1
|
Rass A, Eksteen C, Engelbrecht AM. Paracrine signalling in breast cancer: Insights into the tumour endothelial phenotype. Acta Histochem 2024; 126:152191. [PMID: 39216306 DOI: 10.1016/j.acthis.2024.152191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Tumour endothelial cells (TECs) are genetically and phenotypically distinct from their normal, healthy counterparts and provide various pro-tumourigenic effects. This study aimed to investigate the impact of conditioned media (CM) from non-tumourigenic MCF-12A breast epithelial cells as well as from MCF-7 and MDA-MB-231 breast cancer cells on human umbilical vein endothelial cells (HUVECs). Significant increases in cell viability were observed across all breast CM groups compared to controls, with notable differences between the MCF-12A, MCF-7, and MDA-MB-231 groups. Despite increased viability, no significant differences in MCM2 expression, a marker of cell proliferation, were detected. Morphological changes in HUVECs, including elongation, lumen formation, and branching, were more pronounced in breast cancer CM groups, especially in the MDA-MB-231 CM group. qPCR and Western blot analyses showed increased expression of TEC markers such as MDR1, LOX, and TEM8 in HUVECs treated with MCF-12A CM. The MCF-7 CM group significantly enhanced HUVEC migratory activity compared to MCF-12A CM, as evidenced by a scratch assay. These findings underscore distinct angiogenic responses elicited by non-tumourigenic and tumourigenic breast epithelial cells, with tumourigenic cells inducing a hyperactivated angiogenic response. The study highlights the differential effects of breast cancer cell paracrine signalling on endothelial cells and suggests the need for further investigation into TEC markers' role in both physiological and tumour angiogenesis.
Collapse
Affiliation(s)
- Atarah Rass
- Department of Physiological Sciences, Stellenbosch University, 2nd floor, Mike De Vries Building, Cnr. Merriman Ave & Bosman Street, Stellenbosch, South Africa.
| | - Carla Eksteen
- Department of Physiological Sciences, Stellenbosch University, 2nd floor, Mike De Vries Building, Cnr. Merriman Ave & Bosman Street, Stellenbosch, South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Stellenbosch University, 2nd floor, Mike De Vries Building, Cnr. Merriman Ave & Bosman Street, Stellenbosch, South Africa; African Cancer Institute (ACI), Department of Global Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
2
|
Morimoto M, Maishi N, Hida K. Acquisition of drug resistance in endothelial cells by tumor-derived extracellular vesicles and cancer progression. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:1. [PMID: 38318528 PMCID: PMC10838380 DOI: 10.20517/cdr.2023.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/17/2023] [Indexed: 02/07/2024]
Abstract
Angiogenesis by endothelial cells (ECs) is essential for tumor growth. Angiogenesis inhibitors are used in combination with anticancer drugs in many tumor types, but tumors eventually become resistant. Previously, the underlying mechanism for developing drug resistance was considered to be a change in the characteristics of tumor cells whereas ECs were thought to be genetically stable and do not contribute to drug resistance. However, tumor endothelial cells (TECs) have been shown to differ from normal endothelial cells (NECs) in that they exhibit chromosomal abnormalities, angiogenic potential, and drug resistance. Extracellular vesicles (EVs) secreted by tumor cells have recently attracted attention as a factor involved in the acquisition of such abnormalities. Various cells communicate with each other through EVs, and it has been reported that tumor-derived EVs act on other tumor cells or stromal cells to develop drug resistance. Drug-resistant tumor cells confer drug resistance to recipient cells by transporting mRNAs encoding ATP-binding cassette subfamily B member 1 (ABCB1) and ATP-binding cassette subfamily C member 1 (ABCC1) as well as miRNAs involved in signaling such as Akt, drug efflux transporters, and P-glycoprotein modulators via EVs. However, there are limited reports on the acquisition of drug resistance in ECs by tumor-derived EVs. Since drug resistance of ECs may induce tumor metastasis and support tumor cell proliferation, the mechanism underlying the development of resistance should be elucidated to find therapeutic application. This review provides insight into the acquisition of drug resistance in ECs via tumor EVs in the tumor microenvironment.
Collapse
Affiliation(s)
- Masahiro Morimoto
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo 060-8586, Japan
- Department of Oral Diagnosis and Medicine, Hokkaido University Faculty of Dental Medicine, Sapporo 060-8586, Japan
| | - Nako Maishi
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo 060-8586, Japan
| | - Kyoko Hida
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo 060-8586, Japan
| |
Collapse
|
3
|
Andreadis D, Zisis V, Anastasiadou P, Anagnostou L, Paraskevopoulos K, Poulopoulos A. Aldehyde Dehydrogenase: An Off-Label Marker of Endothelial Activation in Oral Squamous Cell Carcinoma. Cureus 2023; 15:e41596. [PMID: 37559839 PMCID: PMC10408774 DOI: 10.7759/cureus.41596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2023] [Indexed: 08/11/2023] Open
Abstract
The vascular endothelial (VE) expression of aldehyde dehydrogenase (ALDH) 1/2 family in oral leukoplakia (OL) and oral squamous cell carcinoma (OSCC) cases has not been studied so far. The aim of this study was to illustrate the "off-label" endothelial expression of cancer stem cell (CSC) biomarker, ALDH1/2, adjacent to oral potentially malignant and malignant lesions in order to shed some light on the mechanisms leading to oral carcinogenesis. Materials and methods: The expression of CSC protein-biomarker ALDH1/2 was detected through immunohistochemistry (IHC) in 30 paraffin-embedded samples of OL and 21 samples of OSCC compared to five samples of normal oral mucosa. Statistical analysis was done using SPSS, Pearson Chi-square, and Fischer's exact test. The significance level was set at 0.05 (p≤ 0.05). Results: In oral mucosal vessels, ALDH1/2 was not expressed. It was expressed significantly more in the vessels of OSCCs compared to the OLs (Fisher's exact test, p-value= 0,001). However, when endothelial expression of ALDH1/2 in the vasculature of OLs was compared with that of the normal oral mucosa, no significant change was noticed (Fisher's exact test, p-value=1.000). Discussion: The IHC VE expression of ALDH1/2 in OSCC vasculature but not in OL indicates a possible significantly stronger activation of endothelial cells during carcinogenesis, which could be an indicator of the role of inflammation in the development of field cancerization and of prognostic value for (vascular/lymphatic) metastasis.
Collapse
Affiliation(s)
- Dimitrios Andreadis
- Oral Medicine/Pathology, Aristotle University of Thessaloniki, Thessaloniki, GRC
| | - Vasileios Zisis
- Oral Medicine/Pathology, Aristotle University of Thessaloniki, Thessaloniki, GRC
| | | | - Lefteris Anagnostou
- Oral Medicine/Pathology, Aristotle University of Thessaloniki, Thessaloniki, GRC
| | | | | |
Collapse
|
4
|
Tsumita T, Maishi N, Annan DAM, Towfik MA, Matsuda A, Onodera Y, Nam JM, Hida Y, Hida K. The oxidized-LDL/LOX-1 axis in tumor endothelial cells enhances metastasis by recruiting neutrophils and cancer cells. Int J Cancer 2022; 151:944-956. [PMID: 35608341 DOI: 10.1002/ijc.34134] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 04/24/2022] [Accepted: 05/12/2022] [Indexed: 11/07/2022]
Abstract
Epidemiological relationships between cancer and cardiovascular diseases have been reported, but a molecular basis remains unclear. Some proteoglycans that strongly bind low-density-lipoprotein (LDL) are abundant both in atherosclerotic regions and in high metastatic-tumor tissue. LDL retention is crucial for the initiation of atherosclerosis, although its contribution to malignancy of cancer is not known. In this study, we show the importance of the accumulation of LDL in tumor metastasis. We demonstrated that high metastatic-tumor tissue contains high amounts of LDL and forms more oxidized LDL (ox-LDL). Interestingly, lectin-like ox-LDL receptor 1 (LOX-1), a receptor for ox-LDL and a recognized key molecule for cardiovascular diseases, was highly expressed in tumor endothelial cells (TECs). Neutrophils are important for ox-LDL formation. Since we observed the accumulation and activation of neutrophils in HM-tumors, we evaluated the involvement of LOX-1 in neutrophil migration and activation. LOX-1 induced neutrophil migration via CCL2 secretion from TECs, which was enhanced by ox-LDL. Finally, we show genetic manipulation of LOX-1 expression in TECs or tumor stroma tended to reduce lung metastasis. Thus, the LOX-1/ox-LDL axis in TECs may lead to the formation of a high metastatic-tumor microenvironment via attracting neutrophils. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Takuya Tsumita
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
- JSPS Research Fellow for Young Scientists, Tokyo, Japan
| | - Nako Maishi
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Dorcas Akuba-Muhyia Annan
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
- Accra College of Medicine, Accra, Ghana
- West African Genetic Medicine Centre, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Mohammad Alam Towfik
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Aya Matsuda
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Yasuhito Onodera
- Global Center for Biomedical Science and Engineering (GCB), Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Jin-Min Nam
- Global Center for Biomedical Science and Engineering (GCB), Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yasuhiro Hida
- Department of Thoracic Surgery, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Kyoko Hida
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| |
Collapse
|
5
|
Maishi N, Sakurai Y, Hatakeyama H, Umeyama Y, Nakamura T, Endo R, Alam MT, Li C, Annan DAM, Kikuchi H, Morimoto H, Morimoto M, Akiyama K, Ohga N, Hida Y, Harashima H, Hida K. Novel antiangiogenic therapy targeting biglycan using tumor endothelial cell-specific liposomal siRNA delivery system. Cancer Sci 2022; 113:1855-1867. [PMID: 35266253 PMCID: PMC9128192 DOI: 10.1111/cas.15323] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 02/21/2022] [Accepted: 03/02/2022] [Indexed: 12/01/2022] Open
Abstract
Tumor blood vessels play important roles in tumor progression and metastasis. Targeting tumor endothelial cells (TECs) is one of the strategies for cancer therapy. We previously reported that biglycan, a small leucine‐rich proteoglycan, is highly expressed in TECs. TECs utilize biglycan in an autocrine manner for migration and angiogenesis. Furthermore, TEC‐derived biglycan stimulates tumor cell migration in a paracrine manner leading to tumor cell intravasation and metastasis. In this study, we explored the therapeutic effect of biglycan inhibition in the TECs of renal cell carcinoma using an in vivo siRNA delivery system known as a multifunctional envelope‐type nanodevice (MEND), which contains a unique pH‐sensitive cationic lipid. To specifically deliver MEND into TECs, we incorporated cyclo(Arg–Gly–Asp–d–Phe–Lys) (cRGD) into MEND because αVβ3 integrin, a receptor for cRGD, is selective and highly expressed in TECs. We developed RGD‐MEND‐encapsulating siRNA against biglycan. First, we confirmed that MEND was delivered into OS‐RC‐2 tumor‐derived TECs and induced in vitro RNAi‐mediated gene silencing. MEND was then injected intravenously into OS‐RC‐2 tumor‐bearing mice. Flow cytometry analysis demonstrated that MEND was specifically delivered into TECs. Quantitative RT‐PCR indicated that biglycan was knocked down by biglycan siRNA‐containing MEND. Finally, we analyzed the therapeutic effect of biglycan silencing by MEND in TECs. Tumor growth was inhibited by biglycan siRNA‐containing MEND. Tumor microenvironmental factors such as fibrosis were also normalized using biglycan inhibition in TECs. Biglycan in TECs can be a novel target for cancer treatment.
Collapse
Affiliation(s)
- Nako Maishi
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Yu Sakurai
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan.,Membrane Transport and Drug Targeting Laboratory, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hiroto Hatakeyama
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan.,Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Yui Umeyama
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Rikito Endo
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Mohammad Towfik Alam
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Cong Li
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Dorcas Akuba-Muhyia Annan
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroshi Kikuchi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hirofumi Morimoto
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Masahiro Morimoto
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Oral Diagnosis and Medicine, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Kosuke Akiyama
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Noritaka Ohga
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Department of Oral Diagnosis and Medicine, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Yasuhiro Hida
- Department of Cardiovascular and Thoracic Surgery, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | | | - Kyoko Hida
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| |
Collapse
|
6
|
Floranović MP, Petrović AR, Veličković LJ. Expression of the CXCR4 and CXCR7 in renal cancers; can "the orphan receptor" predict the mortality? Ann Diagn Pathol 2021; 55:151829. [PMID: 34563828 DOI: 10.1016/j.anndiagpath.2021.151829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/26/2021] [Accepted: 09/05/2021] [Indexed: 02/05/2023]
Abstract
CXCR4 and CXCR7 are chemokine receptors that bind with chemokine CXCL12 and influence various physiological and pathological processes. In renal cell carcinoma, their expression has been mostly associated with tumour aggressiveness. However, there are some contradictory results regarding the localization of immunohistochemical staining and predictive potential of these markers. The expression of CXCR4 and CXCR7 was immunohistochemicaly analyzed in 98 tumour samples, including 85 clear cell type (ccRCC) and 13 papillary type (pRCC). Depending on the staining localization (cytoplasmatic or membranous), intensity and percentage of stained cells, histoscores were calculated, and their association with clinicopathological parameters was analyzed. PRCC was associated with both CXCR7 and CXCR4 cytoplasmatic expression. We have also found that higher CXCR7 expression can be expected in tumours of greater size. In our study, mortality could be predicted by membranous CXCR7 histoscore, tumour size and pRCC type. With each centimetre in tumour size, survival decreases 1.2 times. CXCR7M histoscore higher by 50 units was associated with 1.5 greater risk of mortality. Neither membranous nor cytoplasmatic CXCR4 histoscore was found to be mortality predictor. Our data showed that CXCR7 could be considered as a valid prognostic marker regarding survival of RCC patients.
Collapse
Affiliation(s)
- Milena Potić Floranović
- Scientific Research Center for Biomedicine, Faculty of Medicine, University of Niš, Zoran Đinđić Boulevard 81, 18000 Niš, Serbia.
| | - Ana Ristić Petrović
- Pathology and Pathological Anatomy Center - Clinical Center of Niš, Zoran Đinđić Boulevard 48, 18000 Niš, Serbia
| | - Ljubinka Janković Veličković
- Pathology and Pathological Anatomy Center - Clinical Center of Niš, Zoran Đinđić Boulevard 48, 18000 Niš, Serbia
| |
Collapse
|
7
|
Expression and prognostic value of CXCL12/CXCR4/CXCR7 axis in clear cell renal cell carcinoma. Clin Exp Nephrol 2021; 25:1057-1069. [PMID: 34109508 DOI: 10.1007/s10157-021-02081-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/18/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND CXCL12 or stromal-derived factor-1 is a chemokine that binds to two receptors CXCR4 and CXCR7 and takes part in both physiological and pathological cell functions. The disruption of the CXCL12/CXCR4/CXCR7 chemokine axis is seen in various types of cancers. METHODS We have immunohistochemically analyzed the expression of CXCL12 and its receptors in clear cell renal cell carcinoma patients. The study included 85 tissue samples. Since samples exhibited heterogeneity of expression intensity and staining localization (cytoplasmatic and membranous), histoscores were calculated, and their associations with clinicopathological parameters were analyzed. RESULTS Both cytoplasmatic CXCR7 and CXCL12 histoscores were associated with greater tumour size, while CXCL12 staining was associated with a higher grade as well. Mortality was associated with tumour size and both membranous and cytoplasmatic CXCL12 histoscores. With each centimetre in tumour size, survival decreases 1.3 times, while CXCL12C histoscore higher than 73 was associated with 2.3 greater risk of mortality. CXCR4 histoscore could only be predicted by female gender and neither cytoplasmatic nor membranous CXCR4 expression was found to be a mortality predictor. CONCLUSION Our data suggest that regarding overall survival, CXCL12 could be considered a valuable prognostic marker.
Collapse
|
8
|
Santagata S, Ieranò C, Trotta AM, Capiluongo A, Auletta F, Guardascione G, Scala S. CXCR4 and CXCR7 Signaling Pathways: A Focus on the Cross-Talk Between Cancer Cells and Tumor Microenvironment. Front Oncol 2021; 11:591386. [PMID: 33937018 PMCID: PMC8082172 DOI: 10.3389/fonc.2021.591386] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 03/25/2021] [Indexed: 12/14/2022] Open
Abstract
The chemokine receptor 4 (CXCR4) and 7 (CXCR7) are G-protein-coupled receptors (GPCRs) activated through their shared ligand CXCL12 in multiple human cancers. They play a key role in the tumor/tumor microenvironment (TME) promoting tumor progression, targeting cell proliferation and migration, while orchestrating the recruitment of immune and stromal cells within the TME. CXCL12 excludes T cells from TME through a concentration gradient that inhibits immunoactive cells access and promotes tumor vascularization. Thus, dual CXCR4/CXCR7 inhibition will target different cancer components. CXCR4/CXCR7 antagonism should prevent the development of metastases by interfering with tumor cell growth, migration and chemotaxis and favoring the frequency of T cells in TME. Herein, we discuss the current understanding on the role of CXCL12/CXCR4/CXCR7 cross-talk in tumor progression and immune cells recruitment providing support for a combined CXCR4/CXCR7 targeting therapy. In addition, we consider emerging approaches that coordinately target both immune checkpoints and CXCL12/CXCR4/CXCR7 axis.
Collapse
Affiliation(s)
- Sara Santagata
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| | - Caterina Ieranò
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| | - Anna Maria Trotta
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| | - Anna Capiluongo
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| | - Federica Auletta
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| | - Giuseppe Guardascione
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| | - Stefania Scala
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| |
Collapse
|
9
|
Yanagiya M, Dawood RIH, Maishi N, Hida Y, Torii C, Annan DA, Kikuchi H, Yanagawa Matsuda A, Kitamura T, Ohiro Y, Shindoh M, Tanaka S, Kitagawa Y, Hida K. Correlation between endothelial CXCR7 expression and clinicopathological factors in oral squamous cell carcinoma. Pathol Int 2021; 71:383-391. [PMID: 33783897 DOI: 10.1111/pin.13094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/05/2021] [Indexed: 11/30/2022]
Abstract
Oral squamous cell carcinoma (OSCC) impairs functionality and sensuousness resulting in poor quality of life. Biomarkers can predict disease trajectory and lead to effective treatments. Transcriptomics have identified genes that are upregulated in tumor endothelial cells (TECs) compared with normal endothelial cells (NECs). Among them, chemokine receptor 7 (CXCR7) is highly expressed in TECs of several cancers and involved in angiogenesis of TECs. However, levels of CXCR7 in OSCC blood vessels have not been fully investigated. In this study, we analyzed the correlation between CXCR7 expression in TECs and clinicopathological factors in OSCC. Immunohistochemistry for CXCR7 and CD34 was performed on 59 OSCC tissue specimens resected between 1996 and 2008 at Hokkaido University Hospital. CXCR7 expression in blood vessels was evaluated by the ratio of CXCR7+/CD34+ blood vessels. CXCR7 expression was 42% and 19% in tumor and non-tumor parts, respectively, suggesting that CXCR7 expression is higher in TECs than in NECs. CXCR7 expression in TECs correlated with advanced T-stage and cancer stage. Overall survival and disease-free survival rates were higher in low-expressing CXCR7 patients than in high-expressing. These results suggest that CXCR7 expression in blood vessels may be a useful diagnostic and prognostic marker for OSCC patients.
Collapse
Affiliation(s)
- Misa Yanagiya
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Hokkaido, Japan.,Department of Oral Diagnosis and Medicine, Hokkaido University Graduate School of Dental Medicine, Hokkaido, Japan
| | - Randa I H Dawood
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Hokkaido, Japan
| | - Nako Maishi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Hokkaido, Japan.,Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Hokkaido, Japan
| | - Yasuhiro Hida
- Department of Cardiovascular Thoracic Surgery, Hokkaido University Faculty of Medicine, Hokkaido, Japan
| | - Chisaho Torii
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Hokkaido, Japan
| | - Dorcas A Annan
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Hokkaido, Japan.,Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Hokkaido, Japan
| | - Hiroshi Kikuchi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Hokkaido, Japan
| | - Aya Yanagawa Matsuda
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Hokkaido, Japan.,Department of Oral Pathology and Biology, Hokkaido University Graduate School of Dental Medicine, Hokkaido, Japan
| | - Tetsuya Kitamura
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Hokkaido, Japan.,Department of Oral Pathology and Biology, Hokkaido University Graduate School of Dental Medicine, Hokkaido, Japan
| | - Yoichi Ohiro
- Department of Oral and Maxillofacial Surgery, Hokkaido University Graduate School of Dental Medicine, Hokkaido, Japan
| | - Masanobu Shindoh
- Department of Oral Pathology and Biology, Hokkaido University Graduate School of Dental Medicine, Hokkaido, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Hokkaido University Faculty of Medicine, Hokkaido, Japan
| | - Yoshimasa Kitagawa
- Department of Oral Diagnosis and Medicine, Hokkaido University Graduate School of Dental Medicine, Hokkaido, Japan
| | - Kyoko Hida
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Hokkaido, Japan.,Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Hokkaido, Japan
| |
Collapse
|
10
|
Zhang J, Xiao C, Feng Z, Gong Y, Sun B, Li Z, Lu Y, Fei X, Wu W, Sun X, Teng L. SOX4 promotes the growth and metastasis of breast cancer. Cancer Cell Int 2020; 20:468. [PMID: 33005101 PMCID: PMC7523060 DOI: 10.1186/s12935-020-01568-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/23/2020] [Indexed: 12/18/2022] Open
Abstract
Purpose Increasing evidence has shown that the transcription factor SOX4 is closely associated with the development and progression of many malignant tumors. However, the effect of SOX4 on breast cancer is unclear. In this study, we purposed to investigate the role of SOX4 in the growth and metastasis in breast cancer and the underlying mechanism. Moreover, the effect of SOX4 on cancer cell resistance to chemotherapeutic agents was also evaluated in vitro and in vivo. Methods We used lentivirus technique to ectopically express SOX4 in MDA-MB-231 and SUM149 cells or knockdown SOX4 in BT474 cells, and examined the effect of these changes on various cellular functions. MTT assay was used to determine the cell viability as well as resistance to chemotherapeutic agents. The regulation of SOX4 on epithelial-mesenchymal transition (EMT)-related genes was analyzed using qRT-PCR. The binding of SOX4 to the CXCR7 gene was demonstrated using chromatin immunoprecipitation assay and dual-luciferase reporter activity assay. The effect of SOX4/CXCR7 axis on metastasis was examined using Transwell migration and Matrigel invasion assays. The expression of SOX4/CXCR7 in primary tumors and metastatic foci in lymph nodes was assessed using immunohistochemistry. Cellular morphology was investigated under phase contrast microscope and transmission electron microscopy. Moreover, the effect of SOX4 on tumor growth, metastasis, and resistance to chemotherapy was also studied in vivo by using bioluminescent imaging. Results SOX4 increased breast cancer cell viability, migration, and invasion in vitro and enhanced tumor growth and metastasis in vivo. It regulated EMT-related genes and bound to CXCR7 promoter to upregulate CXCR7 transcription. Both SOX4 and CXCR7 were highly expressed in human primary tumors and metastatic foci in lymph nodes. Treatment of breast cancer cells with the CXCR7 inhibitor CCX771 reversed the SOX4 effect on cell migration and invasion. Ectopic expression of SOX4 increased the susceptibility of cells to paclitaxel. Conclusions SOX4 plays an important role in the growth and metastasis of breast cancer. SOX4/CXCR7 may serve as potential therapeutic targets for the treatment. Paclitaxel may be a good therapeutic option if the expression level of SOX4 is high.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003 People's Republic of China.,Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030 USA
| | - Chunhua Xiao
- Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030 USA.,First Department of Breast Cancer, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, 1 Huan-Hu Xi Road, Ti-Yuan Bei, He Xi, Tianjin, 300060 People's Republic of China
| | - Zhenbo Feng
- Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030 USA.,Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi 530021 People's Republic of China
| | - Yun Gong
- Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030 USA
| | - Baohua Sun
- Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030 USA
| | - Zhongqi Li
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003 People's Republic of China
| | - Yimin Lu
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003 People's Republic of China
| | - Xiaojie Fei
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003 People's Republic of China
| | - Weizhu Wu
- Department of Breast and Thyroid Surgery, Ningbo Medical Center Lihuili Eastern Hospital, Ningbo, Zhejiang 315000 People's Republic of China
| | - Xiaoping Sun
- Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030 USA
| | - Lisong Teng
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003 People's Republic of China
| |
Collapse
|
11
|
Ciesielski O, Biesiekierska M, Panthu B, Vialichka V, Pirola L, Balcerczyk A. The Epigenetic Profile of Tumor Endothelial Cells. Effects of Combined Therapy with Antiangiogenic and Epigenetic Drugs on Cancer Progression. Int J Mol Sci 2020; 21:ijms21072606. [PMID: 32283668 PMCID: PMC7177242 DOI: 10.3390/ijms21072606] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/04/2020] [Accepted: 04/06/2020] [Indexed: 02/07/2023] Open
Abstract
Tumors require a constant supply of nutrients to grow which are provided through tumor blood vessels. To metastasize, tumors need a route to enter circulation, that route is also provided by tumor blood vessels. Thus, angiogenesis is necessary for both tumor progression and metastasis. Angiogenesis is tightly regulated by a balance of angiogenic and antiangiogenic factors. Angiogenic factors of the vascular endothelial growth factor (VEGF) family lead to the activation of endothelial cells, proliferation, and neovascularization. Significant VEGF-A upregulation is commonly observed in cancer cells, also due to hypoxic conditions, and activates endothelial cells (ECs) by paracrine signaling stimulating cell migration and proliferation, resulting in tumor-dependent angiogenesis. Conversely, antiangiogenic factors inhibit angiogenesis by suppressing ECs activation. One of the best-known anti-angiogenic factors is thrombospondin-1 (TSP-1). In pathological angiogenesis, the balance shifts towards the proangiogenic factors and an angiogenic switch that promotes tumor angiogenesis. Here, we review the current literature supporting the notion of the existence of two different endothelial lineages: normal endothelial cells (NECs), representing the physiological form of vascular endothelium, and tumor endothelial cells (TECs), which are strongly promoted by the tumor microenvironment and are biologically different from NECs. The angiogenic switch would be also important for the explanation of the differences between NECs and TECs, as angiogenic factors, cytokines and growth factors secreted into the tumor microenvironment may cause genetic instability. In this review, we focus on the epigenetic differences between the two endothelial lineages, which provide a possible window for pharmacological targeting of TECs.
Collapse
Affiliation(s)
- Oskar Ciesielski
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (O.C.); (M.B.); (V.V.)
- The Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland
| | - Marta Biesiekierska
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (O.C.); (M.B.); (V.V.)
| | - Baptiste Panthu
- INSERM Unit 1060, CarMeN Laboratory, Lyon 1 University, 165 Chemin du Grand Revoyet—BP12, F-69495 Pierre Bénite CEDEX, France; (B.P.); (L.P.)
| | - Varvara Vialichka
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (O.C.); (M.B.); (V.V.)
| | - Luciano Pirola
- INSERM Unit 1060, CarMeN Laboratory, Lyon 1 University, 165 Chemin du Grand Revoyet—BP12, F-69495 Pierre Bénite CEDEX, France; (B.P.); (L.P.)
| | - Aneta Balcerczyk
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (O.C.); (M.B.); (V.V.)
- Correspondence: ; Tel.: +48-42-635-45-10
| |
Collapse
|
12
|
Sjöberg E, Meyrath M, Chevigné A, Östman A, Augsten M, Szpakowska M. The diverse and complex roles of atypical chemokine receptors in cancer: From molecular biology to clinical relevance and therapy. Adv Cancer Res 2020; 145:99-138. [PMID: 32089166 DOI: 10.1016/bs.acr.2019.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Chemokines regulate directed cell migration, proliferation and survival and are key components in cancer biology. They exert their functions by interacting with seven-transmembrane domain receptors that signal through G proteins (GPCRs). A subgroup of four chemokine receptors known as the atypical chemokine receptors (ACKRs) has emerged as essential regulators of the chemokine functions. ACKRs play diverse and complex roles in tumor biology from tumor initiation to metastasis, including cancer cell proliferation, adherence to endothelium, epithelial-mesenchymal transition (EMT), extravasation from blood vessels, tumor-associated angiogenesis or protection from immunological responses. This chapter gives an overview on the established and emerging roles that the atypical chemokine receptors ACKR1, ACKR2, ACKR3 and ACKR4 play in the different phases of cancer development and dissemination, their clinical relevance, as well as on the hurdles to overcome in ACKRs targeting as cancer therapy.
Collapse
Affiliation(s)
- Elin Sjöberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Max Meyrath
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Andy Chevigné
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Arne Östman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | - Martyna Szpakowska
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
13
|
Quinn KE, Mackie DI, Caron KM. Emerging roles of atypical chemokine receptor 3 (ACKR3) in normal development and physiology. Cytokine 2019; 109:17-23. [PMID: 29903572 DOI: 10.1016/j.cyto.2018.02.024] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 02/21/2018] [Accepted: 02/21/2018] [Indexed: 01/16/2023]
Abstract
The discovery that atypical chemokine receptors (ACKRs) can initiate alternative signaling pathways rather than classical G-protein coupled receptor (GPCR) signaling has changed the paradigm of chemokine receptors and their roles in modulating chemotactic responses. The ACKR family has grown over the years, with discovery of new functions and roles in a variety of pathophysiological conditions. However, the extent to which these receptors regulate normal physiology is still continuously expanding. In particular, atypical chemokine receptor 3 (ACKR3) has proven to be an important receptor in mediating normal biological functions, including cardiac development and migration of cortical neurons. In this review, we illustrate the versatile and intriguing role of ACKR3 in physiology.
Collapse
Affiliation(s)
- K E Quinn
- Department of Cell Biology and Physiology, 111 MasonFarm Rd., 6312B MBRB CB# 7545, The University of North Carolina, Chapel Hill, NC 27599-7545, USA
| | - D I Mackie
- Department of Cell Biology and Physiology, 111 MasonFarm Rd., 6312B MBRB CB# 7545, The University of North Carolina, Chapel Hill, NC 27599-7545, USA
| | - K M Caron
- Department of Cell Biology and Physiology, 111 MasonFarm Rd., 6312B MBRB CB# 7545, The University of North Carolina, Chapel Hill, NC 27599-7545, USA.
| |
Collapse
|
14
|
Maishi N, Kikuchi H, Sato M, Nagao-Kitamoto H, Annan DA, Baba S, Hojo T, Yanagiya M, Ohba Y, Ishii G, Masutomi K, Shinohara N, Hida Y, Hida K. Development of Immortalized Human Tumor Endothelial Cells from Renal Cancer. Int J Mol Sci 2019; 20:ijms20184595. [PMID: 31533313 PMCID: PMC6770423 DOI: 10.3390/ijms20184595] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/13/2019] [Accepted: 09/13/2019] [Indexed: 12/20/2022] Open
Abstract
Tumor angiogenesis research and antiangiogenic drug development make use of cultured endothelial cells (ECs) including the human microvascular ECs among others. However, it has been reported that tumor ECs (TECs) are different from normal ECs (NECs). To functionally validate antiangiogenic drugs, cultured TECs are indispensable tools, but are not commercially available. Primary human TECs are available only in small quantities from surgical specimens and have a short life span in vitro due to their cellular senescence. We established immortalized human TECs (h-imTECs) and their normal counterparts (h-imNECs) by infection with lentivirus producing simian virus 40 large T antigen and human telomerase reverse transcriptase to overcome the replication barriers. These ECs exhibited an extended life span and retained their characteristic endothelial morphology, expression of endothelial marker, and ability of tube formation. Furthermore, h-imTECs showed their specific characteristics as TECs, such as increased proliferation and upregulation of TEC markers. Treatment with bevacizumab, an antiangiogenic drug, dramatically decreased h-imTEC survival, whereas the same treatment failed to alter immortalized NEC survival. Hence, these h-imTECs could be a valuable tool for drug screening to develop novel therapeutic agents specific to TECs or functional biological assays in tumor angiogenesis research.
Collapse
Affiliation(s)
- Nako Maishi
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan.
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan.
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan.
| | - Hiroshi Kikuchi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan.
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan.
| | - Masumi Sato
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan.
| | - Hiroko Nagao-Kitamoto
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan.
| | - Dorcas A Annan
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan.
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan.
| | - Shogo Baba
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan.
| | - Takayuki Hojo
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan.
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan.
- Department of Dental Anesthesiology, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan.
| | - Misa Yanagiya
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan.
- Department of Oral Diagnosis and Medicine, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan.
| | - Yusuke Ohba
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan.
| | - Genichiro Ishii
- Division of Pathology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan.
| | - Kenkichi Masutomi
- Division of Cancer Stem Cell, National Cancer Center Research Institute, Tokyo 104-0045, Japan.
| | - Nobuo Shinohara
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan.
| | - Yasuhiro Hida
- Department of Cardiovascular and Thoracic Surgery, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan.
| | - Kyoko Hida
- Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan.
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan.
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan.
| |
Collapse
|
15
|
Koenen J, Bachelerie F, Balabanian K, Schlecht-Louf G, Gallego C. Atypical Chemokine Receptor 3 (ACKR3): A Comprehensive Overview of its Expression and Potential Roles in the Immune System. Mol Pharmacol 2019; 96:809-818. [PMID: 31040166 DOI: 10.1124/mol.118.115329] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 04/24/2019] [Indexed: 12/24/2022] Open
Abstract
Atypical chemokine receptor 3 (ACKR3), previously known as C-X-C chemokine receptor type 7 (CXCR7), has emerged as a key player in several biologic processes, particularly during development. Its CXCL11 and CXCL12 scavenging activity and atypical signaling properties, together with a new array of other nonchemokine ligands, have established ACKR3 as a main regulator of physiologic processes at steady state and during inflammation. Here, we present a comprehensive review of ACKR3 expression in mammalian tissues in search of a possible connection with the receptor function. Besides the reported roles of ACKR3 during development, we discuss the potential contribution of ACKR3 to the function of the immune system, focusing on the myeloid lineage.
Collapse
Affiliation(s)
- Joyce Koenen
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Université Paris-Sud and Université Paris-Saclay, Clamart, France (J.K., F.B., K.B., G.S.-L., C.G.) and Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (J.K.)
| | - Françoise Bachelerie
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Université Paris-Sud and Université Paris-Saclay, Clamart, France (J.K., F.B., K.B., G.S.-L., C.G.) and Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (J.K.)
| | - Karl Balabanian
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Université Paris-Sud and Université Paris-Saclay, Clamart, France (J.K., F.B., K.B., G.S.-L., C.G.) and Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (J.K.)
| | - Géraldine Schlecht-Louf
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Université Paris-Sud and Université Paris-Saclay, Clamart, France (J.K., F.B., K.B., G.S.-L., C.G.) and Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (J.K.)
| | - Carmen Gallego
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Université Paris-Sud and Université Paris-Saclay, Clamart, France (J.K., F.B., K.B., G.S.-L., C.G.) and Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (J.K.)
| |
Collapse
|
16
|
Mollica Poeta V, Massara M, Capucetti A, Bonecchi R. Chemokines and Chemokine Receptors: New Targets for Cancer Immunotherapy. Front Immunol 2019; 10:379. [PMID: 30894861 PMCID: PMC6414456 DOI: 10.3389/fimmu.2019.00379] [Citation(s) in RCA: 371] [Impact Index Per Article: 74.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 02/14/2019] [Indexed: 12/21/2022] Open
Abstract
Immunotherapy is a clinically validated treatment for many cancers to boost the immune system against tumor growth and dissemination. Several strategies are used to harness immune cells: monoclonal antibodies against tumor antigens, immune checkpoint inhibitors, vaccination, adoptive cell therapies (e.g., CAR-T cells) and cytokine administration. In the last decades, it is emerging that the chemokine system represents a potential target for immunotherapy. Chemokines, a large family of cytokines with chemotactic activity, and their cognate receptors are expressed by both cancer and stromal cells. Their altered expression in malignancies dictates leukocyte recruitment and activation, angiogenesis, cancer cell proliferation, and metastasis in all the stages of the disease. Here, we review first attempts to inhibit the chemokine system in cancer as a monotherapy or in combination with canonical or immuno-mediated therapies. We also provide recent findings about the role in cancer of atypical chemokine receptors that could become future targets for immunotherapy.
Collapse
Affiliation(s)
- Valeria Mollica Poeta
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Matteo Massara
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Italy
| | - Arianna Capucetti
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Raffaella Bonecchi
- Humanitas Clinical and Research Center, IRCCS, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| |
Collapse
|
17
|
Borroni EM, Savino B, Bonecchi R, Locati M. Chemokines sound the alarmin: The role of atypical chemokine in inflammation and cancer. Semin Immunol 2018; 38:63-71. [DOI: 10.1016/j.smim.2018.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/08/2018] [Indexed: 12/17/2022]
|
18
|
Wu Y, Tian L, Xu Y, Zhang M, Xiang S, Zhao J, Wang Z. CXCR7 silencing inhibits the migration and invasion of human tumor endothelial cells derived from hepatocellular carcinoma by suppressing STAT3. Mol Med Rep 2018; 18:1644-1650. [PMID: 29901083 DOI: 10.3892/mmr.2018.9114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 05/11/2018] [Indexed: 12/28/2022] Open
Abstract
C-X-C chemokine receptor type 7 (CXCR7) is reported to be overexpressed in tumor endothelial cells (TECs), which are the primary target cells of antivascular chemotherapy. However, the role of CXCR7 in TECs is not fully understood. In the present study, CXCR7 expression was inhibited in TECs derived from hepatocellular carcinoma (HCC) using short hairpin (sh)RNA plasmids to investigate the role of CXCR7 in the regulation of migration and invasion of TECs as well as its underlying mechanisms. The data showed that the downregulation of CXCR7 significantly inhibited the migration and invasion of TECs. Further study showed that silencing CXCR7 resulted in decreased phosphorylated signal transducer and activator of transcription 3 (STAT3) at Tyr705 and its downstream target genes in TECs, including matrix metalloproteinase‑2 (MMP2) and vascular endothelial growth factor (VEGF). However, restoring STAT3 phosphorylation abolished the CXCR7‑shRNA‑induced decrease in TECs migration and invasion, as well as the downregulation of MMP2 and VEGF in TECs. These findings indicate that CXCR7 may regulate the migration and invasion of TECs derived from HCC via the STAT3 signaling pathway and that CXCR7 could be a potential target for the antivascular therapy of HCC.
Collapse
Affiliation(s)
- Ye Wu
- Department of General Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010059, P.R. China
| | - Lei Tian
- Department of Anesthesia, General Hospital of The PLA Rocket Force, Beijing 100088, P.R. China
| | - Yongle Xu
- Department of Vascular Surgery, General Hospital of PLA, Beijing 100853, P.R. China
| | - Minhong Zhang
- Department of Vascular Surgery, General Hospital of PLA, Beijing 100853, P.R. China
| | - Shengqing Xiang
- Department of Pharmacy, General Hospital of The PLA Rocket Force, Beijing 100088, P.R. China
| | - Jianguo Zhao
- Department of General Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010059, P.R. China
| | - Zhenxia Wang
- Department of General Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010059, P.R. China
| |
Collapse
|
19
|
Hida K, Maishi N, Annan DA, Hida Y. Contribution of Tumor Endothelial Cells in Cancer Progression. Int J Mol Sci 2018; 19:ijms19051272. [PMID: 29695087 PMCID: PMC5983794 DOI: 10.3390/ijms19051272] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/19/2018] [Accepted: 04/19/2018] [Indexed: 12/13/2022] Open
Abstract
Tumor progression depends on the process of angiogenesis, which is the formation of new blood vessels. These newly formed blood vessels supply oxygen and nutrients to the tumor, supporting its progression and providing a gateway for tumor metastasis. Tumor angiogenesis is regulated by the balance between angiogenic activators and inhibitors within the tumor microenvironment. Because the newly formed tumor blood vessels originate from preexisting normal vessels, tumor blood vessels, and tumor endothelial cells (TECs) have historically been considered to be the same as normal blood vessels and endothelial cells; however, evidence of TECs’ distinctive abnormal phenotypes has increased. In addition, it has been revealed that TECs constitute a heterogeneous population. Thus, TECs that line tumor blood vessels are important targets in cancer therapy. We have previously reported that TECs induce cancer metastasis. In this review, we describe recent studies on TEC abnormalities related to cancer progression to provide insight into new anticancer therapies.
Collapse
Affiliation(s)
- Kyoko Hida
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan.
| | - Nako Maishi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan.
| | - Dorcas A Annan
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan.
| | - Yasuhiro Hida
- Department of Cardiovascular and Thoracic Surgery, Hokkaido University Graduate School of Medicine, Sapporo 060-0815, Japan.
| |
Collapse
|
20
|
Hojo T, Maishi N, Towfik AM, Akiyama K, Ohga N, Shindoh M, Hida Y, Minowa K, Fujisawa T, Hida K. ROS enhance angiogenic properties via regulation of NRF2 in tumor endothelial cells. Oncotarget 2018; 8:45484-45495. [PMID: 28525375 PMCID: PMC5542202 DOI: 10.18632/oncotarget.17567] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 04/17/2017] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are unstable molecules that activate oxidative stress. Because of the insufficient blood flow in tumors, the tumor microenvironment is often exposed to hypoxic condition and nutrient deprivation, which induces ROS accumulation. We isolated tumor endothelial cells (TECs) and found that they have various abnormalities, although the underlying mechanisms are not fully understood. Here we showed that ROS were accumulated in tumor blood vessels and ROS enhanced TEC migration with upregulation of several angiogenesis related gene expressions. It was also demonstrated that these genes were upregulated by regulation of Nuclear factor erythroid 2-related factor 2 (NRF2). Among these genes, we focused on Biglycan, a small leucine-rich proteoglycan. Inhibition of Toll-like receptors 2 and 4, known BIGLYCAN (BGN) receptors, cancelled the TEC motility stimulated by ROS. ROS inhibited NRF2 expression in TECs but not in NECs, and NRF2 inhibited phosphorylation of SMAD2/3, which activates transcription of BGN. These results indicated that ROS-induced BGN caused the pro-angiogenic phenotype in TECs via NRF2 dysregulation.
Collapse
Affiliation(s)
- Takayuki Hojo
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan.,Department of Dental Anesthesiology, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan
| | - Nako Maishi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
| | - Alam Mohammad Towfik
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan.,Department of Dental Radiology, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan
| | - Kosuke Akiyama
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
| | - Noritaka Ohga
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan.,Department of Oral Diagnosis and Medicine, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan
| | - Masanobu Shindoh
- Department of Oral Pathology and Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan
| | - Yasuhiro Hida
- Department of Cardiovascular and Thoracic Surgery, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Kazuyuki Minowa
- Department of Dental Radiology, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan
| | - Toshiaki Fujisawa
- Department of Dental Anesthesiology, Hokkaido University Graduate School of Dental Medicine, Sapporo 060-8586, Japan
| | - Kyoko Hida
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
| |
Collapse
|
21
|
Pluchino N, Mamillapalli R, Moridi I, Tal R, Taylor HS. G-Protein-Coupled Receptor CXCR7 Is Overexpressed in Human and Murine Endometriosis. Reprod Sci 2018; 25:1168-1174. [PMID: 29587613 DOI: 10.1177/1933719118766256] [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] [Indexed: 12/21/2022]
Abstract
Endometriosis is a chronic inflammatory disease. Dysfunctional regulation of chemokines and chemokine receptors is a crucial aspect of endometriosis pathogenesis. Chemokine G-protein-coupled receptors (GPCRs) are important drug targets that regulate inflammation and immunity. Recently, CXCR7, a C-X-C motif containing GPCR, has been identified as a receptor for chemokine ligand CXCL12, one of the best characterized chemokines for cell trafficking, angiogenesis, and cell proliferation in cancer and inflammation. Here, we investigated the expression and localization of CXCR7 in human endometriosis and a murine model of the disease. Normal endometrial epithelium and stroma showed undetectable or very low expression of CXCR7, without any significant changes across phases of the menstrual cycle in humans. CXCR7 is significantly upregulated in endometriosis, showing higher staining in glands and in associated vessels. The mouse model recapitulated the human findings. In conclusion, overexpression of CXCR7 in different cellular populations of endometriosis microenvironment may play a role in the pathogenesis and represent a novel target for treatment.
Collapse
Affiliation(s)
- Nicola Pluchino
- 1 Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Ramanaiah Mamillapalli
- 1 Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Irene Moridi
- 1 Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Reshef Tal
- 1 Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Hugh S Taylor
- 1 Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
22
|
Abstract
An apparently balanced t(2;3)(q37.3;q13.2) translocation that appears to segregate with renal cell carcinoma (RCC) has indicated potential areas to search for the elusive genetic basis of clear cell RCC. We applied Hi-Plex targeted sequencing to analyse germline DNA from 479 individuals affected with clear cell RCC for this breakpoint translocation and genetic variants in neighbouring genes on chromosome 2, ACKR3 and COPS8. While only synonymous variants were found in COPS8, one of the missense variants in ACKR3:c.892C>T, observed in 4/479 individuals screened (0.8%), was predicted likely to damage ACKR3 function. Identification of causal genes for RCC has potential clinical utility, where risk assessment and risk management can offer better outcomes, with surveillance for at-risk relatives and nephron sparing surgery through earlier intervention.
Collapse
|
23
|
Spinosa PC, Luker KE, Luker GD, Linderman JJ. The CXCL12/CXCR7 signaling axis, isoforms, circadian rhythms, and tumor cellular composition dictate gradients in tissue. PLoS One 2017; 12:e0187357. [PMID: 29117251 PMCID: PMC5678865 DOI: 10.1371/journal.pone.0187357] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 10/18/2017] [Indexed: 12/24/2022] Open
Abstract
Chemokine CXCL12 gradients drive chemotaxis in a CXCR4-dependent mechanism and have been implicated in cancer metastasis. While CXCL12 gradients are typically studied in organized, defined environments, the tumor microenvironment is disorganized. In vivo, CXCL12 gradients depend on many factors: the number and arrangement of cells secreting and degrading CXCL12, isoform-dependent binding to the extracellular matrix, diffusion, and circadian fluctuations. We developed a computational model of the tumor microenvironment to simulate CXCL12 gradient dynamics in disorganized tissue. There are four major findings from the model. First, CXCL12-β and -γ form higher magnitude (steeper) gradients compared to CXCL12-α. Second, endothelial CXCR7+ cells regulate CXCL12 gradient direction by controlling concentrations near but not far from the vasculature. Third, the magnitude and direction of CXCL12 gradients are dependent on the local composition of secreting and scavenging cells within the tumor. We theorize that "micro-regions" of cellular heterogeneity within the tumor are responsible for forming strong gradients directed into the blood. Fourth, CXCL12 circadian fluctuations influence gradient magnitude but not direction. Our simulations provide predictions for future experiments in animal models. Understanding the generation of CXCL12 gradients is crucial to inhibiting cancer metastasis.
Collapse
Affiliation(s)
- Phillip C. Spinosa
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kathryn E. Luker
- Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Gary D. Luker
- Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jennifer J. Linderman
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
24
|
Reyes N, Benedetti I, Rebollo J, Correa O, Geliebter J. Atypical chemokine receptor CCRL2 is overexpressed in prostate cancer cells. J Biomed Res 2017; 33:17. [PMID: 29497024 PMCID: PMC6352880 DOI: 10.7555/jbr.32.20170057] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 12/05/2017] [Indexed: 12/28/2022] Open
Abstract
Atypical chemokine receptors have recently emerged as important molecular players in health and diseases; they affect chemokine availability and function and impact a multitude of pathophysiological events, including the tumorigenesis process. This family of atypical receptors comprises five members: ACKR1/DARC, ACKR2/D6, ACKR3/CXCR7, ACKR4/CCRL1, and ACKR5/CCRL2. This work evaluated the differential expression of these receptors in prostate cancer using quantitative PCR. Further evaluation of CCRL2 at the protein level confirmed its overexpression in a metastatic cell line and in malignant prostatic tissues from patients. CCRL2, a presumed member of the atypical chemokine receptor family, plays a key role in lung dendritic cell trafficking to peripheral lymph nodes. Recent studies have reported the expression of CCRL2 in different human cancer cell lines and tissues. However, its function and expression in prostate cancer has not been previously addressed.
Collapse
Affiliation(s)
- Niradiz Reyes
- . Department of Basic Sciences, School of Medicine, University of Cartagena, Cartagena, Bolívar, Colombia
- . Research Group of Genetics and Molecular Biology
| | - Ines Benedetti
- . Department of Basic Sciences, School of Medicine, University of Cartagena, Cartagena, Bolívar, Colombia
- . Research Group of Histopathology
| | - Juan Rebollo
- . Department of Basic Sciences, School of Medicine, University of Cartagena, Cartagena, Bolívar, Colombia
- . Research Group of Genetics and Molecular Biology
| | - Oscar Correa
- . Research Group of Genetics and Molecular Biology
| | - Jan Geliebter
- . School of Medicine, Department of Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA
| |
Collapse
|
25
|
Hida K, Maishi N, Akiyama K, Ohmura-Kakutani H, Torii C, Ohga N, Osawa T, Kikuchi H, Morimoto H, Morimoto M, Shindoh M, Shinohara N, Hida Y. Tumor endothelial cells with high aldehyde dehydrogenase activity show drug resistance. Cancer Sci 2017; 108:2195-2203. [PMID: 28851003 PMCID: PMC5666026 DOI: 10.1111/cas.13388] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/11/2017] [Accepted: 08/16/2017] [Indexed: 01/11/2023] Open
Abstract
Tumor blood vessels play an important role in tumor progression and metastasis. We previously reported that tumor endothelial cells (TEC) exhibit several altered phenotypes compared with normal endothelial cells (NEC). For example, TEC have chromosomal abnormalities and are resistant to several anticancer drugs. Furthermore, TEC contain stem cell‐like populations with high aldehyde dehydrogenase (ALDH) activity (ALDHhighTEC). ALDHhighTEC have proangiogenic properties compared with ALDHlowTEC. However, the association between ALDHhighTEC and drug resistance remains unclear. In the present study, we found that ALDH mRNA expression and activity were higher in both human and mouse TEC than in NEC. Human NEC:human microvascular endothelial cells (HMVEC) were treated with tumor‐conditioned medium (tumor CM). The ALDHhigh population increased along with upregulation of stem‐related genes such as multidrug resistance 1, CD90, ALP, and Oct‐4. Tumor CM also induced sphere‐forming ability in HMVEC. Platelet‐derived growth factor (PDGF)‐A in tumor CM was shown to induce ALDH expression in HMVEC. Finally, ALDHhighTEC were resistant to fluorouracil (5‐FU) in vitro and in vivo. ALDHhighTEC showed a higher grade of aneuploidy compared with that in ALDHlowTEC. These results suggested that tumor‐secreting factor increases ALDHhighTEC populations that are resistant to 5‐FU. Therefore, ALDHhighTEC in tumor blood vessels might be an important target to overcome or prevent drug resistance.
Collapse
Affiliation(s)
- Kyoko Hida
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Vascular Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Nako Maishi
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Vascular Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Kosuke Akiyama
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Vascular Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Hitomi Ohmura-Kakutani
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Chisaho Torii
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Vascular Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Noritaka Ohga
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Takahiro Osawa
- Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroshi Kikuchi
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hirofumi Morimoto
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Department of Gastroenterological Surgery II, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masahiro Morimoto
- Vascular Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Masanobu Shindoh
- Department of Oral Pathology and Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Nobuo Shinohara
- Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yasuhiro Hida
- Department of Cardiovascular and Thoracic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
26
|
Maishi N, Hida K. Tumor endothelial cells accelerate tumor metastasis. Cancer Sci 2017; 108:1921-1926. [PMID: 28763139 PMCID: PMC5623747 DOI: 10.1111/cas.13336] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/18/2017] [Accepted: 07/26/2017] [Indexed: 12/13/2022] Open
Abstract
Tumor metastasis is the main cause of cancer-related death. Understanding the molecular mechanisms underlying tumor metastasis is crucial to control this fatal disease. Several molecular pathways orchestrate the complex biological cell events during a metastatic cascade. It is now well known that bidirectional interaction between tumor cells and their microenvironment, including tumor stroma, is important for tumor progression and metastasis. Tumor stromal cells, which acquire their specific characteristics in the tumor microenvironment, accelerate tumor malignancy. The formation of new blood vessels, termed as tumor angiogenesis, is a requirement for tumor progression. Tumor blood vessels supply nutrients and oxygen and also provide the route for metastasis. Tumor endothelial cells, which line tumor blood vessels, also exhibit several altered phenotypes compared with those of their normal counterparts. Recent studies have emphasized "angiocrine factors" that are released from tumor endothelial cells and promote tumor progression. During intravasation, tumor cells physically contact tumor endothelial cells and interact with them by juxtacrine and paracrine signaling. Recently, we observed that in highly metastatic tumors, tumor endothelial cells interact with tumor cells by secretion of a small leucine-rich repeat proteoglycan known as biglycan. Biglycan from tumor endothelial cells stimulates the tumor cells to metastasize. In the present review, we highlight the role of tumor stromal cells, particularly endothelial cells, in the initial steps of tumor metastasis.
Collapse
Affiliation(s)
- Nako Maishi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Kyoko Hida
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
27
|
Juárez-Hernández E, Motola-Kuba D, Chávez-Tapia NC, Uribe M, Barbero Becerra V. Biomarkers in hepatocellular carcinoma: an overview. Expert Rev Gastroenterol Hepatol 2017; 11:549-558. [PMID: 28347162 DOI: 10.1080/17474124.2017.1311785] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Current methods for HCC diagnosis have not an optimal diagnostic accuracy. The detection of more than one biomarker seems to improve their individual performance and provide an accurate HCC diagnosis approach. Individual gene expression seems to influence whether or not the treatment is successful, since several molecules have interfere with cancer associated pathways and have been related to poor prognosis which condition the lack of effective treatment options. Areas covered: Novel biomarkers have been proposed as a useful tool in each patient prognosis. This article aims to review the recent evidence based on HCC biomarkers which seems to have a regulative role according to tumor cell development leading to a specific biological response. Epigenetic regulation, miRNAs, and genome sequencing analysis propose molecular expression signatures as novel biomarkers which allowed achieve the major goal for the use of biomarkers in clinical practice. Moreover, a deeper analysis for determine the diagnostic accuracy of biomarkers has been made. Expert commentary: To improve of methodological designs and sample sizes are needed in order to support the role of biomarkers in HCC. Furthermore, is necessary to consider HCC etiologies and all clinic disease context to carried out clinical phase studies to thrust biomarkers application.
Collapse
Affiliation(s)
- Eva Juárez-Hernández
- a Translational Research Unit , Medica Sur Clinic & Foundation , Mexico City , Mexico
| | - Daniel Motola-Kuba
- b Oncology Center , Medica Sur Clinic & Foundation , Mexico City , Mexico
| | | | - Misael Uribe
- a Translational Research Unit , Medica Sur Clinic & Foundation , Mexico City , Mexico
| | | |
Collapse
|
28
|
Meng M, Liao H, Zhang B, Pan Y, Kong Y, Liu W, Yang P, Huo Z, Cao Z, Zhou Q. Cigarette smoke extracts induce overexpression of the proto-oncogenic gene interleukin-13 receptor α2 through activation of the PKA-CREB signaling pathway to trigger malignant transformation of lung vascular endothelial cells and angiogenesis. Cell Signal 2016; 31:15-25. [PMID: 27986643 DOI: 10.1016/j.cellsig.2016.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/03/2016] [Accepted: 12/12/2016] [Indexed: 01/23/2023]
Abstract
Cigarette smoking is a major cause of lung cancer. Tumor-associated endothelial cells (TAECs) play important roles in tumor angiogenesis and metastasis. However, whether cigarette smoking can trigger genesis of lung TAECs has not been reported yet. In the current study, we used lung endothelial cell (EC) lines as a model to study the pathological effect of cigarette smoke extracts (CSEs) on human lung ECs, and found that a lower dose of 4% CSEs obviously caused abnormal morphological changes in ECs, increased the permeability of endothelial monolayer, while a higher concentration of 8% CSEs caused EC apoptosis. Strikingly, CSEs induced a 117-fold overexpression of a pro-tumorigenic interleukin-13 receptor α2 gene (IL-13Rα2, also named as CT-19) through activation of the protein kinase A (PKA) and cAMP response element-binding protein (CREB) signaling pathway. A PKA specific inhibitor H89 completely abolished CSEs-induced IL-13Rα2 overexpression. The overexpression of IL-13Rα2 in lung ECs significantly increased the tumorigenic, migratory, and angiogenic capabilities of the cells, suggesting that IL-13Rα2 promotes genesis of lung TAECs. Together, our data show that CSEs activate the PKA, CREB, and IL-13Rα2 axis in lung ECs, and IL-13Rα2 promotes the malignant transformation of lung ECs and genesis of TAECs with robust angiogenic and oncogenic capabilities. Our study provides new insight into the mechanism of CSEs-triggered lung cancer angiogenesis and tumorigenesis, suggesting that the PKA-CREB-IL-13Rα2 axis is a potential target for novel anti-lung tumor angiogenesis and anti-lung cancer drug discovery.
Collapse
Affiliation(s)
- Mei Meng
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Huaidong Liao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Bin Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yanyan Pan
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Ying Kong
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Wenming Liu
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Ping Yang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zihe Huo
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhifei Cao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Quansheng Zhou
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
29
|
Hida K, Maishi N, Kawamoto T, Akiyama K, Ohga N, Hida Y, Yamada K, Hojo T, Kikuchi H, Sato M, Torii C, Shinohara N, Shindoh M. Tumor endothelial cells express high pentraxin 3 levels. Pathol Int 2016; 66:687-694. [DOI: 10.1111/pin.12474] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 09/13/2016] [Accepted: 10/07/2016] [Indexed: 01/22/2023]
Affiliation(s)
- Kyoko Hida
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine; Hokkaido University; Sapporo Japan
- Department of Vascular Biology; Hokkaido University Graduate School of Dental Medicine; Sapporo Japan
| | - Nako Maishi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine; Hokkaido University; Sapporo Japan
- Department of Vascular Biology; Hokkaido University Graduate School of Dental Medicine; Sapporo Japan
| | - Taisuke Kawamoto
- Department of Vascular Biology; Hokkaido University Graduate School of Dental Medicine; Sapporo Japan
| | - Kosuke Akiyama
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine; Hokkaido University; Sapporo Japan
- Department of Vascular Biology; Hokkaido University Graduate School of Dental Medicine; Sapporo Japan
| | - Noritaka Ohga
- Department of Vascular Biology; Hokkaido University Graduate School of Dental Medicine; Sapporo Japan
- Department of Oral Diagnosis and Medicine; Hokkaido University Graduate School of Dental Medicine; Sapporo Japan
| | - Yasuhiro Hida
- Department of Cardiovascular and Thoracic Surgery; Hokkaido University Graduate School of Medicine; Sapporo Japan
| | - Kenji Yamada
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine; Hokkaido University; Sapporo Japan
- Department of Gastroenterological Surgery I; Hokkaido University Graduate School of Medicine; Sapporo Japan
| | - Takayuki Hojo
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine; Hokkaido University; Sapporo Japan
- Department of Dental Anesthesiology; Hokkaido University Graduate School of Dental Medicine; Sapporo Japan
| | - Hiroshi Kikuchi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine; Hokkaido University; Sapporo Japan
- Department of Renal and Genitourinary Surgery; Hokkaido University Graduate School of Medicine; Sapporo Japan
| | - Masumi Sato
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine; Hokkaido University; Sapporo Japan
| | - Chisaho Torii
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine; Hokkaido University; Sapporo Japan
- Department of Oral and Maxillofacial Surgery, Graduate School of Dental Medicine; Hokkaido University; Sapporo Japan
| | - Nobuo Shinohara
- Department of Renal and Genitourinary Surgery; Hokkaido University Graduate School of Medicine; Sapporo Japan
| | - Masanobu Shindoh
- Department of Oral Pathology and Biology; Hokkaido University Graduate School of Dental Medicine; Sapporo Japan
| |
Collapse
|
30
|
Yuan ZX, Mo J, Zhao G, Shu G, Fu HL, Zhao W. Targeting Strategies for Renal Cell Carcinoma: From Renal Cancer Cells to Renal Cancer Stem Cells. Front Pharmacol 2016; 7:423. [PMID: 27891093 PMCID: PMC5103413 DOI: 10.3389/fphar.2016.00423] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/25/2016] [Indexed: 12/14/2022] Open
Abstract
Renal cell carcinoma (RCC) is a common form of urologic tumor that originates from the highly heterogeneous epithelium of renal tubules. Over the last decade, targeting therapies to renal cancer cells have transformed clinical care for RCC. Recently, it was proposed that renal cancer stem cells (CSCs) isolated from renal carcinomas were responsible for driving tumor growth and resistance to conventional chemotherapy and radiotherapy, according to the theory of CSCs; this has provided the rationale for therapies targeting this aggressive cell population. Precise identification of renal CSC populations and the complete cell hierarchy will accurately inform characterization of disease subtypes. This will ultimately contribute to more personalized and targeted therapies. Here, we summarize potential targeting strategies for renal cancer cells and renal CSCs, including tyrosine kinase inhibitors, mammalian target of rapamycin inhibitors (mTOR), interleukins, CSC marker inhibitors, bone morphogenetic protein-2, antibody drug conjugates, and nanomedicine. In conclusion, targeting therapies for RCC represent new directions for exploration and clinical investigation and they plant a seed of hope for advanced clinical care.
Collapse
Affiliation(s)
- Zhi-Xiang Yuan
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Jingxin Mo
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Guixian Zhao
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Gang Shu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Hua-Lin Fu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University Chengdu, China
| | - Wei Zhao
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| |
Collapse
|
31
|
McConnell AT, Ellis R, Pathy B, Plummer R, Lovat PE, O'Boyle G. The prognostic significance and impact of the CXCR4-CXCR7-CXCL12 axis in primary cutaneous melanoma. Br J Dermatol 2016; 175:1210-1220. [PMID: 27167239 DOI: 10.1111/bjd.14720] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2016] [Indexed: 01/10/2023]
Abstract
BACKGROUND Expression of the chemokine receptor CXCR4 is known to regulate melanoma metastasis to distant sites with high expression of the CXCL12 ligand. However, the prognostic impact of CXCR4 expression and potential for autocrine-mediated activation of prosurvival mitogen-activated protein kinase signalling remains enigmatic. Furthermore, expression of the decoy receptor CXCR7 within the local cutaneous melanoma microenvironment remains undefined. OBJECTIVES To define the contribution and prognostic impact of CXCR4-CXCR7-CXCL12 signalling in primary cutaneous melanomas and the immediate tumour microenvironment. METHODS Immunohistochemical/immunofluorescent expression of CXCR4, CXCR7 or CXC12 was analysed in human metastatic melanoma cell lines, primary cutaneous cell types and a retrospective cohort of primary melanomas/benign naevi. CXCL12 secretion by melanoma/cutaneous cells was evaluated by enzyme-linked immunosorbent assay, and autocrine CXCR4-CXCL12 signalling was investigated by addition of a CXCL12-neutralizing antibody. RESULTS CXCR4 expression was significantly higher in primary melanomas that subsequently metastasized after 7 years (P = 0·037). Stratification for American Joint Committee on Cancer (AJCC) stage II disease revealed significantly decreased disease-free survival in patients with > 50% CXCR4 expression (P = 0·036), while comparative analysis of CXCL12 expression in the adjacent epidermis of all AJCC stage melanomas revealed increased CXCL12 correlated with prolonged time to metastasis (P = 0·014). CXCR7 was expressed within the primary melanoma microenvironment but was absent on primary tumours. Addition of anti-CXCL12 to BRAF-mutant melanoma cells resulted in downregulation of phospho-CXCR4 and phospho-extracellular signal-related kinase, indicating autocrine CXCR4-CXCL12 signalling. CONCLUSIONS CXCR4 expression defines a potential prognostic biomarker for AJCC stage II melanoma. Moreover, targeting the CXCR4-CXCR7-CXCL12 axis may represent a novel therapeutic strategy to prevent early melanoma progression.
Collapse
Affiliation(s)
- A T McConnell
- Dermatological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - R Ellis
- Dermatological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K.,James Cook University Hospital, Middlesbrough, U.K
| | - B Pathy
- Dermatological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - R Plummer
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, U.K
| | - P E Lovat
- Dermatological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K
| | - G O'Boyle
- Dermatological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, U.K.,Faculty of Applied Sciences, University of Sunderland, Sunderland, U.K
| |
Collapse
|
32
|
Tumour endothelial cells in high metastatic tumours promote metastasis via epigenetic dysregulation of biglycan. Sci Rep 2016; 6:28039. [PMID: 27295191 PMCID: PMC4904795 DOI: 10.1038/srep28039] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/27/2016] [Indexed: 11/15/2022] Open
Abstract
Tumour blood vessels are gateways for distant metastasis. Recent studies have revealed that tumour endothelial cells (TECs) demonstrate distinct phenotypes from their normal counterparts. We have demonstrated that features of TECs are different depending on tumour malignancy, suggesting that TECs communicate with surrounding tumour cells. However, the contribution of TECs to metastasis has not been elucidated. Here, we show that TECs actively promote tumour metastasis through a bidirectional interaction between tumour cells and TECs. Co-implantation of TECs isolated from highly metastatic tumours accelerated lung metastases of low metastatic tumours. Biglycan, a small leucine-rich repeat proteoglycan secreted from TECs, activated tumour cell migration via nuclear factor-κB and extracellular signal–regulated kinase 1/2. Biglycan expression was upregulated by DNA demethylation in TECs. Collectively, our results demonstrate that TECs are altered in their microenvironment and, in turn, instigate tumour cells to metastasize, which is a novel mechanism for tumour metastasis.
Collapse
|
33
|
Hida K, Maishi N, Sakurai Y, Hida Y, Harashima H. Heterogeneity of tumor endothelial cells and drug delivery. Adv Drug Deliv Rev 2016; 99:140-147. [PMID: 26626622 DOI: 10.1016/j.addr.2015.11.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/21/2015] [Accepted: 11/10/2015] [Indexed: 10/22/2022]
Abstract
To date anti-angiogenic therapy has been used for cancer therapy widely, yielding promising results. However, it has been elucidated that current anti-angiogenic drug has several issues to be solved, such as side-effects and drug resistance. It has been reported that tumor endothelial cells (TECs) differ from normal counterparts. In addition, it was shown that the TECs are heterogeneous according to the malignancy status of tumor. The development of novel strategy for targeting tumor vasculature is required. Recently, we have developed an active targeting system, which targets TECs specifically. In this review, we will discuss how TECs in tumor vasculature are heterogeneous and offer new perspectives on a drug delivery system, which can target heterogeneous tumor blood vessels from a viewpoint of personalized medicine.
Collapse
|
34
|
Massara M, Bonavita O, Mantovani A, Locati M, Bonecchi R. Atypical chemokine receptors in cancer: friends or foes? J Leukoc Biol 2016; 99:927-33. [PMID: 26908826 DOI: 10.1189/jlb.3mr0915-431rr] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 02/02/2016] [Indexed: 01/23/2023] Open
Abstract
The chemokine system is a fundamental component of cancer-related inflammation involved in all stages of cancer development. It controls not only leukocyte infiltration in primary tumors but also angiogenesis, cancer cell proliferation, and migration to metastatic sites. Atypical chemokine receptors are a new, emerging class of regulators of the chemokine system. They control chemokine bioavailability by scavenging, transporting, or storing chemokines. They can also regulate the activity of canonical chemokine receptors with which they share the ligands by forming heterodimers or by modulating their expression levels or signaling activity. Here, we summarize recent results about the role of these receptors (atypical chemokine receptor 1/Duffy antigen receptor for chemokine, atypical chemokine receptor 2/D6, atypical chemokine receptor 3/CXC-chemokine receptor 7, and atypical chemokine receptor 4/CC-chemokine receptor-like 1) on the tumorigenesis process, indicating that their effects are strictly dependent on the cell type on which they are expressed and on their coexpression with other chemokine receptors. Indeed, atypical chemokine receptors inhibit tumor growth and progression through their activity as negative regulators of chemokine bioavailability, whereas, on the contrary, they can promote tumorigenesis when they regulate the signaling of other chemokine receptors, such as CXC-chemokine receptor 4. Thus, atypical chemokine receptors are key components of the regulatory network of inflammation and immunity in cancer and may have a major effect on anti-inflammatory and immunotherapeutic strategies.
Collapse
Affiliation(s)
- Matteo Massara
- Humanitas Clinical and Research Center, Rozzano, Italy; Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, Rozzano, Italy; and
| | - Ornella Bonavita
- Humanitas Clinical and Research Center, Rozzano, Italy; Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, Rozzano, Italy; and
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, Rozzano, Italy; Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| | - Massimo Locati
- Humanitas Clinical and Research Center, Rozzano, Italy; Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, Rozzano, Italy; and
| | - Raffaella Bonecchi
- Humanitas Clinical and Research Center, Rozzano, Italy; Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| |
Collapse
|
35
|
Zhang H, Yang L, Teng X, Liu Z, Liu C, Zhang L, Liu Z. The chemokine receptor CXCR7 is a critical regulator for the tumorigenesis and development of papillary thyroid carcinoma by inducing angiogenesis in vitro and in vivo. Tumour Biol 2015; 37:2415-23. [PMID: 26383519 DOI: 10.1007/s13277-015-4051-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/02/2015] [Indexed: 01/20/2023] Open
Abstract
Papillary thyroid carcinoma (PTC) is a well-differentiated neoplasm, but it can transfer early to cervical lymph nodes. Accumulating evidences have confirmed the important roles of CXCR7 in tumor cell proliferation, invasion, metastasis, and angiogenesis. Our previous study demonstrated CXCR7 modulated proliferation, apoptosis, and invasion of PTC cells. In this study, we evaluated the effect of expression of CXCR7 in PTC cells on angiogenesis and whether its expression had an influence on the tumor growth of PTC in vivo. We evaluated the effect of CXCR7 on interleukin-8 (IL-8) and vascular endothelial growth factor (VEGF) secretion, angiogenesis, and tumor growth by ELISA, endothelial tube formation assay, and a xenograft tumor model in nude mice. Immunohistochemistry was used to assess expression of CD34 in tumor of mice. In vitro and in vivo studies in PTC cells suggested that the alteration of CXCR7 expression was correlated with angiogenesis and tumor growth. Moreover, CXCR7 mediated the expression of IL-8 and VEGF, which might be involved in the regulation of tumor angiogenesis. These findings suggest that CXCR7 affects the growth of PTC cells and participates in the tumorigenesis of PTC, probably through regulating angiogenesis by the proangiogenic VEGF or IL-8.
Collapse
Affiliation(s)
- Hengwei Zhang
- Department of General Surgery, Shengjing Hospital, China Medical University, No. 36 Sanhao Street, Shenyang, 110004, China
| | - Lei Yang
- Department of General Surgery, First Affiliated Hospital, China Medical University, Shenyang, 110001, China
| | - Xuyong Teng
- Department of General Surgery, Shengjing Hospital, China Medical University, No. 36 Sanhao Street, Shenyang, 110004, China
| | - Zhangyi Liu
- Department of General Surgery, Shengjing Hospital, China Medical University, No. 36 Sanhao Street, Shenyang, 110004, China
| | - Chenxi Liu
- Department of General Surgery, Shengjing Hospital, China Medical University, No. 36 Sanhao Street, Shenyang, 110004, China
| | - Lei Zhang
- Department of General Surgery, Shengjing Hospital, China Medical University, No. 36 Sanhao Street, Shenyang, 110004, China
| | - Zhen Liu
- Department of General Surgery, Shengjing Hospital, China Medical University, No. 36 Sanhao Street, Shenyang, 110004, China.
| |
Collapse
|
36
|
Yamada K, Maishi N, Akiyama K, Towfik Alam M, Ohga N, Kawamoto T, Shindoh M, Takahashi N, Kamiyama T, Hida Y, Taketomi A, Hida K. CXCL12-CXCR7 axis is important for tumor endothelial cell angiogenic property. Int J Cancer 2015; 137:2825-36. [PMID: 26100110 DOI: 10.1002/ijc.29655] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 05/29/2015] [Accepted: 06/11/2015] [Indexed: 12/27/2022]
Abstract
We reported that tumor endothelial cells (TECs) differ from normal endothelial cells (NECs) in many aspects, such as gene expression profiles. Although CXCR7 is reportedly highly expressed in blood vessels of several tumors, its function in TECs is still unknown. To investigate this role, we isolated TECs from mouse tumor A375SM xenografts, and compared them with NECs from normal mouse dermis. After confirming CXCR7 upregulation in TECs, we analyzed its function using CXCR7 siRNA and CXCR7 inhibitor; CCX771. CXCR7 siRNA and CCX771 inhibited migration, tube formation and resistance to serum starvation in TECs but not in NECs. ERK1/2 phosphorylation was inhibited by CXCR7 knockdown in TECs. These results suggest that CXCR7 promotes angiogenesis in TECs via ERK1/2 phosphorylation. Using ELISA, we also detected CXCL12, a ligand of CXCR7, in conditioned medium from TECs, but not from NECs. CXCL12 neutralizing antibody significantly inhibited TEC random motility. VEGF stimulation upregulated CXCR7 expression in NECs, implying that VEGF mediates CXCR7 expression in endothelial cells. A CXCR7 inhibitor, CCX771 also inhibited tumor growth, lung metastasis and tumor angiogenesis in vivo. Taken together, the CXCL12-CXCR7 autocrine loop affects TEC proangiogenic properties, and could be the basis for an antiangiogenic therapy that specifically targets tumor blood vessels rather than normal vessels.
Collapse
Affiliation(s)
- Kenji Yamada
- Department of Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Nako Maishi
- Department of Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Kosuke Akiyama
- Department of Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Mohammad Towfik Alam
- Department of Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Noritaka Ohga
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Taisuke Kawamoto
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Masanobu Shindoh
- Oral Pathology and Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Norihiko Takahashi
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Toshiya Kamiyama
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yasuhiro Hida
- Department of Cardiovascular and Thoracic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kyoko Hida
- Department of Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| |
Collapse
|
37
|
Stacer AC, Fenner J, Cavnar SP, Xiao A, Zhao S, Chang SL, Salomonnson A, Luker KE, Luker GD. Endothelial CXCR7 regulates breast cancer metastasis. Oncogene 2015; 35:1716-24. [PMID: 26119946 PMCID: PMC4486335 DOI: 10.1038/onc.2015.236] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 04/29/2015] [Accepted: 05/01/2015] [Indexed: 02/08/2023]
Abstract
Atypical chemokine receptor CXCR7 (ACKR3) functions as a scavenger receptor for chemokine CXCL12, a molecule that promotes multiple steps in tumor growth and metastasis in breast cancer and multiple other malignancies. While normal vascular endothelium expresses low levels of CXCR7, marked upregulation of CXCR7 occurs in tumor vasculature in breast cancer and other tumors. To investigate effects of endothelial CXCR7 in breast cancer, we conditionally deleted this receptor from vascular endothelium of adult mice, generating CXCR7ΔEND/ΔEND animals. CXCR7ΔEND/ΔEND mice appeared phenotypically normal, although these animals exhibited a modest 35 ± 3% increase in plasma CXCL12 as compared with control. Using two different syngeneic, orthotopic tumor implant models of breast cancer, we discovered that CXCR7ΔEND/ΔEND mice had significantly greater local recurrence of cancer following resection, elevated numbers of circulating tumor cells, and more spontaneous metastases. CXCR7ΔEND/ΔEND mice also showed greater experimental metastases following intracardiac injection of cancer cells. These results establish that endothelial CXCR7 limits breast cancer metastasis at multiple steps in the metastatic cascade, advancing understanding of CXCL12 pathways in tumor environments and informing ongoing drug development targeting CXCR7 in cancer.
Collapse
Affiliation(s)
- A C Stacer
- University of Michigan Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA
| | - J Fenner
- University of Michigan Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA
| | - S P Cavnar
- Department of Biomedical Engineering, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA
| | - A Xiao
- University of Michigan Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA
| | - S Zhao
- Department of Radiation Oncology, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA
| | - S L Chang
- Depatment of Chemical Engineering, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA
| | - A Salomonnson
- University of Michigan Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA
| | - K E Luker
- University of Michigan Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA
| | - G D Luker
- University of Michigan Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA.,Department of Biomedical Engineering, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA.,Department of Microbiology and Immunology, University of Michigan Medical School and College of Engineering, Ann Arbor, MI, USA
| |
Collapse
|
38
|
Liu X, Dai LI, Zhou R. Association between preeclampsia and the CXC chemokine family (Review). Exp Ther Med 2015; 9:1572-1576. [PMID: 26136860 DOI: 10.3892/etm.2015.2337] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 02/18/2015] [Indexed: 12/30/2022] Open
Abstract
Preeclampsia is a major cause of maternal and perinatal mortality and morbidity, characterized by gestational hypertension, proteinuria, systemic endothelial cell activation and an exaggerated inflammatory response. The precise cause of preeclampsia is not currently known; however, it is widely accepted that the pathogenesis of preeclampsia involves inadequate trophoblast invasion, leading to generalized endothelial dysfunction and an exaggerated inflammatory response. Chemokines are a superfamily of structurally similar proteins that mediate cell recruitment, angiogenesis, immunity and stem cell trafficking. CXC chemokines are a family of cytokines, unique in their ability to behave in a disparate manner in the regulation of angiogenesis. The CXC chemokine family further divides into two subfamilies; CXC ELR+, which promotes angiogenesis, and CXC ELR-, which inhibits angiogenesis. Furthermore, CXC chemokines are involved in the pathogenesis of various conditions, including malignant tumors, wound repair, chronic inflammation, atherosclerosis and potentially preeclampsia.
Collapse
Affiliation(s)
- Xijing Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - L I Dai
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Rong Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
39
|
Izumi Y, Hoshino Y, Hosoya K, Takagi S, Okumura M. Isolation and characterization of canine tumor endothelial cells. J Vet Med Sci 2014; 77:359-63. [PMID: 25482496 PMCID: PMC4383786 DOI: 10.1292/jvms.14-0347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
The present study involved the isolation and characterization of canine tumor endothelial
cells (TECs) from 2 malignancies. TECs were isolated using magnetic cell sorting following
FITC labeling with UEA1 lectin, and they were characterized by measuring genetic and
histopathological endothelial markers. Isolated TECs exhibited a cobblestone-like
morphology and expressed both vascular endothelial growth factor receptor 2 (VEGFR2) and
Von Willebrand factor (vWF). Further, both TECs and tumor cells derived from a seminoma
exhibited increased C-X-C chemokine receptor type 7 (CXCR7) expression. However, CXCR7
expression was not detected in TECs and tumor cells derived from a hepatocellular
carcinoma. Understanding TEC specific traits may be important in the development of more
efficacious anti-angiogenic therapies that do not induce adverse effects.
Collapse
Affiliation(s)
- Yusuke Izumi
- Laboratory of Veterinary Surgery, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | | | | | | | | |
Collapse
|
40
|
Ohmura-Kakutani H, Akiyama K, Maishi N, Ohga N, Hida Y, Kawamoto T, Iida J, Shindoh M, Tsuchiya K, Shinohara N, Hida K. Identification of tumor endothelial cells with high aldehyde dehydrogenase activity and a highly angiogenic phenotype. PLoS One 2014; 9:e113910. [PMID: 25437864 PMCID: PMC4250080 DOI: 10.1371/journal.pone.0113910] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 10/30/2014] [Indexed: 11/19/2022] Open
Abstract
Tumor blood vessels play an important role in tumor progression and metastasis. It has been reported that tumor endothelial cells (TECs) exhibit highly angiogenic phenotypes compared with those of normal endothelial cells (NECs). TECs show higher proliferative and migratory abilities than those NECs, together with upregulation of vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR2). Furthermore, compared with NECs, stem cell markers such as Sca-1, CD90, and multidrug resistance 1 are upregulated in TECs, suggesting that stem-like cells exist in tumor blood vessels. In this study, to reveal the biological role of stem-like TECs, we analyzed expression of the stem cell marker aldehyde dehydrogenase (ALDH) in TECs and characterized ALDHhigh TECs. TECs and NECs were isolated from melanoma-xenografted nude mice and normal dermis, respectively. ALDH mRNA expression and activity were higher in TECs than those in NECs. Next, ALDHhigh/low TECs were isolated by fluorescence-activated cell sorting to compare their characteristics. Compared with ALDHlow TECs, ALDHhigh TECs formed more tubes on Matrigel-coated plates and sustained the tubular networks longer. Furthermore, VEGFR2 expression was higher in ALDHhigh TECs than that in ALDHlow TECs. In addition, ALDH was expressed in the tumor blood vessels of in vivo mouse models of melanoma and oral carcinoma, but not in normal blood vessels. These findings indicate that ALDHhigh TECs exhibit an angiogenic phenotype. Stem-like TECs may have an essential role in tumor angiogenesis.
Collapse
Affiliation(s)
- Hitomi Ohmura-Kakutani
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
- Department of Orthodontics, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Kosuke Akiyama
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
- Division of Vascular Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Nako Maishi
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
- Division of Vascular Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Noritaka Ohga
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Yasuhiro Hida
- Department of Cardiovascular and Thoracic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Taisuke Kawamoto
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Junichiro Iida
- Department of Orthodontics, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Masanobu Shindoh
- Department of Oral Pathology and Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Kunihiko Tsuchiya
- Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Sapporo, Japan
| | - Nobuo Shinohara
- Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Sapporo, Japan
| | - Kyoko Hida
- Department of Vascular Biology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
- Division of Vascular Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- * E-mail:
| |
Collapse
|
41
|
Alam MT, Nagao-Kitamoto H, Ohga N, Akiyama K, Maishi N, Kawamoto T, Shinohara N, Taketomi A, Shindoh M, Hida Y, Hida K. Suprabasin as a novel tumor endothelial cell marker. Cancer Sci 2014; 105:1533-40. [PMID: 25283635 PMCID: PMC4317965 DOI: 10.1111/cas.12549] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 09/26/2014] [Accepted: 09/30/2014] [Indexed: 01/24/2023] Open
Abstract
Recent studies have reported that stromal cells contribute to tumor progression. We previously demonstrated that tumor endothelial cells (TEC) characteristics were different from those of normal endothelial cells (NEC). Furthermore, we performed gene profile analysis in TEC and NEC, revealing that suprabasin (SBSN) was upregulated in TEC compared with NEC. However, its role in TEC is still unknown. Here we showed that SBSN expression was higher in isolated human and mouse TEC than in NEC. SBSN knockdown inhibited the migration and tube formation ability of TEC. We also showed that the AKT pathway was a downstream factor of SBSN. These findings suggest that SBSN is involved in the angiogenic potential of TEC and may be a novel TEC marker.
Collapse
Affiliation(s)
- Mohammad T Alam
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan; Department of Oral Pathology and Biology, Graduate School of Dental Medicine, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Bielecka ZF, Czarnecka AM, Szczylik C. Genomic Analysis as the First Step toward Personalized Treatment in Renal Cell Carcinoma. Front Oncol 2014; 4:194. [PMID: 25120953 PMCID: PMC4110478 DOI: 10.3389/fonc.2014.00194] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 07/09/2014] [Indexed: 12/13/2022] Open
Abstract
Drug resistance mechanisms in renal cell carcinoma (RCC) still remain elusive. Although most patients initially respond to targeted therapy, acquired resistance can still develop eventually. Most of the patients suffer from intrinsic (genetic) resistance as well, suggesting that there is substantial need to broaden our knowledge in the field of RCC genetics. As molecular abnormalities occur for various reasons, ranging from single nucleotide polymorphisms to large chromosomal defects, conducting whole-genome association studies using high-throughput techniques seems inevitable. In principle, data obtained via genome-wide research should be continued and performed on a large scale for the purposes of drug development and identification of biological pathways underlying cancerogenesis. Genetic alterations are mostly unique for each histological RCC subtype. According to recently published data, RCC is a highly heterogeneous tumor. In this paper, the authors discuss the following: (1) current state-of-the-art knowledge on the potential biomarkers of RCC subtypes; (2) significant obstacles encountered in the translational research on RCC; and (3) recent molecular findings that may have a crucial impact on future therapeutic approaches.
Collapse
Affiliation(s)
- Zofia Felicja Bielecka
- Department of Oncology with the Laboratory of Molecular Oncology, Military Institute of Medicine , Warsaw , Poland ; Postgraduate School of Molecular Medicine, Medical University of Warsaw , Warsaw , Poland
| | - Anna Małgorzata Czarnecka
- Department of Oncology with the Laboratory of Molecular Oncology, Military Institute of Medicine , Warsaw , Poland
| | - Cezary Szczylik
- Department of Oncology with the Laboratory of Molecular Oncology, Military Institute of Medicine , Warsaw , Poland
| |
Collapse
|
43
|
Zhou J, Xiang Y, Yoshimura T, Chen K, Gong W, Huang J, Zhou Y, Yao X, Bian X, Wang JM. The role of chemoattractant receptors in shaping the tumor microenvironment. BIOMED RESEARCH INTERNATIONAL 2014; 2014:751392. [PMID: 25110692 PMCID: PMC4119707 DOI: 10.1155/2014/751392] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/17/2014] [Indexed: 12/13/2022]
Abstract
Chemoattractant receptors are a family of seven transmembrane G protein coupled receptors (GPCRs) initially found to mediate the chemotaxis and activation of immune cells. During the past decades, the functions of these GPCRs have been discovered to not only regulate leukocyte trafficking and promote immune responses, but also play important roles in homeostasis, development, angiogenesis, and tumor progression. Accumulating evidence indicates that chemoattractant GPCRs and their ligands promote the progression of malignant tumors based on their capacity to orchestrate the infiltration of the tumor microenvironment by immune cells, endothelial cells, fibroblasts, and mesenchymal cells. This facilitates the interaction of tumor cells with host cells, tumor cells with tumor cells, and host cells with host cells to provide a basis for the expansion of established tumors and development of distant metastasis. In addition, many malignant tumors of the nonhematopoietic origin express multiple chemoattractant GPCRs that increase the invasiveness and metastasis of tumor cells. Therefore, GPCRs and their ligands constitute targets for the development of novel antitumor therapeutics.
Collapse
Affiliation(s)
- Jiamin Zhou
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
- Endoscopic Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yi Xiang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
- Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Teizo Yoshimura
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Keqiang Chen
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Jian Huang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ye Zhou
- Department of Gastric Cancer and Soft Tissue Surgery, Fudan University Cancer Center, Shanghai 200032, China
| | - Xiaohong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ji Ming Wang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
44
|
Bianco AM, Uno M, Oba-Shinjo SM, Clara CA, de Almeida Galatro TF, Rosemberg S, Teixeira MJ, Nagahashi Marie SK. CXCR7 and CXCR4 Expressions in Infiltrative Astrocytomas and Their Interactions with HIF1α Expression and IDH1 Mutation. Pathol Oncol Res 2014; 21:229-40. [PMID: 24970694 DOI: 10.1007/s12253-014-9813-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 06/04/2014] [Indexed: 11/27/2022]
Abstract
The CXCR7, a new receptor for CXCL12 with higher affinity than CXCR4 has raised key issues on glioma cell migration. The aim of this study is to investigate the CXCR7 mRNA expression in diffuse astrocytomas tissues and to evaluate its interactions with CXCR4 and HIF1α expression and IDH1 mutation. CXCR7, CXCR4 and HIF1α mRNA expression were evaluated in 129 frozen samples of astrocytomas. IDH1 mutation status was analyzed with gene expressions, matched with clinicopathological parameters and overall survival time. Protein expression was analyzed by immunohistochemistry in different grades of astrocytoma and in glioma cell line (U87MG) by confocal microscopy. There was significant difference in the expression levels of the genes studied between astrocytomas and non-neoplasic (NN) controls (p < 0.001). AGII showed no significant correlation between CXCR7/HIF1α (p = 0.548); there was significant correlation between CXCR7/CXCR4 (p = 0.042) and CXCR7/IDH1 (p = 0.008). GBM showed significant correlations between CXCR7/CXCR4 (p = 0.002), CXCR7/IDH1 (p < 0.001) and CXCR7/HIF1α (p = 0.008). HIF1α overexpression was associated with higher expressions of CXCR7 (p = 0.01) and CXCR4 (p < 0.0001), while IDH1 mutation was associated with lower CXCR7 (p = 0.009) and CXCR4 (p = 0.0005) mRNA expressions. Protein expression increased with malignancy and in U87MG cell line was mainly localized in the cellular membrane. CXCR7 was overexpressed in astrocytoma and correlates with CXCR4 and IDH1 in AGII and CXCR4, IDH1 and HIF1α in GBM. Overexpression HIF1α was related with higher expressions of CXCR7 and CXCR4, otherwise IDH1 mutation related with lower expression of both genes. No association between CXCR7 and CXCR4 expression and survival data was related.
Collapse
Affiliation(s)
- Andre Macedo Bianco
- Department of Neurology, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil Laboratory of Molecular and Cellular Biology, LIM15 Av. Dr. Arnaldo, 455, 4th floor, r.4110, Sao Paulo, SP, Brazil, 01246-903,
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Endothelial CD276 (B7-H3) expression is increased in human malignancies and distinguishes between normal and tumour-derived circulating endothelial cells. Br J Cancer 2014; 111:149-56. [PMID: 24892449 PMCID: PMC4090744 DOI: 10.1038/bjc.2014.286] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/10/2014] [Accepted: 04/25/2014] [Indexed: 02/07/2023] Open
Abstract
Background: Mature circulating endothelial cells (CEC) are surrogate markers of endothelial damage. CEC measured in patients with advanced cancer are thought not only to derive from damaged normal vasculature (n-CEC), but also from damaged (t-CEC). Therefore, assays that allow the discrimination between these two putative types of CEC are thought to improve the specificity of the enumeration of CEC in cancer. Methods: Identification of tumour-associated endothelial markers (TEM) by comparing antigen expression on normal vs t-CEC and assess the presence of t-CEC in peripheral blood of cancer patients by incorporating TEM in our novel flow cytometry-based CEC detection assay. Results: No difference in antigen expression between normal and malignant endothelial cells (ECs) was found for CD54, CD109, CD137, CD141, CD144 and CXCR7. In contrast, overexpression for CD105, CD146, CD276 and CD309 was observed in tumour ECs compared with normal ECs. CD276 was most differentially expressed and chosen as a marker for further investigation. CD276-expressing CEC were significantly higher in 15 patients with advanced colorectal cancer (median 9 (range 1–293 cell per 4 ml); P<0.005), in 83 patients with a glioblastoma multiforme (median 10 (range 0–804); P<0.0001) and in 14 patients with advanced breast cancer (median 14 (range 0–390) P<0.05) as compared with 24 healthy individuals (median 3 (range 0–11)). Of all patients with malignancies, 58% had CD276+ CEC counts above the ULN (8 cell per 4 ml). Conclusions: The present study shows that CD276 can be used to discriminate ECs from malignant tissue from ECs from normal tissue. In addition, CD276+ CEC do occur in higher frequencies in patients with advanced cancer.
Collapse
|
46
|
Freitas C, Desnoyer A, Meuris F, Bachelerie F, Balabanian K, Machelon V. The relevance of the chemokine receptor ACKR3/CXCR7 on CXCL12-mediated effects in cancers with a focus on virus-related cancers. Cytokine Growth Factor Rev 2014; 25:307-16. [PMID: 24853339 DOI: 10.1016/j.cytogfr.2014.04.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 04/29/2014] [Indexed: 01/18/2023]
Abstract
Recent studies have highlighted the importance of understanding the molecular determinants of CXCL12-mediated effects in cancers. Once previously thought to interact exclusively with CXCR4, CXCL12 also binds with high affinity to CXCR7 (recently renamed ACKR3), which belongs to an atypical chemokine receptor family whose members fail to activate Gαi proteins but interact with β-arrestins. In addition to its capacity to control CXCL12 bioavailability, ACKR3 can either enhance or dampen CXCR4-mediated signaling and activity. In light of the most recent findings, we have examined the role of ACKR3 in cancer, including a subset of virus-related cancers.
Collapse
Affiliation(s)
- Christelle Freitas
- Univ. Paris-Sud, Laboratoire "Cytokines, Chemokines and Immunopathology", UMR_S996, 32, rue des Carnets, Clamart F-92140, France; INSERM, Univ. Paris-Sud, Laboratory of Excellence in Research on Medication and Innovative Therapeutics (LERMIT), Clamart F-92140, France
| | - Aude Desnoyer
- Univ. Paris-Sud, Laboratoire "Cytokines, Chemokines and Immunopathology", UMR_S996, 32, rue des Carnets, Clamart F-92140, France; INSERM, Univ. Paris-Sud, Laboratory of Excellence in Research on Medication and Innovative Therapeutics (LERMIT), Clamart F-92140, France
| | - Floriane Meuris
- Univ. Paris-Sud, Laboratoire "Cytokines, Chemokines and Immunopathology", UMR_S996, 32, rue des Carnets, Clamart F-92140, France; INSERM, Univ. Paris-Sud, Laboratory of Excellence in Research on Medication and Innovative Therapeutics (LERMIT), Clamart F-92140, France
| | - Françoise Bachelerie
- Univ. Paris-Sud, Laboratoire "Cytokines, Chemokines and Immunopathology", UMR_S996, 32, rue des Carnets, Clamart F-92140, France; INSERM, Univ. Paris-Sud, Laboratory of Excellence in Research on Medication and Innovative Therapeutics (LERMIT), Clamart F-92140, France
| | - Karl Balabanian
- Univ. Paris-Sud, Laboratoire "Cytokines, Chemokines and Immunopathology", UMR_S996, 32, rue des Carnets, Clamart F-92140, France; INSERM, Univ. Paris-Sud, Laboratory of Excellence in Research on Medication and Innovative Therapeutics (LERMIT), Clamart F-92140, France.
| | - Véronique Machelon
- Univ. Paris-Sud, Laboratoire "Cytokines, Chemokines and Immunopathology", UMR_S996, 32, rue des Carnets, Clamart F-92140, France; INSERM, Univ. Paris-Sud, Laboratory of Excellence in Research on Medication and Innovative Therapeutics (LERMIT), Clamart F-92140, France.
| |
Collapse
|
47
|
Otsubo T, Hida Y, Ohga N, Sato H, Kai T, Matsuki Y, Takasu H, Akiyama K, Maishi N, Kawamoto T, Shinohara N, Nonomura K, Hida K. Identification of novel targets for antiangiogenic therapy by comparing the gene expressions of tumor and normal endothelial cells. Cancer Sci 2014; 105:560-7. [PMID: 24602018 PMCID: PMC4317838 DOI: 10.1111/cas.12394] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 03/02/2014] [Accepted: 03/03/2014] [Indexed: 01/25/2023] Open
Abstract
Targeting tumor angiogenesis is an established strategy for cancer therapy. Because angiogenesis is not limited to pathological conditions such as cancer, molecular markers that can distinguish between physiological and pathological angiogenesis are required to develop more effective and safer approaches for cancer treatment. To identify such molecules, we determined the gene expression profiles of murine tumor endothelial cells (mTEC) and murine normal endothelial cells using DNA microarray analysis followed by quantitative reverse transcription–polymerase chain reaction analysis. We identified 131 genes that were differentially upregulated in mTEC. Functional analysis using siRNA-mediated gene silencing revealed five novel tumor endothelial cell markers that were involved in the proliferation or migration of mTEC. The expression of DEF6 and TMEM176B was upregulated in tumor vessels of human renal cell carcinoma specimens, suggesting that they are potential targets for antiangiogenic intervention for renal cell carcinoma. Comparative gene expression analysis revealed molecular differences between tumor endothelial cells and normal endothelial cells and identified novel tumor endothelial cell markers that may be exploited to target tumor angiogenesis for cancer treatment.
Collapse
Affiliation(s)
- Tsuguteru Otsubo
- Drug Discovery II, DSP Cancer Institute, Dainippon Sumitomo Pharma Co., Ltd, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Chemokines have fundamental roles in regulating immune and inflammatory responses, primarily through their control of leukocyte migration and localization. The biological functions of chemokines are typically mediated by signalling through G protein-coupled chemokine receptors, but chemokines are also bound by a small family of atypical chemokine receptors (ACKRs), the members of which are unified by their inability to initiate classical signalling pathways after ligand binding. These ACKRs are emerging as crucial regulatory components of chemokine networks in a wide range of developmental, physiological and pathological contexts. In this Review, we discuss the biochemical and immunological properties of ACKRs and the potential unifying themes in this family, and we highlight recent studies that identify novel roles for these molecules in development , homeostasis, inflammatory disease, infection and cancer.
Collapse
|
49
|
Stimulation of TLR4 by LMW-HA induces metastasis in human papillary thyroid carcinoma through CXCR7. Clin Dev Immunol 2013; 2013:712561. [PMID: 24363762 PMCID: PMC3865734 DOI: 10.1155/2013/712561] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 10/12/2013] [Indexed: 12/13/2022]
Abstract
In inflammatory sites, high molecular weight hyaluronan fragments are degraded into lower molecular weight hyaluronan fragments (LMW-HA) to regulate immune responses. However, the function of LMW-HA in PTC progression remains to be elucidated. In this study, we found that receptor of LMW-HA, TLR4, was aberrantly overexpressed in PTC tissues and cell line W3. Exposure of W3 cells to LMW-HA promoted cell proliferation and migration via TLR4. Knockdown of TLR4 has provided evidence that TLR4 is essential for LMW-HA-induced CXCR7 expression, which is responsible for LMW-HA-induced proliferation and migration of W3 cells. In tumor-bearing adult nude mice, stimulation of LMW-HA on W3 cells promotes CXCR7 expression in tumor masses (P = 0.002) and tumor growth (P < 0.001). To further confirm our findings, we investigated the clinicopathologic significance of TLR4 and CXCR7 expression using immumohistochemistry in 135 human PTC tissues and 56 normal thyroid tissue samples. Higher rates of TLR4 (53%) and CXCR7 (24%) expression were found in PTC tissues than in normal tissues. Expression of TLR4 or CXCR7 is associated with tumor size and lymph node metastasis. Therefore, LMW-HA may contribute to the development of PTC via TLR4/CXCR7 pathway, which may be a novel target for PTC immunomodulatory therapy.
Collapse
|
50
|
The peculiarities of the SDF-1/CXCL12 system: in some cells, CXCR4 and CXCR7 sing solos, in others, they sing duets. Cell Tissue Res 2013; 355:239-53. [DOI: 10.1007/s00441-013-1747-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 10/17/2013] [Indexed: 12/26/2022]
|