151
|
Goulet CR, Pouliot F. TGFβ Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1270:89-105. [PMID: 33123995 DOI: 10.1007/978-3-030-47189-7_6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Transforming growth factor beta (TGFβ) is a pleiotropic growth factor. Under normal physiological conditions, TGFβ maintains homeostasis in mammalian tissues by restraining the growth of cells and stimulating apoptosis. However, the role of TGFβ signaling in the carcinogenesis is complex. TGFβ acts as a tumor suppressor in the early stages of disease and as a tumor promoter in its later stages where cancer cells have been relieved from TGFβ growth controls. Overproduction of TGFβ by cancer cells lead to a local fibrotic and immune-suppressive microenvironment that fosters tumor growth and correlates with invasive and metastatic behavior of the cancer cells. Here, we present an overview of the complex biology of the TGFβ family, and we discuss the roles of TGFβ signaling in carcinogenesis and how this knowledge is being leveraged to develop TGFβ inhibition therapies against the tumor.
Collapse
Affiliation(s)
- Cassandra Ringuette Goulet
- Oncology Division, CHU de Québec Research Center, Quebec, QC, Canada
- Department of Surgery, Faculty of Medicine, Laval University, Quebec, QC, Canada
| | - Frédéric Pouliot
- Oncology Division, CHU de Québec Research Center, Quebec, QC, Canada.
- Department of Surgery, Faculty of Medicine, Laval University, Quebec, QC, Canada.
- Department of surgery, CHU de Québec Research Center - Laval University, Quebec City, QC, Canada.
| |
Collapse
|
152
|
Extracellular Vesicles Orchestrate Immune and Tumor Interaction Networks. Cancers (Basel) 2020; 12:cancers12123696. [PMID: 33317058 PMCID: PMC7763968 DOI: 10.3390/cancers12123696] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/29/2022] Open
Abstract
Simple Summary Significant strides have been made to describe the pervasive role of extracellular vesicles (EVs) in health and disease. This work provides an insightful and unifying mechanistic understanding of EVs in immunity and tumorigenesis. This is achieved by dissecting the role of EVs within the continuum of immune cell physiology, immune–infection responses, and the immune–tumor microenvironment. Our work synthesizes important topical findings on immune EV signaling in mediating immune–tumor interaction networks. Abstract Extracellular vesicles (EVs) are emerging as potent and intricate intercellular communication networks. From their first discovery almost forty years ago, several studies have bolstered our understanding of these nano-vesicular structures. EV subpopulations are now characterized by differences in size, surface markers, cargo, and biological effects. Studies have highlighted the importance of EVs in biology and intercellular communication, particularly during immune and tumor interactions. These responses can be equally mediated at the proteomic and epigenomic levels through surface markers or nucleic acid cargo signaling, respectively. Following the exponential growth of EV studies in recent years, we herein synthesize new aspects of the emerging immune–tumor EV-based intercellular communications. We also discuss the potential role of EVs in fundamental immunological processes under physiological conditions, viral infections, and tumorigenic conditions. Finally, we provide insights on the future prospects of immune–tumor EVs and suggest potential avenues for the use of EVs in diagnostics and therapeutics.
Collapse
|
153
|
Kasprzak A. Angiogenesis-Related Functions of Wnt Signaling in Colorectal Carcinogenesis. Cancers (Basel) 2020; 12:cancers12123601. [PMID: 33276489 PMCID: PMC7761462 DOI: 10.3390/cancers12123601] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Angiogenesis belongs to the most clinical characteristics of colorectal cancer (CRC) and is strongly linked to the activation of Wnt/β-catenin signaling. The most prominent factors stimulating constitutive activation of this pathway, and in consequence angiogenesis, are genetic alterations (mainly mutations) concerning APC and the β-catenin encoding gene (CTNNB1), detected in a large majority of CRC patients. Wnt/β-catenin signaling is involved in the basic types of vascularization (sprouting and nonsprouting angiogenesis), vasculogenic mimicry as well as the formation of mosaic vessels. The number of known Wnt/β-catenin signaling components and other pathways interacting with Wnt signaling, regulating angiogenesis, and enabling CRC progression continuously increases. This review summarizes the current knowledge about the role of the Wnt/Fzd/β-catenin signaling pathway in the process of CRC angiogenesis, aiming to improve the understanding of the mechanisms of metastasis as well as improvements in the management of this cancer. Abstract Aberrant activation of the Wnt/Fzd/β-catenin signaling pathway is one of the major molecular mechanisms of colorectal cancer (CRC) development and progression. On the other hand, one of the most common clinical CRC characteristics include high levels of angiogenesis, which is a key event in cancer cell dissemination and distant metastasis. The canonical Wnt/β-catenin downstream signaling regulates the most important pro-angiogenic molecules including vascular endothelial growth factor (VEGF) family members, matrix metalloproteinases (MMPs), and chemokines. Furthermore, mutations of the β-catenin gene associated with nuclear localization of the protein have been mainly detected in microsatellite unstable CRC. Elevated nuclear β-catenin increases the expression of many genes involved in tumor angiogenesis. Factors regulating angiogenesis with the participation of Wnt/β-catenin signaling include different groups of biologically active molecules including Wnt pathway components (e.g., Wnt2, DKK, BCL9 proteins), and non-Wnt pathway factors (e.g., chemoattractant cytokines, enzymatic proteins, and bioactive compounds of plants). Several lines of evidence argue for the use of angiogenesis inhibition in the treatment of CRC. In the context of this paper, components of the Wnt pathway are among the most promising targets for CRC therapy. This review summarizes the current knowledge about the role of the Wnt/Fzd/β-catenin signaling pathway in the process of CRC angiogenesis, aiming to improve the understanding of the mechanisms of metastasis as well as improvements in the management of this cancer.
Collapse
Affiliation(s)
- Aldona Kasprzak
- Department of Histology and Embryology, Poznan University of Medical Sciences, Swiecicki Street 6, 60-781 Poznań, Poland
| |
Collapse
|
154
|
Pietrobono D, Giacomelli C, Marchetti L, Martini C, Trincavelli ML. High Adenosine Extracellular Levels Induce Glioblastoma Aggressive Traits Modulating the Mesenchymal Stromal Cell Secretome. Int J Mol Sci 2020; 21:E7706. [PMID: 33081024 PMCID: PMC7589183 DOI: 10.3390/ijms21207706] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/01/2020] [Accepted: 10/16/2020] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma is an aggressive, fast-growing brain tumor influenced by the composition of the tumor microenvironment (TME) in which mesenchymal stromal cell (MSCs) play a pivotal role. Adenosine (ADO), a purinergic signal molecule, can reach up to high micromolar concentrations in TME. The activity of specific adenosine receptor subtypes on glioma cells has been widely explored, as have the effects of MSCs on tumor progression. However, the effects of high levels of ADO on glioma aggressive traits are still unclear as is its role in cancer cells-MSC cross-talk. Herein, we first studied the role of extracellular Adenosine (ADO) on isolated human U343MG cells as a glioblastoma cellular model, finding that at high concentrations it was able to prompt the gene expression of Snail and ZEB1, which regulate the epithelial-mesenchymal transition (EMT) process, even if a complete transition was not reached. These effects were mediated by the induction of ERK1/2 phosphorylation. Additionally, ADO affected isolated bone marrow derived MSCs (BM-MSCs) by modifying the pattern of secreted inflammatory cytokines. Then, the conditioned medium (CM) of BM-MSCs stimulated with ADO and a co-culture system were used to investigate the role of extracellular ADO in GBM-MSC cross-talk. The CM promoted the increase of glioma motility and induced a partial phenotypic change of glioblastoma cells. These effects were maintained when U343MG cells and BM-MSCs were co-cultured. In conclusion, ADO may affect glioma biology directly and through the modulation of the paracrine factors released by MSCs overall promoting a more aggressive phenotype. These results point out the importance to deeply investigate the role of extracellular soluble factors in the glioma cross-talk with other cell types of the TME to better understand its pathological mechanisms.
Collapse
Affiliation(s)
| | - Chiara Giacomelli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (D.P.); (L.M.); (C.M.); (M.L.T.)
| | | | | | | |
Collapse
|
155
|
Liu YN, Yang JF, Huang DJ, Ni HH, Zhang CX, Zhang L, He J, Gu JM, Chen HX, Mai HQ, Chen QY, Zhang XS, Gao S, Li J. Hypoxia Induces Mitochondrial Defect That Promotes T Cell Exhaustion in Tumor Microenvironment Through MYC-Regulated Pathways. Front Immunol 2020; 11:1906. [PMID: 32973789 PMCID: PMC7472844 DOI: 10.3389/fimmu.2020.01906] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/15/2020] [Indexed: 12/21/2022] Open
Abstract
T cell exhaustion is an obstacle to immunotherapy for solid tumors. An understanding of the mechanism by which T cells develop this phenotype in solid tumors is needed. Here, hypoxia, a feature of the tumor microenvironment, causes T cell exhaustion (TExh) by inducing a mitochondrial defect. Upon exposure to hypoxia, activated T cells with a TExh phenotype are characterized by mitochondrial fragmentation, decreased ATP production, and decreased mitochondrial oxidative phosphorylation activity. The TExh phenotype is correlated with the downregulation of the mitochondrial fusion protein mitofusin 1 (MFN1) and upregulation of miR-24. Overexpression of miR-24 alters the transcription of many metabolism-related genes including its target genes MYC and fibroblast growth factor 11 (FGF11). Downregulation of MYC and FGF11 induces TExh differentiation, reduced ATP production and a loss of the mitochondrial mass in T cell receptor (TCR)-stimulated T cells. In addition, we determined that MYC regulates the transcription of FGF11 and MFN1. In nasopharyngeal carcinoma (NPC) tissues, the T cells exhibit an increased frequency of exhaustion and loss of mitochondrial mass. In addition, inhibition of miR-24 signaling decreases NPC xenograft growth in nude mice. Our findings reveal a mechanism for T cell exhaustion in the tumor environment and provide potential strategies that target mitochondrial metabolism for cancer immunotherapy.
Collapse
Affiliation(s)
- Yi-Na Liu
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Jie-Feng Yang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Dai-Jia Huang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Huan-He Ni
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Chuan-Xia Zhang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Lin Zhang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Jia He
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jia-Mei Gu
- Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hong-Xia Chen
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Hai-Qiang Mai
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qiu-Yan Chen
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiao-Shi Zhang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Song Gao
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Jiang Li
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangzhou, China.,Department of Biotherapy, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Shenzhen, China
| |
Collapse
|
156
|
Hu Q, Yuan Y, Wu Y, Huang Y, Zhao Z, Xiao C. MicroRNA‑137 exerts protective effects on hypoxia‑induced cell injury by inhibiting autophagy/mitophagy and maintaining mitochondrial function in breast cancer stem‑like cells. Oncol Rep 2020; 44:1627-1637. [PMID: 32945512 PMCID: PMC7448477 DOI: 10.3892/or.2020.7714] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/10/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer stem‑like cells (BCSCs) have been identified and proven to play critical roles in tumorigenesis and progression. Hypoxia is a common pathologic feature of breast cancer and potentially, at least in part, regulates the initiation, progression, and recurrence of breast cancer. However, less is known about how hypoxia regulates BCSCs. As several well‑known microRNAs respond to hypoxia, we aimed to determine how hypoxia regulates the physiological processes of BCSCs by regulating the corresponding microRNAs. As expected, microRNA‑137 (miRNA‑137 or miR‑137) was downregulated upon hypoxic exposure, indicating that it may play critical roles in BCSCs. Introduction of miR‑137 mimics promoted cell cycle entry and inhibited hypoxia‑induced cell apoptosis as determined by cell cycle assay and apoptosis assay. By detecting mitochondrial reactive oxygen species (ROS), it was found that miR‑137 inhibited ROS accumulation induced by hypoxic exposure and thus suppressed cell apoptosis. Introduction of miR‑137 mimics under hypoxia inhibited mitophagy/autophagy by targeting FUN14 domain containing 1 (Fundc1) and thus promoted mitochondrial functions, including mitochondrial mass, ATP synthesis and mitochondrial transcriptional activity, which was similar to the effects of Fundc1 knockdown by specific siRNA. Based on these observations, we hypothesized that the survival of BCSCs under hypoxia was mediated by miR‑137 by regulating mitochondrial dysfunction. We demonstrated here that the introduction of exogenous miR‑137 promoted mitochondrial function, indicating that it may be a potential therapeutic target in BCSCs.
Collapse
Affiliation(s)
- Qiongying Hu
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Yun Yuan
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Yeke Wu
- Department of Stomatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Yongliang Huang
- Department of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Ziyi Zhao
- Department of Central Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Chong Xiao
- Teaching and Research Office of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
157
|
Reynolds BA, Oli MW, Oli MK. Eco-oncology: Applying ecological principles to understand and manage cancer. Ecol Evol 2020; 10:8538-8553. [PMID: 32884638 PMCID: PMC7452771 DOI: 10.1002/ece3.6590] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 12/25/2022] Open
Abstract
Cancer is a disease of single cells that expresses itself at the population level. The striking similarities between initiation and growth of tumors and dynamics of biological populations, and between metastasis and ecological invasion and community dynamics suggest that oncology can benefit from an ecological perspective to improve our understanding of cancer biology. Tumors can be viewed as complex, adaptive, and evolving systems as they are spatially and temporally heterogeneous, continually interacting with each other and with the microenvironment and evolving to increase the fitness of the cancer cells. We argue that an eco-evolutionary perspective is essential to understand cancer biology better. Furthermore, we suggest that ecologically informed therapeutic approaches that combine standard of care treatments with strategies aimed at decreasing the evolutionary potential and fitness of neoplastic cells, such as disrupting cell-to-cell communication and cooperation, and preventing successful colonization of distant organs by migrating cancer cells, may be effective in managing cancer as a chronic condition.
Collapse
Affiliation(s)
- Brent A. Reynolds
- Department of NeurosurgeryCollege of MedicineUniversity of FloridaGainesvilleFLUSA
| | - Monika W. Oli
- Department of Microbiology and Cell ScienceInstitute of Food and Agricultural SciencesUniversity of FloridaGainesvilleFLUSA
| | - Madan K. Oli
- Department of Wildlife Ecology and ConservationInstitute of Food and Agricultural SciencesUniversity of FloridaGainesvilleFLUSA
| |
Collapse
|
158
|
Joshi S. Targeting the Tumor Microenvironment in Neuroblastoma: Recent Advances and Future Directions. Cancers (Basel) 2020; 12:E2057. [PMID: 32722460 PMCID: PMC7465822 DOI: 10.3390/cancers12082057] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/30/2022] Open
Abstract
Neuroblastoma (NB) is the most common pediatric tumor malignancy that originates from the neural crest and accounts for more than 15% of all the childhood deaths from cancer. The neuroblastoma cancer research has long been focused on the role of MYCN oncogene amplification and the contribution of other genetic alterations in the progression of this malignancy. However, it is now widely accepted that, not only tumor cells, but the components of tumor microenvironment (TME), including extracellular matrix, stromal cells and immune cells, also contribute to tumor progression in neuroblastoma. The complexity of different components of tumor stroma and their resemblance with surrounding normal tissues pose huge challenges for therapies targeting tumor microenvironment in NB. Hence, the detailed understanding of the composition of the TME of NB is crucial to improve existing and future potential immunotherapeutic approaches against this childhood cancer. In this review article, I will discuss different components of the TME of NB and the recent advances in the strategies, which are used to target the tumor microenvironment in neuroblastoma.
Collapse
Affiliation(s)
- Shweta Joshi
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093-0815, USA
| |
Collapse
|
159
|
Li S, Zhang W, Xue H, Xing R, Yan X. Tumor microenvironment-oriented adaptive nanodrugs based on peptide self-assembly. Chem Sci 2020; 11:8644-8656. [PMID: 34123123 PMCID: PMC8163399 DOI: 10.1039/d0sc02937h] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The aberrant metabolism of tumor cells creates an inimitable microenvironment featuring acidic pH, high glutathione (GSH) levels, and overexpression of certain enzymes, which benefits the overwhelming progress of a tumor. Peptide self-assembly, emerging as a biofriendly and versatile fabrication strategy, harnesses multiple noncovalent interactions to obtain a variety of nanostructures tailored on demand. Orchestrating the reversible nature of noncovalent interactions and abnormal physiological parameters in the tumor microenvironment enables peptide-based nanodrugs to be targetable or switchable, thereby improving the drugs' bioavailability and optimizing the treatment outcome. This review will focus on peptide-modulated self-assembly of photosensitizers, chemotherapeutic drugs, immunoactive agents for tumor microenvironment-oriented adaptive phototherapy, chemotherapy, immunotherapy and combinatorial therapy. We will emphasize the building block design, the intermolecular interaction principle, adaptive structural transformation in the tumor microenvironment and corresponding therapeutic efficacy, and aim to elucidate the critical role of peptide-modulated, tumor microenvironment-oriented adaptive assemblies in improving the therapeutic index. Challenges and opportunities will be covered as well to advance the development and clinical application of tumor therapies based on peptide self-assembly materials and techniques.
Collapse
Affiliation(s)
- Shukun Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Beijing 100190 China http://www.yan-assembly.org/.,School of Chemical Engineering, University of Chinese Academy of Sciences Beijing 100049 China
| | - Wenjia Zhang
- Department of Radiology, Peking Union Medical College Hospital Beijing 100730 China
| | - Huadan Xue
- Department of Radiology, Peking Union Medical College Hospital Beijing 100730 China
| | - Ruirui Xing
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Beijing 100190 China http://www.yan-assembly.org/
| | - Xuehai Yan
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences Beijing 100190 China http://www.yan-assembly.org/.,School of Chemical Engineering, University of Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
160
|
Cheng YQ, Wang SB, Liu JH, Jin L, Liu Y, Li CY, Su YR, Liu YR, Sang X, Wan Q, Liu C, Yang L, Wang ZC. Modifying the tumour microenvironment and reverting tumour cells: New strategies for treating malignant tumours. Cell Prolif 2020; 53:e12865. [PMID: 32588948 PMCID: PMC7445401 DOI: 10.1111/cpr.12865] [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: 04/17/2020] [Revised: 06/02/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
Abstract
The tumour microenvironment (TME) plays a pivotal role in tumour fate determination. The TME acts together with the genetic material of tumour cells to determine their initiation, metastasis and drug resistance. Stromal cells in the TME promote the growth and metastasis of tumour cells by secreting soluble molecules or exosomes. The abnormal microenvironment reduces immune surveillance and tumour killing. The TME causes low anti‐tumour drug penetration and reactivity and high drug resistance. Tumour angiogenesis and microenvironmental hypoxia limit the drug concentration within the TME and enhance the stemness of tumour cells. Therefore, modifying the TME to effectively attack tumour cells could represent a comprehensive and effective anti‐tumour strategy. Normal cells, such as stem cells and immune cells, can penetrate and disrupt the abnormal TME. Reconstruction of the TME with healthy cells is an exciting new direction for tumour treatment. We will elaborate on the mechanism of the TME to support tumours and the current cell therapies for targeting tumours and the TME—such as immune cell therapies, haematopoietic stem cell (HSC) transplantation therapies, mesenchymal stem cell (MSC) transfer and embryonic stem cell‐based microenvironment therapies—to provide novel ideas for producing breakthroughs in tumour therapy strategies.
Collapse
Affiliation(s)
- Ya Qi Cheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shou Bi Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jia Hui Liu
- Affiliated Dongguan People's Hospital, Southern Medical University, Dongguan, China
| | - Lin Jin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ying Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Chao Yang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ya Ru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yu Run Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xuan Sang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Qi Wan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Chang Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Liu Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zhi Chong Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
161
|
Devarasetty M, Dominijanni A, Herberg S, Shelkey E, Skardal A, Soker S. Simulating the human colorectal cancer microenvironment in 3D tumor-stroma co-cultures in vitro and in vivo. Sci Rep 2020; 10:9832. [PMID: 32555362 PMCID: PMC7300090 DOI: 10.1038/s41598-020-66785-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022] Open
Abstract
The tumor microenvironment (TME) plays a significant role in cancer progression and thus modeling it will advance our understanding of cancer growth dynamics and response to therapies. Most in vitro models are not exposed to intact body physiology, and at the same time, fail to recapitulate the extensive features of the tumor stroma. Conversely, animal models do not accurately capture the human tumor architecture. We address these deficiencies with biofabricated colorectal cancer (CRC) tissue equivalents, which are built to replicate architectural features of biopsied CRC tissue. Our data shows that tumor-stroma co-cultures consisting of aligned extracellular matrix (ECM) fibers and ordered micro-architecture induced an epithelial phenotype in CRC cells while disordered ECM drove a mesenchymal phenotype, similar to well and poorly differentiated tumors, respectively. Importantly, co-cultures studied in vitro, and upon implantation in mice, revealed similar tumor growth dynamics and retention of architectural features for 28 days. Altogether, these results are the first demonstration of replicating human tumor ECM architecture in ex vivo and in vivo cultures.
Collapse
Affiliation(s)
| | | | - Samuel Herberg
- SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Ethan Shelkey
- Wake Forest Baptist Medical Center, Winston-Salem, NC, 27101, USA
| | | | - Shay Soker
- Wake Forest Baptist Medical Center, Winston-Salem, NC, 27101, USA.
| |
Collapse
|
162
|
Xuefeng X, Hou MX, Yang ZW, Agudamu A, Wang F, Su XL, Li X, Shi L, Terigele T, Bao LL, Wu XL. Epithelial-mesenchymal transition and metastasis of colon cancer cells induced by the FAK pathway in cancer-associated fibroblasts. J Int Med Res 2020; 48:300060520931242. [PMID: 32588696 PMCID: PMC7323289 DOI: 10.1177/0300060520931242] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 05/12/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE The role and mechanism of tetrathiomolybdate (TM) in cancer-associated fibroblasts (CAFs) in colon cancer using three-dimensional (3D) culture were investigated, and the associations between the focal adhesion kinase (FAK) pathway and epithelial-mesenchymal transition (EMT) in CAFs were explored. METHODS A 3D co-culture model of colon cancer LOVO cells with CAFs and normal fibroblasts (NFs) was established using Matrigel as a scaffold material. The differential expression of LOXL2 (lysyl oxidase-like 2) in the supernatant of CAFs and NFs was determined using ELISA, and expression levels of EMT-related proteins and FAK signaling pathway-related proteins were determined using western blot. RESULTS LOXL2 levels secreted by CAFs were higher compared with that secreted by NFs. In the CAF + LOVO group, compared with the LOVO group, E-cadherin expression decreased significantly, while N-cadherin and F-PAK expression increased significantly. TM results were opposite compared with the above results. CONCLUSIONS CAFs stimulate EMT in human colon cancer LOVO cells by secreting LOXL2 to activate the FAK signaling pathway, thereby promoting tumor metastasis. TM inhibited the occurrence of EMT in the CAF-induced colon cancer LOVO cell line, thereby reducing the invasion and metastasis of colon cancer cells.
Collapse
Affiliation(s)
- Xuefeng Xuefeng
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolian Autonomous Region, China
| | - Ming-Xing Hou
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolian Autonomous Region, China
| | - Zhi-Wen Yang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolian Autonomous Region, China
| | - Agudamu Agudamu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolian Autonomous Region, China
| | - Feng Wang
- Department of Gastrointestinal Surgery, Beijing Tsinghua Changgung Hospital, Beijing, China
| | - Xiu-Lan Su
- Clinical Medicine Research Center, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolian Autonomous Region, China
| | - Xian Li
- Clinical Medicine Research Center, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolian Autonomous Region, China
| | - Lin Shi
- Department of Pathology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolian Autonomous Region, China
| | - Terigele Terigele
- Department of Pathology, Inner Mongolia Autonomous Region Maternal and Child Health Hospital, Hohhot, Inner Mongolian Autonomous Region, China
| | - Li-Li Bao
- Center of Geriatrics, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolian Autonomous Region, China
| | - Xin-Lin Wu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolian Autonomous Region, China
| |
Collapse
|
163
|
Lian Z, Hu Z, Xian H, Jiang R, Huang H, Jiang Y, Zheng Z, Lloyd RS, Yuan J, Sha Y, Wang S, Hu D. Exosomes derived from normal human bronchial epithelial cells down-regulate proliferation and migration of hydroquinone-transformed malignant recipient cells via up-regulating PTEN expression. CHEMOSPHERE 2020; 244:125496. [PMID: 31812062 DOI: 10.1016/j.chemosphere.2019.125496] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/22/2019] [Accepted: 11/26/2019] [Indexed: 06/10/2023]
Abstract
The gene encoding the tumor suppressor, phosphatase and tensin homolog (PTEN), located on chromosome 10, is frequently expressed at low levels in various tumors, resulting in the stimulation of cell proliferation and migration. However, the role of exosomal PTEN in cell-cell communication during the progress of benzene-induced carcinogenesis remains unclear. The goal of this study was to explore whether exosomes derived from normal human bronchial epithelial cells (16HBE) could transmit PTEN to hydroquinone-transformed malignant recipient cells (16HBE-t) and its possible effects on cell proliferation and migration. Consistent with PTEN expression being down-regulated in transformed cells, we found that its expression was significantly decreased in 16HBE-t relative to 16HBE cells and that purified exosomes secreted by 16HBE, up-regulated PTEN levels in recipient 16HBE-t cells. Thus, down-regulating their proliferation and migration. Further, when exosomes derived from 16HBE cells that had been treated with the PTEN inhibitor SF1670, were incubated with recipient 16HBE-t cells, they exhibited decreased PTEN levels, with a corresponding increase in their proliferation and migration. In conclusion, our study demonstrates that exosomes derived from 16HBE cells can down-regulate proliferation and migration of recipient 16HBE-t cells via transferring PTEN.
Collapse
Affiliation(s)
- Zhenwei Lian
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Zuqing Hu
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China; Department of Medicine, Jiamusi University, Jiamusi, 154007, China
| | - Hongyi Xian
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Ran Jiang
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Haoyu Huang
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Yunxia Jiang
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Zhongdaixi Zheng
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - R Stephen Lloyd
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, 3181 S. W. Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - Jianhui Yuan
- Nanshan District Center for Disease Control and Prevention, Shenzhen, 518054, China
| | - Yan Sha
- Institute of Occupational Disease, Shenzhen Prevention and Treatment Center for Occupational Disease, Shenzhen, 518020, China
| | - Sanming Wang
- Faculty of Health Sciences, University of Macau, Taipa, SAR, Macau, China
| | - Dalin Hu
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
164
|
Li ZL, Wang ZJ, Wei GH, Yang Y, Wang XW. Changes in extracellular matrix in different stages of colorectal cancer and their effects on proliferation of cancer cells. World J Gastrointest Oncol 2020; 12:267-275. [PMID: 32206177 PMCID: PMC7081112 DOI: 10.4251/wjgo.v12.i3.267] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 01/12/2020] [Accepted: 02/07/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The extracellular matrix is the main component of the tumor microenvironment. Extracellular matrix remodels with the oncogenesis and development of tumors. Previous studies usually focused on the changes of proteins in normal colorectal tissues and colorectal cancers. Little is known about the changes in the extracellular matrix in different stages of colorectal cancer and the effects of these changes on the development of this cancer. AIM To test the changes of type I collagen, type IV collagen, matrix metalloproteinase-2 (MMP-2), matrix metalloproteinase-9 (MMP-9), and tissue inhibitor of metalloproteinase-3 (TIMP-3) in different stages of colorectal cancer and the effects of these changes on the proliferation of cancer cells. METHODS The extracellular matrix from various stages of colorectal cancer and normal colon tissue was obtained by using acellular technology. We used proteomics to detect the differential expression of proteins between normal colon tissues and colorectal cancer tissues, and then we used Western blot to observe their expression in each stage of colorectal cancer and in normal colon tissue. By co-culturing the extracellular matrix and HT29 colon cancer cells in vivo and in vitro, we tested the cancer cell proliferation rate in vitro by methyl thiazolyl tetrazolium (MTT) assay and in vivo by measuring the tumor volume. RESULTS The expression of type I collagen and MMP-2 increased with increased tumor stage. The expression of MMP-9 was higher in colorectal cancer tissues and was highest in stage III cancer. The expression of type IV collagen and TIMP-3 decreased with increased tumor stage. The proliferation rate of cancer cells in the extracellular matrix of colorectal cancer was higher than that in the extracellular matrix of the normal colon. CONCLUSION These data suggest that the extracellular matrix structure and composition become disorganized during the development of tumors, which is more conducive for the growth of cancer cells.
Collapse
Affiliation(s)
- Zhu-Lin Li
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Zhen-Jun Wang
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Guang-Hui Wei
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yong Yang
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiao-Wan Wang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, China
| |
Collapse
|
165
|
Fu R, Li Y, Jiang N, Ren BX, Zang CZ, Liu LJ, Lv WC, Li HM, Weiss S, Li ZY, Lu T, Wu ZQ. Inactivation of endothelial ZEB1 impedes tumor progression and sensitizes tumors to conventional therapies. J Clin Invest 2020; 130:1252-1270. [PMID: 32039918 PMCID: PMC7269596 DOI: 10.1172/jci131507] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 12/03/2019] [Indexed: 12/29/2022] Open
Abstract
Current antiangiogenic therapy is limited by its cytostatic property, scarce drug delivery to the tumor, and side toxicity. To address these limitations, we unveiled the role of ZEB1, a tumor endothelium-enriched zinc-finger transcription factor, during tumor progression. We discovered that the patients who had lung adenocarcinomas with high ZEB1 expression in tumor endothelium had increased prevalence of metastases and markedly reduced overall survival after the diagnosis of lung cancer. Endothelial ZEB1 deletion in tumor-bearing mice diminished tumor angiogenesis while eliciting persistent tumor vascular normalization by epigenetically repressing TGF-β signaling. This consequently led to improved blood and oxygen perfusion, enhanced chemotherapy delivery and immune effector cell infiltration, and reduced tumor growth and metastasis. Moreover, targeting vascular ZEB1 remarkably potentiated the anticancer activity of nontoxic low-dose cisplatin. Treatment with low-dose anti-programmed cell death protein 1 (anti-PD-1) antibody elicited tumor regression and markedly extended survival in ZEB1-deleted mice, conferring long-term protective anticancer immunity. Collectively, we demonstrated that inactivation of endothelial ZEB1 may offer alternative opportunities for cancer therapy with minimal side effects. Targeting endothelium-derived ZEB1 in combination with conventional chemotherapy or immune checkpoint blockade therapy may yield a potent and superior anticancer effect.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Cisplatin/pharmacology
- Endothelium/immunology
- Epigenesis, Genetic/drug effects
- Epigenesis, Genetic/immunology
- Gene Deletion
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/immunology
- Mice
- Mice, Transgenic
- Neoplasms, Experimental/blood supply
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/immunology
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/immunology
- Zinc Finger E-box-Binding Homeobox 1/deficiency
- Zinc Finger E-box-Binding Homeobox 1/immunology
Collapse
Affiliation(s)
- Rong Fu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yi Li
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Nan Jiang
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Bo-Xue Ren
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chen-Zi Zang
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Li-Juan Liu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wen-Cong Lv
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hong-Mei Li
- State Key Laboratory of Natural Medicines, Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, Nanjing, China
| | - Stephen Weiss
- Life Sciences Institute, Rogel Cancer Center, Department of Internal Medicine, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Zheng-Yu Li
- Department of Medicinal Chemistry, College of Chemistry, University of Glasgow, Glasgow, United Kingdom
| | - Tao Lu
- State Key Laboratory of Natural Medicines, Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, Nanjing, China
| | - Zhao-Qiu Wu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
166
|
Lin Z, Luo G, Du W, Kong T, Liu C, Liu Z. Recent Advances in Microfluidic Platforms Applied in Cancer Metastasis: Circulating Tumor Cells' (CTCs) Isolation and Tumor-On-A-Chip. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1903899. [PMID: 31747120 DOI: 10.1002/smll.201903899] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/13/2019] [Indexed: 05/03/2023]
Abstract
Cancer remains the leading cause of death worldwide despite the enormous efforts that are made in the development of cancer biology and anticancer therapeutic treatment. Furthermore, recent studies in oncology have focused on the complex cancer metastatic process as metastatic disease contributes to more than 90% of tumor-related death. In the metastatic process, isolation and analysis of circulating tumor cells (CTCs) play a vital role in diagnosis and prognosis of cancer patients at an early stage. To obtain relevant information on cancer metastasis and progression from CTCs, reliable approaches are required for CTC detection and isolation. Additionally, experimental platforms mimicking the tumor microenvironment in vitro give a better understanding of the metastatic microenvironment and antimetastatic drugs' screening. With the advancement of microfabrication and rapid prototyping, microfluidic techniques are now increasingly being exploited to study cancer metastasis as they allow precise control of fluids in small volume and rapid sample processing at relatively low cost and with high sensitivity. Recent advancements in microfluidic platforms utilized in various methods for CTCs' isolation and tumor models recapitulating the metastatic microenvironment (tumor-on-a-chip) are comprehensively reviewed. Future perspectives on microfluidics for cancer metastasis are proposed.
Collapse
Affiliation(s)
- Zhengjie Lin
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Guanyi Luo
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Weixiang Du
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Tiantian Kong
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Changkun Liu
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Zhou Liu
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| |
Collapse
|
167
|
Saber SH, Ali HEA, Gaballa R, Gaballah M, Ali HI, Zerfaoui M, Abd Elmageed ZY. Exosomes are the Driving Force in Preparing the Soil for the Metastatic Seeds: Lessons from the Prostate Cancer. Cells 2020; 9:E564. [PMID: 32121073 PMCID: PMC7140426 DOI: 10.3390/cells9030564] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/17/2020] [Accepted: 02/20/2020] [Indexed: 12/13/2022] Open
Abstract
Exosomes are nano-membrane vesicles that various cell types secrete during physiological and pathophysiological conditions. By shuttling bioactive molecules such as nucleic acids, proteins, and lipids to target cells, exosomes serve as key regulators for multiple cellular processes, including cancer metastasis. Recently, microvesicles have emerged as a challenge in the treatment of prostate cancer (PCa), encountered either when the number of vesicles increases or when the vesicles move into circulation, potentially with an ability to induce drug resistance, angiogenesis, and metastasis. Notably, the exosomal cargo can induce the desmoplastic response of PCa-associated cells in a tumor microenvironment (TME) to promote PCa metastasis. However, the crosstalk between PCa-derived exosomes and the TME remains only partially understood. In this review, we provide new insights into the metabolic and molecular signatures of PCa-associated exosomes in reprogramming the TME, and the subsequent promotion of aggressive phenotypes of PCa cells. Elucidating the molecular mechanisms of TME reprogramming by exosomes draws more practical and universal conclusions for the development of new therapeutic interventions when considering TME in the treatment of PCa patients.
Collapse
Affiliation(s)
- Saber H. Saber
- Laboratory of Molecular Cell Biology, Department of Zoology, Faculty of Science, Assiut University, Assiut 71515, Egypt;
| | - Hamdy E. A. Ali
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M Health Science Center, College Station, TX 77843, USA; (H.E.A.A.); (R.G.); (M.G.); (H.I.A.)
| | - Rofaida Gaballa
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M Health Science Center, College Station, TX 77843, USA; (H.E.A.A.); (R.G.); (M.G.); (H.I.A.)
| | - Mohamed Gaballah
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M Health Science Center, College Station, TX 77843, USA; (H.E.A.A.); (R.G.); (M.G.); (H.I.A.)
| | - Hamed I. Ali
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M Health Science Center, College Station, TX 77843, USA; (H.E.A.A.); (R.G.); (M.G.); (H.I.A.)
| | - Mourad Zerfaoui
- Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA;
| | - Zakaria Y. Abd Elmageed
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M Health Science Center, College Station, TX 77843, USA; (H.E.A.A.); (R.G.); (M.G.); (H.I.A.)
| |
Collapse
|
168
|
Zhang Z, Zhang J, Jiang M, Zhao L, Li S, Sun H, Yang F, Liang H. Human Serum Albumin-Based Dual-Agent Delivery Systems for Combination Therapy: Acting against Cancer Cells and Inhibiting Neovascularization in the Tumor Microenvironment. Mol Pharm 2020; 17:1405-1414. [DOI: 10.1021/acs.molpharmaceut.0c00133] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Zhenlei Zhang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin, Guangxi 541004 China
| | - Juzheng Zhang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin, Guangxi 541004 China
| | - Ming Jiang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin, Guangxi 541004 China
| | - Lei Zhao
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin, Guangxi 541004 China
| | - Shanhe Li
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin, Guangxi 541004 China
| | - Hongbin Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, Jiangsu 210009 China
| | - Feng Yang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin, Guangxi 541004 China
| | - Hong Liang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin, Guangxi 541004 China
| |
Collapse
|
169
|
Ge Y, Chen W, Zhang X, Wang H, Cui J, Liu Y, Ju S, Tian X, Ju S. Nuclear-localized costimulatory molecule 4-1BBL promotes colon cancer cell proliferation and migration by regulating nuclear Gsk3β, and is linked to the poor outcomes associated with colon cancer. Cell Cycle 2020; 19:577-591. [PMID: 31992123 DOI: 10.1080/15384101.2020.1719308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Anti-tumor immune response and the prognosis of tumor are the results of competition between stimulatory and inhibitory checkpoints. Except for upregulating inhibitory checkpoints, lowering some immune accelerating molecules to convert an immunostimulatory microenvironment into an immunodormant one through "decelerating the accelerator" might be another effective immune escape pattern. 4-1BBL is a classical transmembrane costimulatory molecule involving in antitumor immune responses. In contrast, we demonstrated that 4-1BBL is predominantly localized in the nuclei of cancer cells in colon cancer specimens and is positively correlated with tumor size, lymph node metastasis, and a lower survival ratio. Furthermore, the nuclear localization of 4-1BBL was also ascertained in vitro. 4-1BBL knockout (KO) arrests the proliferation and impaired the migration and invasion ability of colon cancer cells in vitro and retarded tumor growth in vivo. 4-1BBL KO increased the accumulation of Gsk3β in the nuclei of colon cancer cells and consequently decreased the expression of Wnt pathway target genes and thus alter tumor biological behavior. We hypothesized that unlike membrane-expressed 4-1BBL, which stimulates the 4-1BB signaling of antitumor cytotoxic T cells, the nuclear-localized 4-1BBL could facilitate the malignant behavior of colon cancer cells by circumventing antitumor signaling and driving some key oncotropic signal pathway in the nucleus. Nuclear-localized 4-1BBL might be an indicator of colon cancer malignancy and serve as a promising target of immunotherapy.
Collapse
Affiliation(s)
- Yan Ge
- Department of Immunology, School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Wei Chen
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xueguang Zhang
- Department of Immunology, School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Haiyan Wang
- Department of Immunology, School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Juanjuan Cui
- Department of Immunology, School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Yue Liu
- Department of Immunology, School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Songwen Ju
- Central Laboratory, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Xinxin Tian
- Department of Immunology, School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu Province, China.,Departmemt of Medical Care for Cadres, Nanjing Municipal Government Hospital, Nanjing, Jiangsu Province, China
| | - Songguang Ju
- Department of Immunology, School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
170
|
Zhang FP, Huang YP, Luo WX, Deng WY, Liu CQ, Xu LB, Liu C. Construction of a risk score prognosis model based on hepatocellular carcinoma microenvironment. World J Gastroenterol 2020; 26:134-153. [PMID: 31969776 PMCID: PMC6962430 DOI: 10.3748/wjg.v26.i2.134] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/23/2019] [Accepted: 12/06/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a common cancer with a poor prognosis. Previous studies revealed that the tumor microenvironment (TME) plays an important role in HCC progression, recurrence, and metastasis, leading to poor prognosis. However, the effects of genes involved in TME on the prognosis of HCC patients remain unclear. Here, we investigated the HCC microenvironment to identify prognostic genes for HCC.
AIM To identify a robust gene signature associated with the HCC microenvironment to improve prognosis prediction of HCC.
METHODS We computed the immune/stromal scores of HCC patients obtained from The Cancer Genome Atlas based on the ESTIMATE algorithm. Additionally, a risk score model was established based on Differentially Expressed Genes (DEGs) between high‐ and low‐immune/stromal score patients.
RESULTS The risk score model consisting of eight genes was constructed and validated in the HCC patients. The patients were divided into high- or low-risk groups. The genes (Disabled homolog 2, Musculin, C-X-C motif chemokine ligand 8, Galectin 3, B-cell-activating transcription factor, Killer cell lectin like receptor B1, Endoglin and adenomatosis polyposis coli tumor suppressor) involved in our risk score model were considered to be potential immunotherapy targets, and they may provide better performance in combination. Functional enrichment analysis showed that the immune response and T cell receptor signaling pathway represented the major function and pathway, respectively, related to the immune-related genes in the DEGs between high- and low-risk groups. The receiver operating characteristic (ROC) curve analysis confirmed the good potency of the risk score prognostic model. Moreover, we validated the risk score model using the International Cancer Genome Consortium and the Gene Expression Omnibus database. A nomogram was established to predict the overall survival of HCC patients.
CONCLUSION The risk score model and the nomogram will benefit HCC patients through personalized immunotherapy.
Collapse
MESH Headings
- Aged
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Biomarkers, Tumor/antagonists & inhibitors
- Biomarkers, Tumor/genetics
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/mortality
- Databases, Genetic/statistics & numerical data
- Datasets as Topic
- Female
- Gene Expression Profiling/methods
- Gene Expression Regulation, Neoplastic/immunology
- Humans
- Kaplan-Meier Estimate
- Liver/immunology
- Liver/pathology
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/immunology
- Liver Neoplasms/mortality
- Male
- Middle Aged
- Models, Genetic
- Neoplasm Staging
- Nomograms
- Precision Medicine/methods
- ROC Curve
- Risk Assessment/methods
- Treatment Outcome
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Fa-Peng Zhang
- Department of Biliary Pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
| | - Yi-Pei Huang
- Department of Biliary Pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
| | - Wei-Xin Luo
- Department of Biliary Pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
| | - Wan-Yu Deng
- Department of Biliary Pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
- College of Life Science, Shangrao Normal University, Shangrao 334001, Jiangxi Province, China
| | - Chao-Qun Liu
- Department of Biliary Pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
| | - Lei-Bo Xu
- Department of Biliary Pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
| | - Chao Liu
- Department of Biliary Pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
| |
Collapse
|
171
|
Microenvironment in Cardiac Tumor Development: What Lies Beyond the Event Horizon? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1226:51-56. [PMID: 32030675 DOI: 10.1007/978-3-030-36214-0_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cardiac tumors are found in less than 1% of adult and pediatric autopsies. More than three-fourths of primary cardiac neoplasms are benign, with myxomas and rhabdomyomas being the most common cardiac tumors seen in adults and children, respectively. Primary malignant cardiac tumors are extremely rare, whereas metastatic lesions can be seen in approximately 8% of patients dying from cancer. Attempting to understand why the heart is so resistant to carcinogenesis and which fail-safe mechanisms malfunction when cardiac tumors do develop is particularly challenging considering the rarity of these tumors and the fact that when relevant clinical studies are published, they rarely focus on molecular pathogenesis. Apart from cancer cells, solid tumors are comprised of a concoction of noncancerous cells, and extracellular matrix constituents, which along with pH and oxygen levels jointly constitute the so-called tumor microenvironment (TME). In the present chapter, we explore mechanisms through which TME may influence cardiac carcinogenesis.
Collapse
|
172
|
Xiao W, Sun G, Fan T, Liu J, Zhang N, Zhao L, Zhong R. Reductive Activity and Mechanism of Hypoxia- Targeted AGT Inhibitors: An Experimental and Theoretical Investigation. Int J Mol Sci 2019; 20:6308. [PMID: 31847200 PMCID: PMC6941096 DOI: 10.3390/ijms20246308] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/05/2019] [Accepted: 12/10/2019] [Indexed: 11/16/2022] Open
Abstract
O6-alkylguanine-DNA alkyltransferase (AGT) is the main cause of tumor cell resistance to DNA-alkylating agents, so it is valuable to design tumor-targeted AGT inhibitors with hypoxia activation. Based on the existing benchmark inhibitor O6-benzylguanine (O6-BG), four derivatives with hypoxia-reduced potential and their corresponding reduction products were synthesized. A reductase system consisting of glucose/glucose oxidase, xanthine/xanthine oxidase, and catalase were constructed, and the reduction products of the hypoxia-activated prodrugs under normoxic and hypoxic conditions were determined by high-performance liquid chromatography electrospray ionization tandem mass spectrometry (HPLC-ESI-MS/MS). The results showed that the reduction products produced under hypoxic conditions were significantly higher than that under normoxic condition. The amount of the reduction product yielded from ANBP (2-nitro-6-(3-amino) benzyloxypurine) under hypoxic conditions was the highest, followed by AMNBP (2-nitro-6-(3-aminomethyl)benzyloxypurine), 2-NBP (2-nitro-6-benzyloxypurine), and 3-NBG (O6-(3-nitro)benzylguanine). It should be noted that although the levels of the reduction products of 2-NBP and 3-NBG were lower than those of ANBP and AMNBP, their maximal hypoxic/normoxic ratios were higher than those of the other two prodrugs. Meanwhile, we also investigated the single electron reduction mechanism of the hypoxia-activated prodrugs using density functional theory (DFT) calculations. As a result, the reduction of the nitro group to the nitroso was proven to be a rate-limiting step. Moreover, the 2-nitro group of purine ring was more ready to be reduced than the 3-nitro group of benzyl. The energy barriers of the rate-limiting steps were 34-37 kcal/mol. The interactions between these prodrugs and nitroreductase were explored via molecular docking study, and ANBP was observed to have the highest affinity to nitroreductase, followed by AMNBP, 2-NBP, and 3-NBG. Interestingly, the theoretical results were generally in a good agreement with the experimental results. Finally, molecular docking and molecular dynamics simulations were performed to predict the AGT-inhibitory activity of the four prodrugs and their reduction products. In summary, simultaneous consideration of reduction potential and hypoxic selectivity is necessary to ensure that such prodrugs have good hypoxic tumor targeting. This study provides insights into the hypoxia-activated mechanism of nitro-substituted prodrugs as AGT inhibitors, which may contribute to reasonable design and development of novel tumor-targeted AGT inhibitors.
Collapse
Affiliation(s)
- Weinan Xiao
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China; (W.X.); (G.S.); (T.F.); (J.L.); (N.Z.); (R.Z.)
| | - Guohui Sun
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China; (W.X.); (G.S.); (T.F.); (J.L.); (N.Z.); (R.Z.)
| | - Tengjiao Fan
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China; (W.X.); (G.S.); (T.F.); (J.L.); (N.Z.); (R.Z.)
- Department of Medical Technology, Beijing Pharmaceutical University of Staff and Workers, Beijing 100079, China
| | - Junjun Liu
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China; (W.X.); (G.S.); (T.F.); (J.L.); (N.Z.); (R.Z.)
| | - Na Zhang
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China; (W.X.); (G.S.); (T.F.); (J.L.); (N.Z.); (R.Z.)
| | - Lijiao Zhao
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China; (W.X.); (G.S.); (T.F.); (J.L.); (N.Z.); (R.Z.)
| | - Rugang Zhong
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China; (W.X.); (G.S.); (T.F.); (J.L.); (N.Z.); (R.Z.)
| |
Collapse
|
173
|
Francisco BJ, Palumbo JS. New insights into cancer's exploitation of platelets. J Thromb Haemost 2019; 17:2000-2003. [PMID: 31797541 DOI: 10.1111/jth.14624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/19/2019] [Indexed: 11/30/2022]
Affiliation(s)
- Brenton J Francisco
- Department of Pediatrics, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital, The University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joseph S Palumbo
- Department of Pediatrics, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital, The University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
174
|
|
175
|
Yan J, Yan S, Hou P, Lu W, Ma PX, He W, Lei B. A Hierarchical Peptide-Lanthanide Framework To Accurately Redress Intracellular Carcinogenic Protein-Protein Interaction. NANO LETTERS 2019; 19:7918-7926. [PMID: 31645103 DOI: 10.1021/acs.nanolett.9b03028] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Intracellular protein-protein interactions (PPIs) are a vital and yet underexploited class of therapeutic targets for their crucial roles in cellular processes and involvement in disease initiation and progression. Although some successful chemistry and nanotechnologies have been introduced into peptide PPI modulators to allow cell and tissue permeability, significant challenges remain with regard to the efficient and precise modulation of PPIs within specific cells of diseased tissues, such as solid tumors. Herein, an intratumoral transformable hierarchical framework, termed iPLF, was fabricated via a two-step self-assembly between peptides and lanthanide-doped nanocrystals. In this proof-of-concept study, using NanoEL effect, TME response, and tumor marker targeting, iPLF in vivo delivered the p53-MDM2 modulator DPMI into tumor cells and β-catenin-Bcl9 modulator Bcl9p into tumor stem cells. This crafted programmed nanomedicine with triple-stage delivery and responsiveness accurately modulated the specific intracellular protein-protein interactions, resulting in the suppression of tumor growth and metastasis in vivo, while maintaining a highly favorable safety profile. iPLF reached the goal of accurate, potent, and hazard-free intracellular PPI modulation, thereby providing a means to improve current knowledge of PPI networks and a novel therapeutic strategy for a great variety of diseases.
Collapse
Affiliation(s)
- Jin Yan
- Frontier Institute of Science and Technology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology , Xi'an Jiaotong University , Xi'an 710054 , China
| | - Siqi Yan
- The First Affiliated Hospital of Xi'an Jiaotong University , Xi'an 710061 , P.R. China
| | - Peng Hou
- The First Affiliated Hospital of Xi'an Jiaotong University , Xi'an 710061 , P.R. China
| | - Wuyuan Lu
- Institute of Human Virology and Department of Biochemistry and Molecular Biology , University of Maryland School of Medicine , Baltimore , Maryland 21201 , United States
| | - Peter X Ma
- Department of Biologic and Materials Sciences, Department of Biomedical Engineering, Macromolecular Science and Engineering Center, Department of Materials Science and Engineering , University of Michigan , Ann Arbor , Michigan 48109 , United States
| | - Wangxiao He
- Department of Talent Highland , The First Affiliated Hospital of Xi'an Jiao Tong University , Xi'an 710061 , China
| | - Bo Lei
- Frontier Institute of Science and Technology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology , Xi'an Jiaotong University , Xi'an 710054 , China
| |
Collapse
|
176
|
Relationships between hypoxia induced factor-1α and 18F-FDG PET/CT parameters in colorectal cancer. Rev Esp Med Nucl Imagen Mol 2019. [DOI: 10.1016/j.remnie.2019.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
177
|
Hasbek Z, Ozer H, Erturk SA, Erdiş E, Yucel B, Çiftçi E, Çakmakcilar A. Relationships between hypoxia induced factor-1α and 18F-FDG PET/CT parameters in colorectal cancer. Rev Esp Med Nucl Imagen Mol 2019; 38:355-361. [PMID: 31672495 DOI: 10.1016/j.remn.2019.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/28/2019] [Accepted: 05/02/2019] [Indexed: 10/25/2022]
Abstract
AIM The hypoxia-inducible factor 1 (HIF-1) has a critical role in oxygen homeostasis and it is a transcriptional activator of angiogenesis, erythropoiesis, iron and glucose metabolism. Glucose metabolism rate is increased in some tumours via HIF-1α. Our aim is to evaluate the relationship between hypoxia in colorectal cancer, PET parameters, necrotic tissue size and pathologic prognostic factors via using HIF-1α. MATERIALS/METHODS 70 patients (28 female/42 male; median age: 63 years) who were diagnosed with colorectal cancer via biopsy were staged with preoperative PET/CT and operated subsequently. Immunohistochemical evaluation scoring was done according to nuclear HIF-1α expression, staining density and intensity. Metabolic tumour volume (MTV), total lesion glycolysis (TLG) and tumour volume (TV) were calculated by using volume of an ellipsoid formula via CT images, and percentage of tumour necrosis (%TmNcr) that was calculated by the difference between TV and recorded MTV. RESULTS There was a moderately meaningful positive correlation between tumour SUVmax and TV and %TmNcr (r=0.403, p=0.001 and r=0.500, p=0.0001, respectively). There were no statistically significant relationships between HIF-1α expression levels and tumour SUVmax, TLG, MTV, TV, %TmNcr, tumour stage, lymphovascular invasion, perineural invasion and extracapsular/capsular lymph node involvement. On the other hand, strong nuclear immunohistochemical staining was seen in tumour cells adjacent to invasive border, inflammatory cells. Although not statistically significant, moderate or strong nuclear staining were seen in 64.9% of metastatic patients. CONCLUSION Although the presence of a positive correlation between tumour SUVmax and %TmNcr shows that there are hypoxic cells in cancer tissue with high FDG uptake, the relationship between the presence of HIF-1α and enhanced glucose metabolism and pathological prognostic factors of tumour was not shown. Strong nuclear immunohistochemical staining in tumour cells adjacent to invasive border and inflammatory cells leads us to believe that HIF-1α plays a role in the invasion area of tumour microenvironment.
Collapse
Affiliation(s)
- Z Hasbek
- Cumhuriyet University, School of Medicine, Department of Nuclear Medicine, Sivas, Turquía.
| | - H Ozer
- Cumhuriyet University, School of Medicine, Department of Pathology, Sivas, Turquía
| | - S A Erturk
- Cumhuriyet University, School of Medicine, Department of Nuclear Medicine, Sivas, Turquía
| | - E Erdiş
- Cumhuriyet University, School of Medicine, Department of Radiation Oncology, Sivas, Turquía
| | - B Yucel
- Cumhuriyet University, School of Medicine, Department of Radiation Oncology, Sivas, Turquía
| | - E Çiftçi
- Sakarya University, School of Medicine, Research and Training Hospital, Department of Nuclear Medicine, Sakarya, Turquía
| | - A Çakmakcilar
- Cumhuriyet University, School of Medicine, Department of Nuclear Medicine, Sivas, Turquía
| |
Collapse
|
178
|
Valès S, Bacola G, Biraud M, Touvron M, Bessard A, Geraldo F, Dougherty KA, Lashani S, Bossard C, Flamant M, Duchalais E, Marionneau-Lambot S, Oullier T, Oliver L, Neunlist M, Vallette FM, Van Landeghem L. Tumor cells hijack enteric glia to activate colon cancer stem cells and stimulate tumorigenesis. EBioMedicine 2019; 49:172-188. [PMID: 31662289 PMCID: PMC6945247 DOI: 10.1016/j.ebiom.2019.09.045] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/25/2019] [Accepted: 09/25/2019] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Colon cancer stem cells (CSCs), considered responsible for tumor initiation and cancer relapse, are constantly exposed to regulatory cues emanating from neighboring cells present in the tumor microenvironment. Among these cells are enteric glial cells (EGCs) that are potent regulators of the epithelium functions in a healthy intestine. However, whether EGCs impact CSC-driven tumorigenesis remains unknown. METHODS Impact of human EGC primary cultures or a non-transformed EGC line on CSCs isolated from human primary colon adenocarcinomas or colon cancer cell lines with different p53, MMR system and stemness status was determined using murine xenograft models and 3D co-culture systems. Supernatants of patient-matched human primary colon adenocarcinomas and non-adjacent healthy mucosa were used to mimic tumor versus healthy mucosa secretomes and compare their effects on EGCs. FINDINGS Our data show that EGCs stimulate CSC expansion and ability to give rise to tumors via paracrine signaling. Importantly, only EGCs that were pre-activated by tumor epithelial cell-derived soluble factors increased CSC tumorigenicity. Pharmacological inhibition of PGE2 biosynthesis in EGCs or IL-1 knockdown in tumor epithelial cells prevented EGC acquisition of a pro-tumorigenic phenotype. Inhibition of PGE2 receptor EP4 and EGFR in CSCs inhibited the effects of tumor-activated EGCs. INTERPRETATION Altogether, our results show that EGCs, once activated by the tumor, acquire a pro-tumorigenic phenotype and stimulate CSC-driven tumorigenesis via a PGE2/EP4/EGFR-dependent pathway. FUNDING This work was supported by grants from the French National Cancer Institute, La Ligue contre le Cancer, the 'Région des Pays de la Loire' and the UNC Lineberger Comprehensive Cancer Center.
Collapse
Affiliation(s)
- Simon Valès
- Bretagne Loire University, Nantes University, INSERM 1235, IMAD, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France
| | - Gregory Bacola
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Mandy Biraud
- Bretagne Loire University, Nantes University, INSERM 1235, IMAD, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France
| | - Mélissa Touvron
- Bretagne Loire University, Nantes University, INSERM 1235, IMAD, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France,Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Anne Bessard
- Bretagne Loire University, Nantes University, INSERM 1235, IMAD, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France
| | - Fanny Geraldo
- Nantes University, INSERM 1232, CRCINA, Nantes, France
| | - Kelsie A. Dougherty
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Shaian Lashani
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | | | - Mathurin Flamant
- Nantes University Hospital, Nantes, France,Jules Verne Clinic, Nantes, France
| | - Emilie Duchalais
- Bretagne Loire University, Nantes University, INSERM 1235, IMAD, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France,Nantes University Hospital, Nantes, France
| | | | | | - Lisa Oliver
- Nantes University, INSERM 1232, CRCINA, Nantes, France,Nantes University Hospital, Nantes, France
| | - Michel Neunlist
- Bretagne Loire University, Nantes University, INSERM 1235, IMAD, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France,Nantes University Hospital, Nantes, France
| | | | - Laurianne Van Landeghem
- Bretagne Loire University, Nantes University, INSERM 1235, IMAD, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France,Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA,Corresponding author at: North Carolina State University, College of Veterinary Medicine, 1060 William Moore Drive, CB# 8401, Raleigh, NC 27607, USA.
| |
Collapse
|
179
|
Chen R, Gong Y, Zou D, Wang L, Yuan L, Zhou Q. Correlation between subsets of tumor-infiltrating immune cells and risk stratification in patients with cervical cancer. PeerJ 2019; 7:e7804. [PMID: 31616592 PMCID: PMC6791348 DOI: 10.7717/peerj.7804] [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: 06/14/2019] [Accepted: 08/31/2019] [Indexed: 12/15/2022] Open
Abstract
Aim To investigate the correlation between clinicopathological features and risk stratification in cervical cancer patients, and evaluate the feasibility of tumor-infiltrating immune cells as prognostic biomarkers in clinical practice. Methods CD3+ tumor infiltrating T cells (TILs), CD45RO+ TILs, CD4+ TILs, CD8+ TILs, FOXP3+ TILs (regulatory T cells, Tregs), CD68+ tumor associated macrophages (TAMs), CD163+ TAMs, and PD-L1+ tumor cells were immunostained in formalin-fixed paraffin-embedded (PPFE) tissues from 96 cervical cancer patients. Immunostaining density and other clinicopathological features such as age, FIGO stage, histopathologic type, Ki67 index, HPV status, lymhovasular invasion status (LVI), lymph node metastasis, tumor size, stromal invasion status, surgical margin status, and parametrial invasion, were evaluated for their roles in risk stratification of cervical cancer patients. Results The results showed that significant differences of lymph node metastasis (p = 0.003), surgical margin status (p = 0.020), and stromal invasion status (p = 0.004) existed between lVI(−) and LVI(+) patients. CD3+ TILs in the central tumor area (p = 0.010), CD4+ TILs in the central tumor area (p = 0.045), CD8 + TILs in the central tumor area (p = 0.033), and CD8+ TILs in the invasive margin area (p = 0.004) showed significant differences between lVI(−) and LVI(+) patients. When patients were grouped by status of lymph node metastasis, significant differences of FIGO stage (p = 0.005), LVI status (p = 0.003), CD3+ TILs in the central tumor area (p = 0.045), CD45RO+ TILs in the central tumor area (p = 0.033), and CD45RO+ TILs in the invasive margin area (p = 0.028) were also observed. After the patients were stratified into low-, intermediate-, and high risk groups, significant differences of FIGO stage (p = 0.018), status of lymph node metastasis (p = 0.000), LVI status (p = 0.000), parametrial invasion status (p=0.012), stromal invasion status (p = 0.000), tumor growth pattern (p = 0.015) and tumor size (p = 0.000) were identified among 3 groups of patients, while only CD45RO+ TILs in the invasive margin area (p = 0.018) and FOXP3+ TILs in the central tumor area (p = 0.009) were statistically different among three groups of patients. Spearman’s correlation analysis demonstrated that FIGO stage, LVI status, status of lymph node metastasis, parametrial invasion, stromal invasion status, and tumor size positively correlated with risk stratification (P = 0.005, 0.020, 0.000, 0.022, 0.000, and 0.000 respectively), while CD45RO+ TILs in the invasive margin area and FOXP3+ TILs in the central tumor area showed statistically negative correlation with risk stratification (P = 0.031, 0.009 respectively). Conclusion Our study suggested that CD45RO+ TILs in the invasive margin area and FOXP3+ TILs in the central tumor area might be useful biomarkers for risk stratification in cervical cancer patients. Large cohort studies of cervical cancer patients are required to validate our hypothesis.
Collapse
Affiliation(s)
- Rui Chen
- Department of Pathology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Yi Gong
- Department of Hematology-Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Dongling Zou
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Lifeng Wang
- Department of Pathology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Li Yuan
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Qi Zhou
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| |
Collapse
|
180
|
Jimenez O, Barros MH, De Matteo E, Garcia Lombardi M, Preciado MV, Niedobitek G, Chabay P. M1-like macrophage polarization prevails in young children with classic Hodgkin Lymphoma from Argentina. Sci Rep 2019; 9:12687. [PMID: 31481738 PMCID: PMC6722052 DOI: 10.1038/s41598-019-49015-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 08/08/2019] [Indexed: 12/23/2022] Open
Abstract
The microenvironment in classical Hodgkin lymphoma (cHL) comprises a mixture of different types of cells, which are responsible for lymphoma pathogenesis and progression. Even though microenvironment composition in adult cHL has been largely studied, only few groups studied pediatric cHL, in which both Epstein Barr virus (EBV) infection and age may display a role in their pathogenesis. Furthermore, our group described that EBV is significantly associated with cHL in Argentina in patients under the age of 10 years old. For that reason, our aim was to describe the microenvironment composition in 46 pediatric cHL patients. M1-like polarization status prevailed in the whole series independently of EBV association. On the other hand, in children older than 10 years, a tolerogenic environment illustrated by higher FOXP3 expression was proved, accompanied by a macrophage polarization status towards M2. In contrast, in children younger than 10 years, M1-like was prevalent, along with an increase in cytotoxic GrB+ cells. This study supports the notion that pediatric cHL exhibits a particular tumor microenvironment composition.
Collapse
Affiliation(s)
- O Jimenez
- Multidisciplinary Institute for Investigation in Pediatric Pathologies (IMIPP), CONICET-GCBA, Molecular Biology Laboratory, Pathology Division, Ricardo Gutiérrez Children's Hospital, Buenos Aires, Argentina
| | - M H Barros
- Institute for Pathology, Unfallkrankenhaus Berlin, Berlin, Germany
| | - E De Matteo
- Multidisciplinary Institute for Investigation in Pediatric Pathologies (IMIPP), CONICET-GCBA, Molecular Biology Laboratory, Pathology Division, Ricardo Gutiérrez Children's Hospital, Buenos Aires, Argentina
| | - M Garcia Lombardi
- Oncology Division, Ricardo Gutiérrez Children's Hospital, Buenos Aires, Argentina
| | - M V Preciado
- Multidisciplinary Institute for Investigation in Pediatric Pathologies (IMIPP), CONICET-GCBA, Molecular Biology Laboratory, Pathology Division, Ricardo Gutiérrez Children's Hospital, Buenos Aires, Argentina
| | - G Niedobitek
- Institute for Pathology, Unfallkrankenhaus Berlin, Berlin, Germany
| | - P Chabay
- Multidisciplinary Institute for Investigation in Pediatric Pathologies (IMIPP), CONICET-GCBA, Molecular Biology Laboratory, Pathology Division, Ricardo Gutiérrez Children's Hospital, Buenos Aires, Argentina.
| |
Collapse
|
181
|
Karlsson H, Senkowski W, Fryknäs M, Mansoori S, Linder S, Gullbo J, Larsson R, Nygren P. A novel tumor spheroid model identifies selective enhancement of radiation by an inhibitor of oxidative phosphorylation. Oncotarget 2019; 10:5372-5382. [PMID: 31523395 PMCID: PMC6731106 DOI: 10.18632/oncotarget.27166] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 07/30/2019] [Indexed: 01/08/2023] Open
Abstract
There is a need for preclinical models that can enable identification of novel radiosensitizing drugs in clinically relevant high-throughput experiments. We used a new high-throughput compatible total cell kill spheroid assay to study the interaction between drugs and radiation in order to identify compounds with radiosensitizing activity. Experimental drugs were compared to known radiosensitizers and cytotoxic drugs clinically used in combination with radiotherapy. VLX600, a novel iron-chelating inhibitor of oxidative phosphorylation, potentiated the effect of radiation in tumor spheroids in a synergistic manner. This effect was specific to spheroids and not observed in monolayer cell cultures. In conclusion, the total cell kill spheroid assay is a feasible high-throughput method in the search for novel radiosensitizers. VLX600 shows encouraging characteristics for development as a novel radiosensitizer.
Collapse
Affiliation(s)
- Henning Karlsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Mårten Fryknäs
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Stig Linder
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden.,Cancer Center Karolinska, Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Joachim Gullbo
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Rolf Larsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Peter Nygren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
182
|
Nitric oxide and interactions with reactive oxygen species in the development of melanoma, breast, and colon cancer: A redox signaling perspective. Nitric Oxide 2019; 89:1-13. [DOI: 10.1016/j.niox.2019.04.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 04/09/2019] [Accepted: 04/15/2019] [Indexed: 12/13/2022]
|
183
|
S100B Protein Stimulates Proliferation and Angiogenic Mediators Release through RAGE/pAkt/mTOR Pathway in Human Colon Adenocarcinoma Caco-2 Cells. Int J Mol Sci 2019; 20:ijms20133240. [PMID: 31266264 PMCID: PMC6651655 DOI: 10.3390/ijms20133240] [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: 04/10/2019] [Revised: 06/24/2019] [Accepted: 06/29/2019] [Indexed: 12/15/2022] Open
Abstract
Chronic inflammation and angiogenesis are associated with colonic carcinogenesis. Enteric glia-derived S100B protein has been proposed as an "ideal bridge", linking colonic inflammation and cancer, given its dual ability to up-regulate nuclear factor-kappaB (NF-κB) transcription via receptor for advanced glycation end products (RAGE) signaling and to sequestrate wild type pro-apoptotic wild type (wt)p53. However, its pro-angiogenic effects on cancer cells are still uninvestigated. To this aim, we evaluated the effect of exogenous S100B (0.05-5 µM) protein alone or in the presence of S100B blocking monoclonal antibody (mAb) (1:105-1:104 v/v diluted) on (1) cultured Caco-2 cells proliferation, migration and invasiveness in vitro, respectively by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT)-formazan, wound healing and matrigel invasion assays and (2) its effect on the release of pro-angiogenic factors, such as vascular endothelial growth factor (VEGF) by ELISA and immunofluorescence analyses. The effect of S100B alone or in the presence of S100BmAb was then investigated on RAGE/pAkt/mammalian target of rapamycin (mTOR) signaling pathway by immunoblot analysis. Our results showed that S100B markedly increases proliferation and invasiveness of Caco-2 cells, through the release of pro-angiogenic VEGF and NO paralleled to a significant decrease of wtp53 expression mediated by RAGE-p38 mitogen-activated protein kinase (MAPK)/pAkt-mTOR and hypoxia-inducible factor 1-alpha (HIF1α) pathways. Such effects were counteracted by S100BmAb, indicating that S100B targeting is a potential approach to inhibit colon carcinoma proliferation and angiogenesis.
Collapse
|
184
|
Caires-Dos-Santos L, da Silva SV, Smuczek B, de Siqueira AS, Cruz KSP, Barbuto JAM, Augusto TM, Freitas VM, Carvalho HF, Jaeger RG. Laminin-derived peptide C16 regulates Tks expression and reactive oxygen species generation in human prostate cancer cells. J Cell Physiol 2019; 235:587-598. [PMID: 31254281 DOI: 10.1002/jcp.28997] [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: 09/20/2018] [Revised: 06/01/2019] [Accepted: 06/04/2019] [Indexed: 01/05/2023]
Abstract
Laminin peptides influence cancer biology. We investigated the role of a laminin-derived peptide C16 regulating invadopodia molecules in human prostate cancer cells (DU145). C16 augmented invadopodia activity of DU145 cells, and stimulated expression Tks4, Tks5, cortactin, and membrane-type matrix metalloproteinase 1. Reactive oxygen species generation is also related to invadopodia formation. This prompted us to address whether C16 would induce reactive oxygen species generation in DU145 cells. Quantitative fluorescence and flow cytometry showed that the peptide C16 increased reactive oxygen species in DU145 cells. Furthermore, significant colocalization between Tks5 and reactive oxygen species was observed in C16-treated cells. Results suggested that the peptide C16 increased Tks5 and reactive oxygen species in prostate cancer cells. The role of C16 increasing Tks and reactive oxygen species are novel findings on invadopodia activity.
Collapse
Affiliation(s)
- Livia Caires-Dos-Santos
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Suély V da Silva
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Basilio Smuczek
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.,Department of Biology, UNICENTRO State University, Guarapuava, PR, Brazil
| | - Adriane S de Siqueira
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.,School of Dentistry, Positivo University, Curitiba, PR, Brazil
| | - Karen S P Cruz
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil.,Cell Biology Laboratory, Institute of Health and Biological Sciences, Federal University of Alagoas, Maceio, AL, Brazil.,Faculty of Nutrition, Federal University of Alagoas, Maceio, AL, Brazil
| | - José Alexandre M Barbuto
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Taize M Augusto
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil.,Department of Morphology and Basic Pathology, School of Medicine of Jundiai, Jundiai, SP, Brazil
| | - Vanessa M Freitas
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | | - Ruy G Jaeger
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
185
|
Chen CL, Zhang L, Jiao YR, Zhou Y, Ge QF, Li PC, Sun XJ, Lv Z. miR-134 inhibits osteosarcoma cell invasion and metastasis through targeting MMP1 and MMP3 in vitro and in vivo. FEBS Lett 2019; 593:1089-1101. [PMID: 30977909 DOI: 10.1002/1873-3468.13387] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/07/2019] [Accepted: 04/08/2019] [Indexed: 01/15/2023]
Abstract
miR-134 has been shown to be associated with angiogenesis and the progression of osteosarcoma. This study further assessed the effects of miR-134 expression on osteosarcoma cell migration, invasion, and metastasis in vitro and in a nude mouse xenograft model, exploring the underlying molecular events. Luciferase reporter assays revealed that miR-134 directly targets the 3'-UTRs of MMP1 and MMP3 to reduce their expression in osteosarcoma cells. In conclusion, overexpression of miR-134 suppresses osteosarcoma cell invasion and metastasis through the inhibition of MMP1 and MMP3 expression. We propose miR-134 as an attractive novel therapeutic target for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Cheng-Long Chen
- Second Clinical Medical College of Shanxi Medical University, TaiYuan, China
| | - Long Zhang
- Department of Orthopaedics, Second Hospital of Shanxi Medical University, TaiYuan, China
| | - Yu-Rui Jiao
- Second Clinical Medical College of Shanxi Medical University, TaiYuan, China
| | - Yi Zhou
- First Clinical Medical School of Southern Medical University, GuangZhou, China
| | - Qiao-Feng Ge
- Second Clinical Medical College of Shanxi Medical University, TaiYuan, China
| | - Peng-Cui Li
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Second Hospital of Shanxi Medical University, TaiYuan, China
| | - Xiao-Juan Sun
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Second Hospital of Shanxi Medical University, TaiYuan, China
| | - Zhi Lv
- Department of Orthopaedics, Second Hospital of Shanxi Medical University, TaiYuan, China
| |
Collapse
|
186
|
Ungaro F, Colombo P, Massimino L, Ugolini GS, Correale C, Rasponi M, Garlatti V, Rubbino F, Tacconi C, Spaggiari P, Spinelli A, Carvello M, Sacchi M, Spanò S, Vetrano S, Malesci A, Peyrin-Biroulet L, Danese S, D'Alessio S. Lymphatic endothelium contributes to colorectal cancer growth via the soluble matrisome component GDF11. Int J Cancer 2019; 145:1913-1920. [PMID: 30889293 DOI: 10.1002/ijc.32286] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 01/25/2019] [Accepted: 02/28/2019] [Indexed: 12/17/2022]
Abstract
Colorectal cancer (CRC) is one of the most malignant tumors worldwide. Stromal cells residing in the tumor microenvironment strongly contribute to cancer progression through their crosstalk with cancer cells and extracellular matrix. Here we provide the first evidence that CRC-associated lymphatic endothelium displays a distinct matrisome-associated transcriptomic signature, which distinguishes them from healthy intestinal lymphatics. We also demonstrate that CRC-associated human intestinal lymphatic endothelial cells regulate tumor cell growth via growth differentiation factor 11, a soluble matrisome component which in CRC patients was found to be associated with tumor progression. Our data provide new insights into lymphatic contribution to CRC growth, aside from their conventional role as conduits of metastasis.
Collapse
Affiliation(s)
- Federica Ungaro
- IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | | | - Luca Massimino
- Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | | | - Carmen Correale
- IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center, Milan, Italy
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Milan, Italy
| | | | - Federica Rubbino
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Carlotta Tacconi
- Institute of Pharmaceutical Sciences, Pharmacogenomics, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | - Paola Spaggiari
- Department of Pathology, Humanitas Clinical and Research Center, Milan, Italy
| | - Antonino Spinelli
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,Colon and Rectal Surgery Unit, Humanitas Clinical and Research Center, Milan, Italy
| | - Michele Carvello
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,Colon and Rectal Surgery Unit, Humanitas Clinical and Research Center, Milan, Italy
| | - Matteo Sacchi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,Colon and Rectal Surgery Unit, Humanitas Clinical and Research Center, Milan, Italy
| | - Salvatore Spanò
- IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Stefania Vetrano
- IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Alberto Malesci
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy.,Department of Gastroenterology, Humanitas Clinical and Research Center, Milan, Italy
| | - Laurent Peyrin-Biroulet
- Institut National de la Santé et de la Recherche Médicale U954 and Department of Gastroenterology, Nancy University Hospital, Lorraine University, Nancy, France
| | - Silvio Danese
- IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Silvia D'Alessio
- IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
187
|
Extracellular Acidosis Modulates the Expression of Epithelial-Mesenchymal Transition (EMT) Markers and Adhesion of Epithelial and Tumor Cells. Neoplasia 2019; 21:450-458. [PMID: 30953950 PMCID: PMC6447725 DOI: 10.1016/j.neo.2019.03.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/04/2019] [Accepted: 03/07/2019] [Indexed: 12/12/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is an important process of tumor progression associated with increased metastatic potential. EMT can be activated by external triggers such as cytokines or metabolic parameters (e.g. hypoxia). Since extracellular acidosis is a common finding in tumors, the aim of the study is to analyze its impact on the expression of EMT markers in vitro and in vivo as well as the functional impact on cell adhesion. Therefore, three tumor and two normal epithelial cell lines were incubated for 24 h at pH 6.6 and the expression of EMT markers was studied. In addition, mRNA expression of transcription and metabolic factors related to EMT was measured as well as the functional impact on cell adhesion, either during acidic incubation or after priming cells in an acidic milieu. E-cadherin and N-cadherin were down-regulated in all tumor and normal cell lines studied, whereas vimentin expression increased in only two tumor and one normal cell line. Down-regulation of the cadherins was seen in total protein and to a lesser extent in surface protein. In vivo an increase in N-cadherin and vimentin expression was found. Acidosis up-regulated Twist1 and Acsl1 but down-regulated fumarate hydratase (Fh). Cell adhesion during acidic incubation decreased in AT1 prostate carcinoma cells whereas preceding acidic priming increased their subsequent adhesion. Low tumor pH is able to modulate the expression EMT-related proteins and by this may affect the stability of the tissue structure.
Collapse
|
188
|
Izumi D, Toden S, Ureta E, Ishimoto T, Baba H, Goel A. TIAM1 promotes chemoresistance and tumor invasiveness in colorectal cancer. Cell Death Dis 2019; 10:267. [PMID: 30890693 PMCID: PMC6425043 DOI: 10.1038/s41419-019-1493-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 02/18/2019] [Accepted: 02/28/2019] [Indexed: 12/21/2022]
Abstract
Accumulating evidence suggests that cancer cells with stem cell-like features have higher resistance to chemotherapeutic agents. Herein, we identified T-lymphoma invasion and metastasis-inducing protein-1 (TIAM1) as one of the Wnt-signaling associated genes which drives self-renewal and its expression is upregulated by cancer associated fibroblasts (CAFs). TIAM1 expression was assessed in resected colorectal cancer (CRC) tissues from 300 patients who did or did not respond to chemotherapy. siRNA and CRISPR/Cas9 was used to examine whether the inhibition of TIAM1 affects chemosensitivity of CRC. We demonstrate that stemness through Wnt signaling regulates chemosensitivity and this phenomenon occurs exclusively in cancer stem cells. Subsequently, we established patient-derived CAFs and tested whether the drug sensitivity of CRC cell lines is altered with CAF-derived conditioned medium. High-TIAM1 expression correlated significantly with poor prognosis of CRC patients, and was overexpressed in patients who did not respond to chemotherapy. We demonstrated that the inhibition of TIAM1 enhanced sensitivity to chemotherapeutic drugs and reduced tumor invasiveness in a series of experiments in vitro. Moreover, CAF-derived conditioned media increased stemness and chemoresistance in CRC cell lines through TIAM1 overexpression. In addition, we validated TIAM1 associated drug sensitivity using a xenograft model. We have demonstrated that TIAM1 is overexpressed in CRC tumors from patients who did not respond to chemotherapeutic drugs and levels of TIAM1 expression served as an independent prognostic factor. Mechanistically, CAFs enhanced CRC chemoresistance through TIAM1 overexpression. Collectively, these results suggest that TIAM1 regulates chemosensitivity in tumors and stroma and thus may be an attractive therapeutic target.
Collapse
Affiliation(s)
- Daisuke Izumi
- Center for Gastrointestinal Research, Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA.,Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Department of Surgery, Japan Community Health Care Organization Kumamoto General Hospital, Yatsushiro, Japan
| | - Shusuke Toden
- Center for Gastrointestinal Research, Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
| | - Elsie Ureta
- Center for Gastrointestinal Research, Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
| | - Takatsugu Ishimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,The International Research Center for Medicine Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Ajay Goel
- Center for Gastrointestinal Research, Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA.
| |
Collapse
|
189
|
Gooding AJ, Zhang B, Gunawardane L, Beard A, Valadkhan S, Schiemann WP. The lncRNA BORG facilitates the survival and chemoresistance of triple-negative breast cancers. Oncogene 2019; 38:2020-2041. [PMID: 30467380 PMCID: PMC6430670 DOI: 10.1038/s41388-018-0586-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/10/2018] [Accepted: 10/12/2018] [Indexed: 12/31/2022]
Abstract
Disseminated breast cancer cells employ adaptive molecular responses following cytotoxic therapeutic insult which promotes their survival and subsequent outgrowth. Here we demonstrate that expression of the pro-metastatic lncRNA BORG (BMP/OP-Responsive Gene) is greatly induced within triple-negative breast cancer (TNBC) cells subjected to environmental and chemotherapeutic stresses commonly faced by TNBC cells throughout the metastatic cascade. This stress-mediated induction of BORG expression fosters the survival of TNBC cells and renders them resistant to the cytotoxic effects of doxorubicin both in vitro and in vivo. The chemoresistant traits of BORG depend upon its robust activation of the NF-κB signaling axis via a novel BORG-mediated feed-forward signaling loop, and via its ability to bind and activate RPA1. Indeed, genetic and pharmacologic inhibition of NF-κB signaling or the DNA-binding activity of RPA1 abrogates the pro-survival features of BORG and renders BORG-expressing TNBCs sensitive to doxorubicin-induced cytotoxicity. These findings suggest that therapeutic targeting of BORG or its downstream molecular effectors may provide a novel means to alleviate TNBC recurrence.
Collapse
Affiliation(s)
- Alex J Gooding
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Bing Zhang
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Lalith Gunawardane
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Abigail Beard
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Saba Valadkhan
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH, 44106, USA.
| | - William P Schiemann
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
190
|
Targeting Tumor Microenvironment for Cancer Therapy. Int J Mol Sci 2019; 20:ijms20040840. [PMID: 30781344 PMCID: PMC6413095 DOI: 10.3390/ijms20040840] [Citation(s) in RCA: 826] [Impact Index Per Article: 137.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/12/2019] [Accepted: 02/13/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer development is highly associated to the physiological state of the tumor microenvironment (TME). Despite the existing heterogeneity of tumors from the same or from different anatomical locations, common features can be found in the TME maturation of epithelial-derived tumors. Genetic alterations in tumor cells result in hyperplasia, uncontrolled growth, resistance to apoptosis, and metabolic shift towards anaerobic glycolysis (Warburg effect). These events create hypoxia, oxidative stress and acidosis within the TME triggering an adjustment of the extracellular matrix (ECM), a response from neighbor stromal cells (e.g., fibroblasts) and immune cells (lymphocytes and macrophages), inducing angiogenesis and, ultimately, resulting in metastasis. Exosomes secreted by TME cells are central players in all these events. The TME profile is preponderant on prognosis and impacts efficacy of anti-cancer therapies. Hence, a big effort has been made to develop new therapeutic strategies towards a more efficient targeting of TME. These efforts focus on: (i) therapeutic strategies targeting TME components, extending from conventional therapeutics, to combined therapies and nanomedicines; and (ii) the development of models that accurately resemble the TME for bench investigations, including tumor-tissue explants, “tumor on a chip” or multicellular tumor-spheroids.
Collapse
|
191
|
Goulet CR, Champagne A, Bernard G, Vandal D, Chabaud S, Pouliot F, Bolduc S. Cancer-associated fibroblasts induce epithelial-mesenchymal transition of bladder cancer cells through paracrine IL-6 signalling. BMC Cancer 2019; 19:137. [PMID: 30744595 PMCID: PMC6371428 DOI: 10.1186/s12885-019-5353-6] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 02/05/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs), activated by tumour cells, are the predominant type of stromal cells in cancer tissue and play an important role in interacting with neoplastic cells to promote cancer progression. Epithelial-mesenchymal transition (EMT) is a key feature of metastatic cells. However, the mechanism by which CAFs induce EMT program in bladder cancer cells remains unclear. METHODS To investigate the role of CAFs in bladder cancer progression, healthy primary bladder fibroblasts (HFs) were induced into CAFs (iCAFs) by bladder cancer-derived exosomes. Effect of conditioned medium from iCAFs (CM iCAF) on EMT markers expression of non-invasive RT4 bladder cancer cell line was determined by qPCR and Western blot. IL6 expression in iCAFs was evaluated by ELISA and Western blot. RT4 cell proliferation, migration and invasion were assessed in CM iCAF +/- anti-IL6 neutralizing antibody using cyQUANT assay, scratch test and transwell chamber respectively. We investigated IL6 expression relevance for bladder cancer progression by querying gene expression datasets of human bladder cancer specimens from TCGA and GEO genomic data platforms. RESULTS Cancer exosome-treated HFs showed CAFs characteristics with high expression levels of αSMA and FAP. We showed that the CM iCAF induces the upregulation of mesenchymal markers, such as N-cadherin and vimentin, while repressing epithelial markers E-cadherin and p-ß-catenin expression in non-invasive RT4 cells. Moreover, EMT transcription factors SNAIL1, TWIST1 and ZEB1 were upregulated in CM iCAF-cultured RT4 cells compared to control. We also showed that the IL-6 cytokine was highly expressed by CAFs, and its receptor IL-6R was found on RT4 bladder cancer cells. The culture of RT4 bladder cancer cells with CM iCAF resulted in markedly promoted cell growth, migration and invasion. Importantly, inhibition of CAFs-secreted IL-6 by neutralizing antibody significantly reversed the IL-6-induced EMT phenotype, suggesting that this cytokine is necessary for CAF-induced EMT in the progression of human bladder cancer. Finally, we observed that IL6 expression is up-regulated in aggressive bladder cancer and correlate with CAF marker ACTA2. CONCLUSIONS We conclude that CAFs promote aggressive phenotypes of non-invasive bladder cancer cells through an EMT induced by the secretion of IL-6.
Collapse
Affiliation(s)
- Cassandra Ringuette Goulet
- Centre de recherche en organogénèse expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, QC, Québec Canada
- Department of Surgery, Faculty of Medicine, Laval University, QC, Quebec Canada
- Oncology Division, CHU de Québec Research Center, QC, Quebec Canada
| | - Audrey Champagne
- Department of Surgery, Faculty of Medicine, Laval University, QC, Quebec Canada
- Oncology Division, CHU de Québec Research Center, QC, Quebec Canada
| | - Geneviève Bernard
- Centre de recherche en organogénèse expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, QC, Québec Canada
- Department of Surgery, Faculty of Medicine, Laval University, QC, Quebec Canada
| | - Dominique Vandal
- Centre de recherche en organogénèse expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, QC, Québec Canada
| | - Stéphane Chabaud
- Centre de recherche en organogénèse expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, QC, Québec Canada
- Department of Surgery, Faculty of Medicine, Laval University, QC, Quebec Canada
| | - Frédéric Pouliot
- Department of Surgery, Faculty of Medicine, Laval University, QC, Quebec Canada
- Oncology Division, CHU de Québec Research Center, QC, Quebec Canada
| | - Stéphane Bolduc
- Centre de recherche en organogénèse expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, QC, Québec Canada
- Department of Surgery, Faculty of Medicine, Laval University, QC, Quebec Canada
- Centre de recherche du CHU de Québec-Université Laval, Centre de recherche en organogénèse expérimentale de l’Université Laval/LOEX, 1401, 18e rue, Quebec city, Québec G1J 1Z4 Canada
| |
Collapse
|
192
|
|
193
|
Kim JY, Kim YM. Tumor endothelial cells as a potential target of metronomic chemotherapy. Arch Pharm Res 2019; 42:1-13. [PMID: 30604201 DOI: 10.1007/s12272-018-01102-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022]
Abstract
Drug resistance and toxic side effects are major therapeutic hurdles affecting cancer patients receiving conventional chemotherapy based on the maximum tolerated dose. Metronomic chemotherapy (MCT), a new therapeutic approach developed to avoid these problems generally, consists of the continuous administration of low-dose cytotoxic agents without extended intervals. This therapy targets the tumor microenvironment, rather than exerting a direct effect on tumor cells. As a result, the MCT regimen functionally impairs tumor endothelial cells and circulating endothelial progenitor cells, leading to tumor dormancy via anti-angiogenesis. Over the past 10 years, several studies have highlighted the impact of MCT on the tumor microenvironment and angiogenesis and demonstrated its potential as a switch from the pro-angiogenic to the anti-angiogenic state. However, the mechanisms of action are still obscure. Here, we systematically review the evidence regarding the anti-angiogenic potential of MCT as a crucial determinant of tumor dormancy and cancer treatment.
Collapse
Affiliation(s)
- Ji Yoon Kim
- Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul, 04763, South Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry School of Medicine, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, 24341, South Korea.
| |
Collapse
|
194
|
Yin Z, Jiang K, Li R, Dong C, Wang L. Multipotent mesenchymal stromal cells play critical roles in hepatocellular carcinoma initiation, progression and therapy. Mol Cancer 2018; 17:178. [PMID: 30593276 PMCID: PMC6309092 DOI: 10.1186/s12943-018-0926-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 12/16/2018] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, with high morbidity, relapse and mortality rates. Multipotent mesenchymal stromal cells (MSCs) can be recruited to and become integral components of the HCC microenvironment and can influence tumor progression. This review discusses MSC migration to liver fibrosis and the HCC microenvironment, MSC involvement in HCC initiation and progression and the widespread application of MSCs in HCC-targeted therapy, thus clarifying the critical roles of MSCs in HCC.
Collapse
Affiliation(s)
- Zeli Yin
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027, Liaoning, China
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, 116027, Liaoning, China
- Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, 116027, Liaoning, China
| | - Keqiu Jiang
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027, Liaoning, China
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, 116027, Liaoning, China
- Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, 116027, Liaoning, China
| | - Rui Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027, Liaoning, China
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, 116027, Liaoning, China
- Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, 116027, Liaoning, China
| | - Chengyong Dong
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027, Liaoning, China.
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, 116027, Liaoning, China.
- Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, 116027, Liaoning, China.
| | - Liming Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027, Liaoning, China.
- Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, 116027, Liaoning, China.
- Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, 116027, Liaoning, China.
| |
Collapse
|
195
|
Yang Y, Zhang L, La X, Li Z, Li H, Guo S. Salvianolic acid A inhibits tumor-associated angiogenesis by blocking GRP78 secretion. Naunyn Schmiedebergs Arch Pharmacol 2018; 392:467-480. [DOI: 10.1007/s00210-018-1585-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/12/2018] [Indexed: 12/31/2022]
|
196
|
Matteucci C, Balestrieri E, Argaw-Denboba A, Sinibaldi-Vallebona P. Human endogenous retroviruses role in cancer cell stemness. Semin Cancer Biol 2018; 53:17-30. [PMID: 30317035 DOI: 10.1016/j.semcancer.2018.10.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/30/2018] [Accepted: 10/05/2018] [Indexed: 12/18/2022]
Abstract
Cancer incidence and mortality, metastasis, drug resistance and recurrence are still the critical issues of oncological diseases. In this scenario, increasing scientific evidences demonstrate that the activation of human endogenous retroviruses (HERVs) is involved in the aggressiveness of tumors such as melanoma, breast, germ cell, renal, ovarian, liver and haematological cancers. In their dynamic regulation, HERVs have also proved to be important determinants of pluripotency in human embryonic stem cells (ESC) and of the reprogramming process of induced pluripotent stem cells (iPSCs). In many types of tumors, essential characteristics of aggressiveness have been associated with the achievement of stemness features, often accompanied with the identification of defined subpopulations, termed cancer stem cells (CSCs), which possess stem cell-like properties and sustain tumorigenesis. Indeed, CSCs show high self-renewal capacity with a peculiar potential in tumor initiation, progression, metastasis, heterogeneity, recurrence, radiotherapy and drug resistance. However, HERVs role in CSCs biology is still not fully elucidated. In this regard, CD133 is a widely recognized marker of CSCs, and our group demonstrated, for the first time, the requirement of HERV-K activation to expand and maintain a CD133+ melanoma cell subpopulation with stemness features in response to microenvironmental modifications. The review will discuss HERVs expression as cancer hallmark, with particular focus on their role in the regulation of cancer stemness features and the potential involvement as targets for therapy.
Collapse
Affiliation(s)
- Claudia Matteucci
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Via Montpellier, 1, 00133, Rome, Italy.
| | - Emanuela Balestrieri
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Via Montpellier, 1, 00133, Rome, Italy
| | - Ayele Argaw-Denboba
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Via Montpellier, 1, 00133, Rome, Italy; European Molecular Biology Laboratory (EMBL), Adriano Buzzati-Traverso Campus, Monterotondo, Rome, Italy
| | - Paola Sinibaldi-Vallebona
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Via Montpellier, 1, 00133, Rome, Italy; Institute of Translational Pharmacology, National Research Council, Via Fosso del Cavaliere, 100, 00133, Rome, Italy
| |
Collapse
|
197
|
Zhou Y, Su Y, Zhu H, Wang X, Li X, Dai C, Xu C, Zheng T, Mao C, Chen D. Interleukin-23 receptor signaling mediates cancer dormancy and radioresistance in human esophageal squamous carcinoma cells via the Wnt/Notch pathway. J Mol Med (Berl) 2018; 97:177-188. [PMID: 30483821 PMCID: PMC6348073 DOI: 10.1007/s00109-018-1724-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/30/2018] [Accepted: 11/13/2018] [Indexed: 01/05/2023]
Abstract
Abstract In the tumor microenvironment, inflammatory cells and molecules influence almost every process; among them, interleukin-23 (IL-23) is a pro-inflammatory molecule that exhibits pro- or anti-tumor properties, but both activities remain poorly understood. In this study, we investigated the effect of extracellular IL-23 in IL-23 receptor-positive (IL-23R+) esophageal squamous cell carcinoma (ESCC) and explored the mechanisms underlying this effect. We analyzed ESCC tumor tissues by immunohistochemical and immunofluorescence staining and found that IL-23, which was highly expressed, co-localized with Oct-4A in IL-23R+ ESCC cells. In addition, IL-23 treatment significantly increased the accumulation of CD133+ cells and activated the Wnt and Notch signaling pathways in CD133−IL-23R+ ESCC cell lines. Consistently, CD133−IL-23R+ cells pretreated with IL-23 showed stronger anti-apoptosis activity when exposed to radiation and higher survival than untreated groups. Moreover, the inhibition of Wnt/Notch signaling by a small-molecule inhibitor or siRNA abolished the effect of IL-23-induced dormancy and consequent radioresistance. Taken together, these results suggested that IL-23 facilitates radioresistance in ESCC by activating Wnt/Notch-mediated G0/1 phase arrest, and attenuating these detrimental changes by blocking the formation of dormancy may prove to be an effective pretreatment for radiotherapy. Key messages IL-23/IL-23R is correlated with the acquisition of stem-like potential in ESCC. CD133−IL-23R+ ESCCs acquired dormancy via IL-23. Radioresistance depends on IL-23-mediated Wnt/Notch pathway activation in vitro and vivo.
Electronic supplementary material The online version of this article (10.1007/s00109-018-1724-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuepeng Zhou
- Institute of Oncology, Affiliated Hospital of Jiangsu University, Jiefang Road 438, Zhenjiang, 212001, China
| | - Yuting Su
- Institute of Oncology, Affiliated Hospital of Jiangsu University, Jiefang Road 438, Zhenjiang, 212001, China
| | - Haitao Zhu
- Department of Medical Imaging, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Xuefeng Wang
- Department of Nuclear Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Xiaoqin Li
- Institute of Oncology, Affiliated Hospital of Jiangsu University, Jiefang Road 438, Zhenjiang, 212001, China
| | - Chunhua Dai
- Institute of Oncology, Affiliated Hospital of Jiangsu University, Jiefang Road 438, Zhenjiang, 212001, China
| | - Chengcheng Xu
- Department of Nuclear Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Tingting Zheng
- Department of Nuclear Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Chaoming Mao
- Institute of Oncology, Affiliated Hospital of Jiangsu University, Jiefang Road 438, Zhenjiang, 212001, China.
- Department of Nuclear Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| | - Deyu Chen
- Institute of Oncology, Affiliated Hospital of Jiangsu University, Jiefang Road 438, Zhenjiang, 212001, China.
| |
Collapse
|
198
|
Higher Ki67 expression in fibroblast like cells at invasive front indicates better clinical outcomes in oral squamous cell carcinoma patients. Biosci Rep 2018; 38:BSR20181271. [PMID: 30341240 PMCID: PMC6246770 DOI: 10.1042/bsr20181271] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/21/2018] [Accepted: 10/03/2018] [Indexed: 01/05/2023] Open
Abstract
Background: Ki67 has been a key role for the treatment options and prognosis evaluation in some kinds of tumors; however, the spatial expression of Ki67 in oral squamous cell carcinoma (OSCC) has not been fully-evaluated. Therefore, in the present study, we aimed to elucidate the prognosis value of Ki67 spatial expression including in different cell types and at different compartments of tumor in OSCC patients. Methods: Immunohistochemical expression of Ki67 in tumor cells (TCs) and fibroblast like cells (FLCs) at center of tumor (CT) and invasive front (IF) was evaluated in 109 OSCC patients. Then correlations of Ki67 expressions with clinicopathological parameters were analyzed by Chi-square test, and survival curves were evaluated by Kaplan-Meier methods. Furthermore, univariate and multivariate analysis were performed to assess the diagnostic values of Ki67 expression by the Cox regression model. Results: Ki67 expression in TCs was much higher than in FLCs both at CT and IF compartments, but Ki67 expression in TCs was simultaneously higher at CT than that at IF (P=0.0004), which was converse to Ki67 expression in FLCs (P<0.0001). Additionally, high Ki67 expression in FLCs at IF was significantly associated with poor tumor differentiation (P=0.003), worse depth of invasion (DOI, P=0.027) and worst pattern of invasion (WPOI, P=0.041), but Ki67 expression in TCs had no correlation with clinical parameters no matter at CT or IF. Moreover, patients with higher Ki67 expression in TCs at CT had significantly increased risk for OS (overall survival; HR:1.935, 95% CI: 1.181-4.823, P=0.0395) and DFS (disease-free survival; HR: 2.974, 95% CI:1.189-5.023, P=0.046). On contrary, higher Ki67 expression in FLCs at IF was correlated with better OS (HR: 0.15, 95% CI: 0.018-0.846, P=0.0396) and DFS (HR: 0.15, 95% CI: 0.018-0.947, P=0.0445). Whereas, Ki67 expression both at TCs in IF and at FLCs in CT had no significant prognostic value for OS and DFS. Furthermore, Cox multivariate analysis revealed that Ki67 expression in FLCs at IF could not be an independent prognostic factor for OSCC patients. Conclusion: These results show that higher Ki67 expression in FLCs at IF indicated better clinical outcomes for OSCC patients.
Collapse
|
199
|
ZNF32 induces anoikis resistance through maintaining redox homeostasis and activating Src/FAK signaling in hepatocellular carcinoma. Cancer Lett 2018; 442:271-278. [PMID: 30439540 DOI: 10.1016/j.canlet.2018.09.033] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/07/2018] [Accepted: 09/06/2018] [Indexed: 02/06/2023]
Abstract
Tumor cells need to attain anoikis resistance to survive prior to metastasis making it a vital trait of malignancy. The molecular mechanism by which hepatocellular carcinoma (HCC) cells resist anoikis remains not fully understood. Here, we report that ZNF32 expression is markedly upregulated in HCC cells upon detachment. Enforced ZNF32 expression significantly promotes the anchorage-independent growth capability of HepG2 and Huh7 cells, whereas knockdown of ZNF32 results in increased apoptosis of HCC cells after detachment. Mechanistically, we demonstrate that ZNF32 overexpression suppresses the reactive oxygen species (ROS) accumulation and maintains mitochondrial membrane potential, leading to ATP, GSH and NADPH elevation and promoting HCC cell survival in response to suspension. Moreover, ZNF32 enhances the phosphorylation and activation of Src/FAK signaling. Src and FAK inhibitors effectively reverse ZNF32-induced anoikis resistance in HCC cells. Collectively, our findings not only reveal a novel and important mechanism by which ZNF32 contributes to anoikis resistance through maintaining redox homeostasis and activating Src/FAK signaling, but also suggest the potential therapeutic value of ZNF32 in HCC patients.
Collapse
|
200
|
Zhou S, He Y, Yang S, Hu J, Zhang Q, Chen W, Xu H, Zhang H, Zhong S, Zhao J, Tang J. The regulatory roles of lncRNAs in the process of breast cancer invasion and metastasis. Biosci Rep 2018; 38:BSR20180772. [PMID: 30217944 PMCID: PMC6165837 DOI: 10.1042/bsr20180772] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 09/03/2018] [Accepted: 09/11/2018] [Indexed: 12/28/2022] Open
Abstract
Breast cancer (BC) is the most common cancer and principal cause of death among females worldwide. Invasion and metastasis are major causes which influence the survival and prognosis of BC. Therefore, to understand the molecule mechanism underlying invasion and metastasis is paramount for developing strategies to improve survival and prognosis in BC patients. Recent studies have reported that long non-coding RNAs (lncRNAs) play critical roles in the regulation of BC invasion and metastasis through a variety of molecule mechanisms that endow cells with an aggressive phenotype. In this article, we focused on the function of lncRNAs on BC invasion and metastasis through participating in epithelial-to-mesenchymal transition, strengthening cancer stem cells generation, serving as competing endogenous lncRNAs, influencing multiple signaling pathways as well as regulating expressions of invasion-metastasis related factors, including cells adhesion molecules, extracellular matrix, and matrix metallo-proteinases. The published work described has provided a better understanding of the mechanisms underpinning the contribution of lncRNAs to BC invasion and metastasis, which may lay the foundation for the development of new strategies to prevent BC invasion and metastasis.
Collapse
Affiliation(s)
- Siying Zhou
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Xianlin Road 138, Nanjing 210023, P.R. China
| | - Yunjie He
- The First Clinical School of Nanjing Medical University, Nanjing 210029, P.R. China
| | - Sujin Yang
- The First Clinical School of Nanjing Medical University, Nanjing 210029, P.R. China
| | - Jiahua Hu
- The Fourth Clinical School of Nanjing Medical University, Nanjing 210029, P.R. China
- Center of Clinical Laboratory Science, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Baiziting 42, Nanjing 210029, P.R. China
| | - Qian Zhang
- The First Clinical School of Nanjing Medical University, Nanjing 210029, P.R. China
| | - Wei Chen
- Department of Head and Neck Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Baiziting 42, Nanjing 210029, P.R. China
| | - Hanzi Xu
- Department of Radiotherapy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Baiziting 42, Nanjing 210029, P.R. China
| | - Heda Zhang
- Department of General Surgery, School of Medicine, Southeast University, 87 Ding Jia Qiao, Nanjing 210009, P.R. China
| | - Shanliang Zhong
- Center of Clinical Laboratory Science, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Baiziting 42, Nanjing 210029, P.R. China
| | - Jianhua Zhao
- Center of Clinical Laboratory Science, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Baiziting 42, Nanjing 210029, P.R. China
| | - Jinhai Tang
- Department of General Surgery, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, P.R. China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Xianlin Road 138, Nanjing 210023, P.R. China
| |
Collapse
|