937 cells. Mechanistically, MDA-MB-231 and HCC1937 cells secreted more macrophage colony-stimulating factor (M-CSF) than MCF-7, which is the main inducer of macrophage migration, and the secreted M-CSF promoted the increase in actin and the elongation of pseudopodia. When M-CSF was neutralized by antibody, the elongation of macrophage pseudopodia was disappeared and the migration was inhibited. In vivo, there were more macrophages in tumors induced by MDA-MB-231 than MCF-7 did. Therefore, M-CSF specially secreted by TNBC was the important cause of macrophages aggregation in TNBC, which further promoted the aggressiveness of TNBC.
Collapse
Affiliation(s)
- Xiaoqing Lu
- Breast surgery, the second Hospital of Shanxi Medical University, Taiyuan 030001,
| | - Rui Yang
- The second Clinical Medical College, Shanxi Medical University, Taiyuan 030001,
| | - Lichao Zhang
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China.
| | - Yanfeng Xi
- Department of Pathology, Shanxi Cancer Hospital, Taiyuan 030001, China.
| | - Jiping Zhao
- Breast surgery, the second Hospital of Shanxi Medical University, Taiyuan 030001,
| | - Fusheng Wang
- Breast surgery, the second Hospital of Shanxi Medical University, Taiyuan 030001, China. . 13099096632
| | - Huanhu Zhang
- Department of Digestive Sciences, Shanxi Cancer Hospital, Taiyuan 030001, China. . 18035119990
| | - Zhuoyu Li
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan, 030006, China. . 13934565188.,School of Life Science, Shanxi University, Taiyuan, 030006, China. . 13934565188
| |
Collapse
|
637
|
Fänder J, Kielstein H, Büttner M, Koelblinger P, Dummer R, Bauer M, Handke D, Wickenhauser C, Seliger B, Jasinski-Bergner S. Characterizing CD44 regulatory microRNAs as putative therapeutic agents in human melanoma. Oncotarget 2019; 10:6509-6525. [PMID: 31741714 PMCID: PMC6849650 DOI: 10.18632/oncotarget.27305] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/21/2019] [Indexed: 01/22/2023] Open
Abstract
The multistructural and multifunctional transmembrane glycoprotein CD44 is overexpressed in many tumors of distinct origin including malignant melanoma and contributes to a poor prognosis by affecting cell proliferation, cell migration, and also the sensitivity for apoptosis induction. Previous studies reported so far 15 CD44 regulatory microRNAs (miRs) in different cell systems. Using a novel method for miR affinity purification miR-143-3p was identified as most potent binder to the 3' untranslated region (UTR) of CD44. Overexpression of miR-143-3p in melanoma cells inhibits CD44 translation, which is accompanied by a reduced proliferation, migration and enhanced daunorubicin induced apoptosis of melanoma cells in vitro. Analyses of discordant CD44 and miR-143-3p expression levels in human melanocytic nevi and dermal melanoma samples demonstrated medium to high CD44 levels with no association to tumor grading or staging. The CD44 expression correlated to PD-L1, but not to MART-1 expression in malignant melanoma. Interestingly, the CD44 expression was inversely correlated to the infiltration of pro-inflammatory immune effector cells. In conclusion, the tumor suppressive miR-143-3p was identified as the most potent CD44 inhibitory miR, which affects growth characteristics of melanoma cells suggesting the implementation of miR-143-3p as as a potential anti-CD44 therapy of malignant melanoma.
Collapse
Affiliation(s)
- Johannes Fänder
- Institute of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Heike Kielstein
- Institute of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Maximilian Büttner
- Institute of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Peter Koelblinger
- Department of Dermatology, Paracelsus Medical University, Salzburg, Austria
| | - Reinhard Dummer
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | - Marcus Bauer
- Institute for Pathology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Diana Handke
- Institute for Medical Immunology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Claudia Wickenhauser
- Institute for Pathology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Barbara Seliger
- Institute for Medical Immunology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Simon Jasinski-Bergner
- Institute of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany.,Institute for Medical Immunology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
638
|
Hou C, Ishi Y, Motegi H, Okamoto M, Ou Y, Chen J, Yamaguchi S. Overexpression of CD44 is associated with a poor prognosis in grade II/III gliomas. J Neurooncol 2019; 145:201-210. [PMID: 31506754 DOI: 10.1007/s11060-019-03288-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/05/2019] [Indexed: 02/05/2023]
Abstract
PURPOSE Overexpression of CD44 has been detected in many types of tumor tissues. Moreover, CD44 is recognized as a cancer stem cell marker for many cancers. However, the prognostic value of CD44 for glioma patients has not yet been clarified. The authors tried to explore the impact of CD44 expression on grade II/III glioma patients. METHODS To assess the RNA expression levels of CD44 in glioma tissues and normal brain tissues, meta-analyses were conducted in the online Oncomine database. The mRNA expression levels of CD44, CD44s, and CD44v2-v10 in 112 grade II/III glioma patients in Hokkaido University Hospital (HUH) were detected by qPCR. The RNA-seq data and clinical data of grade II/III glioma patients were obtained from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA) databases. RESULTS Based on the Oncomine database, CD44 has significantly high expression in glioma tissues as compared with normal tissues. We explored the clinical relevance of CD44 mRNA expression based on the HUH cohorts, the TCGA cohorts, and the CGGA cohorts. In survival analysis, high mRNA expression of CD44 was correlated with poor overall survival and poor progression-free survival in grade II/III glioma patients. Multivariate Cox regression analyses confirmed CD44 as an independent prognostic factor for grade II/III glioma patients. CONCLUSIONS The present study suggests that overexpression of CD44 is associated with a poor prognosis for grade II/III glioma patients. Moreover, our findings suggest that CD44 could serve as a prognostic biomarker in grade II/III glioma patients.
Collapse
Affiliation(s)
- Chongxian Hou
- Department of Neurosurgery, Faculty of Medicine, Hokkaido University, North 15 West 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Yukitomo Ishi
- Department of Neurosurgery, Faculty of Medicine, Hokkaido University, North 15 West 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Hiroaki Motegi
- Department of Neurosurgery, Faculty of Medicine, Hokkaido University, North 15 West 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Michinari Okamoto
- Department of Neurosurgery, Faculty of Medicine, Hokkaido University, North 15 West 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Yafei Ou
- Graduate School of Information Science and Technology, Hokkaido University, Sapporo, 060-0814, Japan
| | - Jiawei Chen
- Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Shigeru Yamaguchi
- Department of Neurosurgery, Faculty of Medicine, Hokkaido University, North 15 West 7, Kita-ku, Sapporo, 060-8638, Japan.
| |
Collapse
|
639
|
A CTC-Cluster-Specific Signature Derived from OMICS Analysis of Patient-Derived Xenograft Tumors Predicts Outcomes in Basal-Like Breast Cancer. J Clin Med 2019; 8:jcm8111772. [PMID: 31652963 PMCID: PMC6912280 DOI: 10.3390/jcm8111772] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/12/2019] [Accepted: 10/21/2019] [Indexed: 12/24/2022] Open
Abstract
Circulating tumor cell clusters (CTCcl) have a higher metastatic potential compared to single CTCs and predict long-term outcomes in breast cancer (BC) patients. Because of the rarity of CTCcls, molecular characterization of primary tumors that give rise to CTCcl hold significant promise for better diagnosis and target discovery to combat metastatic BC. In our study, we utilized the reverse-phase protein array (RPPA) and transcriptomic (RNA-Seq) data of 10 triple-negative BC patient-derived xenograft (TNBC PDX) transplantable models with CTCs and evaluated expression of upregulated candidate protein Bcl2 (B-cell lymphoma 2) by immunohistochemistry (IHC). The sample-set consisted of six CTCcl-negative (CTCcl-) and four CTCcl-positive (CTCcl+) models. We analyzed the RPPA and transcriptomic profiles of CTCcl- and CTCcl+ TNBC PDX models. In addition, we derived a CTCcl-specific gene signature for testing if it predicted outcomes using a publicly available dataset from 360 patients with basal-like BC. The RPPA analysis of CTCcl+ vs. CTCcl- TNBC PDX tumors revealed elevated expression of Bcl2 (false discovery rate (FDR) < 0.0001, fold change (FC) = 3.5) and reduced acetyl coenzyme A carboxylase-1 (ACC1) (FDR = 0.0005, FC = 0.3) in CTCcl+ compared to CTCcl- tumors. Genome-wide transcriptomic analysis of CTCcl+ vs. CTCcl- tumors revealed 549 differentially expressed genes associated with the presence of CTCcls. Apoptosis was one of the significantly downregulated pathways (normalized enrichment score (NES) = -1.69; FDR < 0.05) in TNBC PDX tumors associated with CTCcl positivity. Two out of four CTCcl+ TNBC PDX primary tumors had high Bcl2 expression by IHC (H-score > 34); whereas, only one of six CTCcl- TNBC PDX primary tumors met this criterion. Evaluation of epithelial-mesenchymal transition (EMT)-specific signature did not show significant differences between CTCcl+ and CTCcl- tumors. However, a gene signature associated with the presence of CTCcls in TNBC PDX models was associated with worse relapse-free survival in the publicly available dataset from 360 patients with basal-like BC. In summary, we identified the multigene signature of primary PDX tumors associated with the presence of CTCcls. Evaluation of additional TNBC PDX models and patients can further illuminate cellular and molecular pathways facilitating CTCcl formation.
Collapse
|
640
|
Wang C, Shi M, Ji J, Cai Q, Jiang J, Zhang H, Zhu Z, Zhang J. A self-enforcing HOXA11/Stat3 feedback loop promotes stemness properties and peritoneal metastasis in gastric cancer cells. Theranostics 2019; 9:7628-7647. [PMID: 31695791 PMCID: PMC6831465 DOI: 10.7150/thno.36277] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/05/2019] [Indexed: 12/13/2022] Open
Abstract
Rationale: Peritoneal metastasis is one of the most common and life-threatening metastases in gastric cancer patients. The disseminated gastric cancer cells forming peritoneal metastasis exhibit a variety of characteristics that contrast with those of adjacent epithelial cell of gastric mucosa and even primary gastric cancer cells. We hypothesized that the gene expression profiles of peritoneal foci could reveal the identities of genes that might function as metastatic activator. Methods: In this study, we show, using in vitro, in vivo, in silico and gastric cancer tissues studies in humans and mice, that Homoebox A11 (HOXA11) potently promote peritoneal metastasis of gastric cancer cells. Results: Its mechanism of action involves alternation of cancer stemness and subsequently enhancement of the adhesion, migration and invasion and anti-apoptosis. This is achieved, mainly, through formation of a positive feedback loop between HOXA11 and Stat3, which is involved in the stimulation of Stat3 signaling pathway. Conclusions: These observations uncover a novel peritoneal metastatic activator and demonstrate the association between HOXA11, Stat3 and cancer stemness of gastric cancer cells, thereby revealing a previously undescribed mechanism of peritoneal metastasis.
Collapse
Affiliation(s)
- Chao Wang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Min Shi
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Jun Ji
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Qu Cai
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Jinling Jiang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Huan Zhang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Zhenggang Zhu
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Ruijin er Road, Shanghai, 200025, China
| |
Collapse
|
641
|
Transcriptional profiling of circulating tumor cells in multiple myeloma: a new model to understand disease dissemination. Leukemia 2019; 34:589-603. [PMID: 31595039 DOI: 10.1038/s41375-019-0588-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/25/2019] [Accepted: 07/23/2019] [Indexed: 12/28/2022]
Abstract
The reason why a few myeloma cells egress from the bone marrow (BM) into peripheral blood (PB) remains unknown. Here, we investigated molecular hallmarks of circulating tumor cells (CTCs) to identify the events leading to myeloma trafficking into the bloodstream. After using next-generation flow to isolate matched CTCs and BM tumor cells from 32 patients, we found high correlation in gene expression at single-cell and bulk levels (r ≥ 0.94, P = 10-16), with only 55 genes differentially expressed between CTCs and BM tumor cells. CTCs overexpressed genes involved in inflammation, hypoxia, or epithelial-mesenchymal transition, whereas genes related with proliferation were downregulated in CTCs. The cancer stem cell marker CD44 was overexpressed in CTCs, and its knockdown significantly reduced migration of MM cells towards SDF1-α and their adhesion to fibronectin. Approximately half (29/55) of genes differentially expressed in CTCs were prognostic in patients with newly-diagnosed myeloma (n = 553; CoMMpass). In a multivariate analysis including the R-ISS, overexpression of CENPF and LGALS1 was significantly associated with inferior survival. Altogether, these results help understanding the presence of CTCs in PB and suggest that hypoxic BM niches together with a pro-inflammatory microenvironment induce an arrest in proliferation, forcing tumor cells to circulate in PB and seek other BM niches to continue growing.
Collapse
|
642
|
Zou H, Chen H, Zhou Z, Wan Y, Liu Z. ATXN3 promotes breast cancer metastasis by deubiquitinating KLF4. Cancer Lett 2019; 467:19-28. [PMID: 31563563 DOI: 10.1016/j.canlet.2019.09.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/19/2019] [Accepted: 09/24/2019] [Indexed: 12/24/2022]
Abstract
Krüppel-like factor 4 (KLF4) is an important transcription factor implicated in a variety of essential cellular processes. Aberrant KLF4 expression is closely related to tumourigenesis and tumour progression. The rapid turnover of the KLF4 protein indicates an important role for the posttranslational modifications (PTMs) of KLF4. To date, E3 ligases mediating KLF4 ubiquitination have been widely reported, yet the deubiquitinating mechanism of KLF4 remains largely unknown. We screened a library of 65 deubiquitinating enzymes and identified ATXN3 as a deubiquitinating enzyme of KLF4. Subsequent immunoprecipitation assays confirmed that ATXN3 bound to KLF4, mediating the deubiquitination and stabilization of KLF4 protein levels. Furthermore, we demonstrated that ATXN3 promoted breast cancer cell metastasis via KLF4 in vitro and in vivo. Finally, the protein expression analysis of human breast cancer specimens demonstrated that ATXN3 significantly correlated with KLF4. High ATXN3/KLF4 expression was associated with a poor prognosis in breast cancer patients. Collectively, we identified ATXN3 as a novel deubiquitinating enzyme of KLF4, providing a new explanation for breast cancer metastasis, and proposed ATXN3 as a potential target for breast cancer metastasis treatment.
Collapse
Affiliation(s)
- Haojing Zou
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Hongyan Chen
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhuan Zhou
- Department of Obstetrics and Gynecology, Department of Pharmacology, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Yong Wan
- Department of Obstetrics and Gynecology, Department of Pharmacology, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
643
|
Lai HT, Tseng WK, Huang SW, Chao TC, Su Y. MicroRNA-203 diminishes the stemness of human colon cancer cells by suppressing GATA6 expression. J Cell Physiol 2019; 235:2866-2880. [PMID: 31544978 DOI: 10.1002/jcp.29192] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/03/2019] [Indexed: 12/27/2022]
Abstract
The interaction between hyaluronan and CD44, an important cancer stem-cell marker, stimulates various tumor cell-specific functions such as the stemness of tumor cells. microRNA-203 (miR-203) can be downregulated by this interaction in human colorectal cancer (CRC) cells, which increases their stemness; however, the underlying mechanism is not yet defined. Here, we show that overexpression and sequestration of miR-203 in HCT-116 and HT-29 human CRC cells reduces and enhances their stemness, respectively. We also show that GATA-binding factor 6 (GATA6) is a direct target of miR-203. Our results indicate that upregulated expression of this transcription factor not only restores the self-renewal abilities of miR-203-overexpressing HCT-116 and HT-29 cells but also promotes the stemness properties of their parental counterparts. More important, we show that silencing the expression of either LRH-1 or Hes-1 is sufficient to diminish the stemness-promoting effects of GATA6 in human CRC cells. Together, our findings delineate the stemness-inhibitory mechanism of miR-203 in human CRC cells and suggest that this miR is a potential therapeutic agent for colorectal cancer.
Collapse
Affiliation(s)
- Hung-Tzi Lai
- Institute of Biopharmaceutical Sciences, School of Pharmaceutical Sciences, National Yang-Ming University, Shi-Pai, Taipei, Taiwan, R.O.C
| | - Wen-Ko Tseng
- Program in Molecular Medicine, School of Life Sciences, National Yang-Ming University, Shi-Pai, Taipei, Taiwan, R.O.C.,Colorectal Surgery Department, Chung-Gung Memorial Hospital, Keelung Branch, Keelung, Taipei, Taiwan, R.O.C
| | - Shi-Wei Huang
- Institute of Biopharmaceutical Sciences, School of Pharmaceutical Sciences, National Yang-Ming University, Shi-Pai, Taipei, Taiwan, R.O.C
| | - Ta-Chung Chao
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C.,Faculty of Medicine, School of Medicine, National Yang-Ming University, Shi-Pai, Taipei, Taiwan, R.O.C
| | - Yeu Su
- Institute of Biopharmaceutical Sciences, School of Pharmaceutical Sciences, National Yang-Ming University, Shi-Pai, Taipei, Taiwan, R.O.C.,Program in Molecular Medicine, School of Life Sciences, National Yang-Ming University, Shi-Pai, Taipei, Taiwan, R.O.C
| |
Collapse
|
644
|
Wu Q, Wu Z, Bao C, Li W, He H, Sun Y, Chen Z, Zhang H, Ning Z. Cancer stem cells in esophageal squamous cell cancer. Oncol Lett 2019; 18:5022-5032. [PMID: 31612013 PMCID: PMC6781610 DOI: 10.3892/ol.2019.10900] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 05/29/2019] [Indexed: 12/13/2022] Open
Abstract
Cancer stem cells (CSCs) are hypothesized to govern the origin, progression, drug resistance, recurrence and metastasis of human cancer. CSCs have been identified in nearly all types of human cancer, including esophageal squamous cell cancer (ESCC). Four major methods are typically used to isolate or enrich CSCs, including: i) fluorescence-activated cell sorting or magnetic-activated cell sorting using cell-specific surface markers; ii) stem cell markers, including aldehyde dehydrogenase 1 family member A1; iii) side population cell phenotype markers; and iv) microsphere culture methods. ESCC stem cells have been identified using a number of these methods. An increasing number of stem cell signatures and pathways have been identified, which have assisted in the clarification of molecular mechanisms that regulate the stemness of ESCC stem cells. Certain viruses, such as human papillomavirus and hepatitis B virus, are also considered to be important in the formation of CSCs, and there is a crosstalk between stemness and viruses-associated genes/pathways, which may suggest a potential therapeutic strategy for the eradication of CSCs. In the present review, findings are summarized along these lines of inquiry.
Collapse
Affiliation(s)
- Qian Wu
- Basic Medical School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China.,Nurse School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Zhe Wu
- Basic Medical School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Cuiyu Bao
- Nurse School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Wenjing Li
- Basic Medical School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Hui He
- Basic Medical School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Yanling Sun
- Basic Medical School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Zimin Chen
- Basic Medical School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Hao Zhang
- Basic Medical School, Ji'nan University Medical School, Guangzhou, Guangdong 510632, P.R. China
| | - Zhifeng Ning
- Basic Medical School, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| |
Collapse
|
645
|
Johnson BL, d’Alincourt Salazar M, Mackenzie-Dyck S, D’Apuzzo M, Shih HP, Manuel ER, Diamond DJ. Desmoplasia and oncogene driven acinar-to-ductal metaplasia are concurrent events during acinar cell-derived pancreatic cancer initiation in young adult mice. PLoS One 2019; 14:e0221810. [PMID: 31490946 PMCID: PMC6731019 DOI: 10.1371/journal.pone.0221810] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 08/15/2019] [Indexed: 01/04/2023] Open
Abstract
The five-year survival rate of patients diagnosed with advanced pancreatic ductal adenocarcinoma (PDAC) has remained static at <5% despite decades of research. With the exception of erlotinib, clinical trials have failed to demonstrate the benefit of any targeted therapy for PDAC despite promising results in preclinical animal studies. The development of more refined mouse models of PDAC which recapitulate the carcinogenic progression from non-neoplastic, adult exocrine subsets of pancreatic cells to invasive carcinoma in humans are needed to facilitate the accurate translation of therapies to the clinic. To study acinar cell-derived PDAC initiation, we developed a genetically engineered mouse model of PDAC, called KPT, utilizing a tamoxifen-inducible Cre recombinase/estrogen receptor (ESR1) fusion protein knocked into the Ptf1a locus to activate the expression of oncogenic KrasG12D and Trp53R270H alleles in mature pancreatic acinar cells. Oncogene-expressing acinar cells underwent acinar-to-ductal metaplasia, and formed pancreatic intraepithelial neoplasia lesions following the induction of oncogene expression. After a defined latency period, oncogene-expressing acinar cells initiated the formation of highly differentiated and fibrotic tumors, which metastasized to the lungs and liver. Whole-transcriptome analysis of microdissected regions of acinar-to-ductal metaplasia and histological validation experiments demonstrated that regions of acinar-to-ductal metaplasia are characterized by the deposition of the extracellular matrix component hyaluronan. These results indicate that acinar cells expressing KrasG12D and Trp53R270H can initiate PDAC development in young adult mice and implicate hyaluronan deposition in the formation of the earliest characterized PDAC precursor lesions (and the progression of pancreatic cancer). Further studies are necessary to provide a comprehensive characterization of PDAC progression and treatment response in KPT mice and to investigate whether the KPT model could be used as a tool to study translational aspects of acinar cell-derived PDAC tumorigenesis.
Collapse
Affiliation(s)
- Benjamin L. Johnson
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute of City of Hope, Duarte, CA, United States of America
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, United States of America
| | - Marcela d’Alincourt Salazar
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute of City of Hope, Duarte, CA, United States of America
| | - Sarah Mackenzie-Dyck
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute of City of Hope, Duarte, CA, United States of America
| | - Massimo D’Apuzzo
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, CA, United States of America
| | - Hung Ping Shih
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolic Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, United States of America
| | - Edwin R. Manuel
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States of America
| | - Don J. Diamond
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute of City of Hope, Duarte, CA, United States of America
- * E-mail:
| |
Collapse
|
646
|
Mori T, Katayama Y. Signal amplification in flow cytometry for cell surface antigen analysis. J Biochem 2019; 166:205-212. [PMID: 31251348 DOI: 10.1093/jb/mvz052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 06/25/2019] [Indexed: 01/12/2023] Open
Abstract
Signal enhancing systems have been introduced to enable detection of cell surface antigens by flow cytometry. Cell surface antigens are important targets that describe the function and lineage of cells. Although flow cytometry is an effective tool for analysing cell surface antigens, this technique has poor sensitivity, which prohibits the detection of many important antigens on cell membranes. Thus, signal amplification is essential for developing practical tools for evaluating cell surface antigens by flow cytometry. Using a bright fluorophore or fluorescent polymer incorporated into antibodies is a straightforward strategy to improve flow cytometry sensitivity but may affect the functional characteristics of the labelled antibody. In contrast, enzymatic signal amplification is a more practical and efficient strategy to improve sensitivity that should not affect antibody activity. Although enzymatic signal amplification still has a number of drawbacks, this approach is a promising strategy to analyse cell surface antigens.
Collapse
Affiliation(s)
- Takeshi Mori
- Faculty of Engineering, Department of Chemistry and Biochemistry, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, Japan
| | - Yoshiki Katayama
- Faculty of Engineering, Department of Chemistry and Biochemistry, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, Japan
| |
Collapse
|
647
|
Wang Q, Xu J, Chen Y, Liu L. KLF4 overexpression decreases the viability, invasion and migration of papillary thyroid cancer cells. Exp Ther Med 2019; 18:3493-3501. [PMID: 31602225 PMCID: PMC6777314 DOI: 10.3892/etm.2019.7969] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 06/06/2019] [Indexed: 12/24/2022] Open
Abstract
Kruppel-like factor 4 (KLF4) has been implicated in a number of different types of cancer; however, the role of KLF4 in papillary thyroid cancer remains elusive. The present study aimed to investigate the role of KLF4 in papillary thyroid cancer and its potential underlying molecular mechanisms. The expression of KLF4 in thyroid tumor tissue and adjacent non-cancerous tissues were detected via immunohistochemistry and western blotting. The papillary thyroid cancer cell line, KTC1, was transfected with viruses carrying KLF4 overexpression vectors. The relative expression of KLF4, E-cadherin, N-cadherin, Vimentin, matrix metalloproteinase (MMP)2, MMP9 and collagen was detected via quantitative-PCR. The viability of KTC1 cells was detected using a cell counting kit-8 assay at 24, 48 and 72 h. Cell invasion was examined via a transwell invasion assay. Cell migration was examined via a scratch migration assay at 0 and 24 h. Compared with adjacent non-cancerous tissues, the expression of KLF4 was significantly lower in thyroid tumor tissues. The expression of KLF4 in KTC1 cells were significantly increased compared with the blank or negative control groups. The expression of N-cadherin, MMP2, MMP9 and collagen was significantly decreased in the KLF4 overexpression group. The viability of KTC1 cells was markedly decreased in KLF4 overexpression group at 24, 48 and 72 h when compared with the blank or negative control groups. The invasion of KTC1 cells in the KLF4 overexpression group was markedly decreased. Compared with the negative control group, the KTC1 cell migration in the KLF4 overexpression group was markedly decreased at 24 h. The expression of KLF4 was also significantly lower in thyroid tumor tissues. The cell viability, tumor invasion and migration ability and expression levels of N-cadherin, MMP2, MMP9 and collagen in papillary thyroid cancer cells were markedly decreased with KLF4 overexpression.
Collapse
Affiliation(s)
- Qianzhu Wang
- Department of General Surgery, Baoshan District Integrated Traditional Chinese and Western Medicine Hospital, Shanghai 201999, P.R. China
| | - Jian Xu
- Department of General Surgery, Baoshan District Integrated Traditional Chinese and Western Medicine Hospital, Shanghai 201999, P.R. China
| | - Yong Chen
- Department of General Surgery, Baoshan District Integrated Traditional Chinese and Western Medicine Hospital, Shanghai 201999, P.R. China
| | - Limin Liu
- Department of General Surgery, Baoshan District Integrated Traditional Chinese and Western Medicine Hospital, Shanghai 201999, P.R. China
| |
Collapse
|
648
|
Ibrahim HM, AbdElbary AM, Mohamed SY, Elwan A, Abdelhamid MI, Ibrahim A. Prognostic Value of Cyclin D1 and CD44 Expression in Gastric Adenocarcinoma. J Gastrointest Cancer 2019; 50:370-379. [PMID: 29497929 DOI: 10.1007/s12029-018-0079-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Worldwide, gastric carcinoma (GC) is the 5th most common malignancies in both sexes representing 6.8% of the total fatalities and is the 3rd leading cause of cancer death representing 8.8% of total fatalities. In Egypt, GC considers the 12th leading cause of cancer death representing 2.2% of the total cancer mortality. A growing body of evidence supports that cancer stem cells (CSCs) are resistant to chemotherapy or radiation, and the cell adhesion molecule CD44 has been identified as a cell surface marker associated with cancer stem cell in several types of tumors including gastric cancer. CD44 regulates gastric stem cell proliferation by increasing cyclin D1 expression which represents an important regulatory protein in the cell cycle transition from G1 phase to S phase. This study aimed to investigate whether cyclin D1 and CD44 can be used as prognostic indicators in gastric cancer. MATERIAL AND METHODS Forty formalin-fixed and paraffin-embedded gastric tissues, obtained from patients who underwent endoscopic resection or surgical resection, constituted the group of our study. The immunohistochemical expression of cyclin D1 and CD44 was examined and correlated with clinical-pathological parameters and outcome of the patients. RESULTS Overexpression of CD44 and cyclin D1 was noted (in of 55 and 50% respectively). Cyclin D1 and CD44 positive expressions in GC were positively correlated with tumor differentiation (p = 0.020, p = 0.004 respectively), TNM stage (p < 0.001 for both), poor survival (p < 0.001 for both), and with increased rate of recurrence (p = 0.020, p = 0.005 respectively). CONCLUSION CD44 and cyclin D1 were associated with poor prognosis in gastric cancer, and so, they comprise an attractive target for anticancer drug development.
Collapse
Affiliation(s)
- Hanaa M Ibrahim
- Pathology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Abeer M AbdElbary
- Pathology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Salem Y Mohamed
- Internal Medicine Department, Gastroenterology and Hepatology Unit, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Amira Elwan
- Clinical Oncology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mohamed I Abdelhamid
- General Surgery Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Amr Ibrahim
- General Surgery Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
649
|
Bayat Mokhtari R, Baluch N, Morgatskaya E, Kumar S, Sparaneo A, Muscarella LA, Zhao S, Cheng HL, Das B, Yeger H. Human bronchial carcinoid tumor initiating cells are targeted by the combination of acetazolamide and sulforaphane. BMC Cancer 2019; 19:864. [PMID: 31470802 PMCID: PMC6716820 DOI: 10.1186/s12885-019-6018-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 08/06/2019] [Indexed: 12/19/2022] Open
Abstract
Background Bronchial carcinoids are neuroendocrine tumors that present as typical (TC) and atypical (AC) variants, the latter being more aggressive, invasive and metastatic. Studies of tumor initiating cell (TIC) biology in bronchial carcinoids has been hindered by the lack of appropriate in-vitro and xenograft models representing the bronchial carcinoid phenotype and behavior. Methods Bronchial carcinoid cell lines (H727, TC and H720, AC) were cultured in serum-free growth factor supplemented medium to form 3D spheroids and serially passaged up to the 3rd generation permitting expansion of the TIC population as verified by expression of stemness markers, clonogenicity in-vitro and tumorigenicity in both subcutaneous and orthotopic (lung) models. Acetazolamide (AZ), sulforaphane (SFN) and the AZ + SFN combination were evaluated for targeting TIC in bronchial carcinoids. Results Data demonstrate that bronchial carcinoid cell line 3rd generation spheroid cells show increased drug resistance, clonogenicity, and tumorigenic potential compared with the parental cells, suggesting selection and expansion of a TIC fraction. Gene expression and immunolabeling studies demonstrated that the TIC expressed stemness factors Oct-4, Sox-2 and Nanog. In a lung orthotopic model bronchial carcinoid, cell line derived spheroids, and patient tumor derived 3rd generation spheroids when supported by a stroma, showed robust tumor formation. SFN and especially the AZ + SFN combination were effective in inhibiting tumor cell growth, spheroid formation and in reducing tumor formation in immunocompromised mice. Conclusions Human bronchial carcinoid tumor cells serially passaged as spheroids contain a higher fraction of TIC exhibiting a stemness phenotype. This TIC population can be effectively targeted by the combination of AZ + SFN. Our work portends clinical relevance and supports the therapeutic use of the novel AZ+ SFN combination that may target the TIC population of bronchial carcinoids.
Collapse
Affiliation(s)
- Reza Bayat Mokhtari
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada. .,Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, Canada. .,The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay St., Rm 15.9714, Toronto, Ontario, M5G 0A4, Canada.
| | - Narges Baluch
- Department of Pediatrics, Queen's University, 76 Stuart St, Kingston, ON, K7L 2V7, Canada
| | - Evgeniya Morgatskaya
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Sushil Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Williams Science Hall 3035, Department of Pharmaceutical Sciences 601 S. Saddle Creek Rd, Omaha, NE, 68106, USA
| | - Angelo Sparaneo
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, viale Cappuccini, 71013, San Giovanni Rotondo, FG, Italy
| | - Lucia Anna Muscarella
- Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, viale Cappuccini, 71013, San Giovanni Rotondo, FG, Italy
| | - Sheyun Zhao
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Hai-Ling Cheng
- Institute of Biomaterials & Biomedical Engineering, University of Toronto, 164 College Street, Rosebrugh Building, Room 407, Toronto, ON, M5S 3G9, Canada
| | - Bikul Das
- Thoreau Laboratory for Global Health, M2D2, University of Massachusetts-Lowell, Innovation Hub, 110 Canal St, Lowell, MA, 01852, USA.,KaviKrishna Laboratory, Indian Institute of Technology Complex, Guwahati, India
| | - Herman Yeger
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay St., Rm 15.9714, Toronto, Ontario, M5G 0A4, Canada
| |
Collapse
|
650
|
Sirkisoon SR, Carpenter RL, Rimkus T, Doheny D, Zhu D, Aguayo NR, Xing F, Chan M, Ruiz J, Metheny-Barlow LJ, Strowd R, Lin J, Regua AT, Arrigo A, Anguelov M, Pasche B, Debinski W, Watabe K, Lo HW. TGLI1 transcription factor mediates breast cancer brain metastasis via activating metastasis-initiating cancer stem cells and astrocytes in the tumor microenvironment. Oncogene 2019; 39:64-78. [PMID: 31462709 PMCID: PMC6938539 DOI: 10.1038/s41388-019-0959-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/24/2019] [Accepted: 07/25/2019] [Indexed: 11/24/2022]
Abstract
Mechanisms for breast cancer metastasis remain unclear. Whether truncated glioma-associated oncogene homolog 1 (TGLI1), a transcription factor known to promote angiogenesis, migration and invasion, plays any role in metastasis of any tumor type has never been investigated. In this study, results of two mouse models of breast cancer metastasis showed that ectopic expression of TGLI1, but not GLI1, promoted preferential metastasis to the brain. Conversely, selective TGLI1 knockdown using antisense oligonucleotides led to decreased breast cancer brain metastasis (BCBM) in vivo. Immunohistochemical staining showed that TGLI1, but not GLI1, was increased in lymph node metastases compared to matched primary tumors, and that TGLI1 was expressed at higher levels in BCBM specimens compared to primary tumors. TGLI1 activation is associated with a shortened time to develop BCBM and enriched in HER2-enriched and triple-negative breast cancers. Radioresistant BCBM cell lines and specimens expressed higher levels of TGLI1, but not GLI1, than radiosensitive counterparts. Since cancer stem cells (CSCs) are radioresistant and metastasis-initiating cells, we examined TGLI1 for its involvement in breast CSCs and found TGLI1 to transcriptionally activate stemness genes CD44, Nanog, Sox2, and OCT4 leading to CSC renewal, and TGLI1 outcompetes with GLI1 for binding to target promoters. We next examined whether astrocyte-priming underlies TGLI1-mediated brain tropism and found that TGLI1-positive CSCs strongly activated and interacted with astrocytes in vitro and in vivo. These findings demonstrate, for the first time, that TGLI1 mediates breast cancer metastasis to the brain, in part, through promoting metastasis-initiating CSCs and activating astrocytes in BCBM microenvironment.
Collapse
Affiliation(s)
- Sherona R Sirkisoon
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Richard L Carpenter
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Bloomington, IN, USA
| | - Tadas Rimkus
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Daniel Doheny
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Dongqin Zhu
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Noah R Aguayo
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Fei Xing
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA.,Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Michael Chan
- Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA.,Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Jimmy Ruiz
- Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA.,Department of Hematology and Oncology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Linda J Metheny-Barlow
- Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA.,Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Roy Strowd
- Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA.,Department of Neurology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Jiayuh Lin
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Angelina T Regua
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Austin Arrigo
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Marlyn Anguelov
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Boris Pasche
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA.,Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Waldemar Debinski
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA.,Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA.,Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Hui-Wen Lo
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA. .,Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|