1
|
Wang P, Jie Y, Yao L, Sun YM, Jiang DP, Zhang SQ, Wang XY, Fan Y. Cells in the liver microenvironment regulate the process of liver metastasis. Cell Biochem Funct 2024; 42:e3969. [PMID: 38459746 DOI: 10.1002/cbf.3969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/31/2024] [Accepted: 02/26/2024] [Indexed: 03/10/2024]
Abstract
The research of liver metastasis is a developing field. The ability of tumor cells to invade the liver depends on the complicated interactions between metastatic cells and local subpopulations in the liver (including Kupffer cells, hepatic stellate cells, liver sinusoidal endothelial cells, and immune-related cells). These interactions are mainly mediated by intercellular adhesion and the release of cytokines. Cell populations in the liver microenvironment can play a dual role in the progression of liver metastasis through different mechanisms. At the same time, we can see the participation of liver parenchymal cells and nonparenchymal cells in the process of liver metastasis of different tumors. Therefore, the purpose of this article is to summarize the relationship between cellular components of liver microenvironment and metastasis and emphasize the importance of different cells in the occurrence or potential regression of liver metastasis.
Collapse
Affiliation(s)
- Pei Wang
- Cancer Institute, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yu Jie
- Cancer Institute, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Lin Yao
- Cancer Institute, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yi-Meng Sun
- Cancer Institute, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Da-Peng Jiang
- Cancer Institute, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Shi-Qi Zhang
- Department of Gastroenterology, The Affiliated Suqian First People's Hospital of Xuzhou Medical University, Suqian, Jiangsu, China
| | - Xiao-Yan Wang
- Department of Gastroenterology, The Affiliated Suqian First People's Hospital of Xuzhou Medical University, Suqian, Jiangsu, China
| | - Yu Fan
- Cancer Institute, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
2
|
Huang R, Ding J, Xie WF. Liver cancer. SINUSOIDAL CELLS IN LIVER DISEASES 2024:349-366. [DOI: 10.1016/b978-0-323-95262-0.00017-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
3
|
Tsilimigras DI, Ntanasis-Stathopoulos I, Pawlik TM. Molecular Mechanisms of Colorectal Liver Metastases. Cells 2023; 12:1657. [PMID: 37371127 DOI: 10.3390/cells12121657] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
The liver is the most frequently target for metastasis among patients with colorectal cancer mainly because of the portal vein circulation that directly connects the colon and rectum with the liver. The liver tumor microenvironment consists of different cell types each with unique characteristics and functions that modulate the antigen recognition and immune system activation. Primary tumors from other sites "prime" the liver prior to the seeding of cancer cells, creating a pre-metastatic niche. Following invasion into the liver, four different phases are key to the development of liver metastases: a microvascular phase in which cancer cells infiltrate and become trapped in sinusoidal vessels; an extravascular, pre-angiogenic phase; an angiogenic phase that supplies oxygen and nutrients to cancer cells; and a growth phase in which metastatic cells multiply and enlarge to form detectable tumors. Exosomes carry proteins, lipids, as well as genetic information that can create a pre-metastatic niche in distant sites, including the liver. The complexity of angiogenic mechanisms and the exploitation of the vasculature in situ by cancer cells have limited the efficacy of currently available anti-angiogenic therapies. Delineating the molecular mechanisms implicated in colorectal liver metastases is crucial to understand and predict tumor progression; the development of distant metastases; and resistance to chemotherapy, immunotherapy, and targeted treatment.
Collapse
Affiliation(s)
- Diamantis I Tsilimigras
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, 395 W. 12th Ave., Columbus, OH 43210, USA
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, Alexandra General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, 395 W. 12th Ave., Columbus, OH 43210, USA
| |
Collapse
|
4
|
Goïta AA, Guenot D. Colorectal Cancer: The Contribution of CXCL12 and Its Receptors CXCR4 and CXCR7. Cancers (Basel) 2022; 14:1810. [PMID: 35406582 PMCID: PMC8997717 DOI: 10.3390/cancers14071810] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer is one of the most common cancers, and diagnosis at late metastatic stages is the main cause of death related to this cancer. This progression to metastasis is complex and involves different molecules such as the chemokine CXCL12 and its two receptors CXCR4 and CXCR7. The high expression of receptors in CRC is often associated with a poor prognosis and aggressiveness of the tumor. The interaction of CXCL12 and its receptors activates signaling pathways that induce chemotaxis, proliferation, migration, and cell invasion. To this end, receptor inhibitors were developed, and their use in preclinical and clinical studies is ongoing. This review provides an overview of studies involving CXCR4 and CXCR7 in CRC with an update on their targeting in anti-cancer therapies.
Collapse
Affiliation(s)
| | - Dominique Guenot
- INSERM U1113/Unistra, IRFAC—Interface de Recherche Fondamentale et Appliquée en Cancérologie, 67200 Strasbourg, France;
| |
Collapse
|
5
|
Amer S, Nabil M, Negm M. Expression of Podoplanin in Hepatocellular Carcinoma in a Sample of Egyptian Population – Immunohistopathological Study. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.8460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Hepatocellular carcinoma (HCC) is a highly incident malignancy with a dreadful prognosis. It evolves through a multistep process, with a contribution from different stromal cells like cancer associated fibroblasts. Podoplanin is a glycoprotein that influences epithelial mesenchymal interplay facilitating the tumor invasion.
AIM: The aim of the study was to evaluate the immunohistochemical expression of Podoplanin in HCC in cancer associated fibroblasts (CAFs) and malignant hepatocytes as well as assessing the lymphovascular density, and correlating them with the clinicopathological parameters.
METHODS: Sixty formalin-fixed paraffin-embedded HCC tissue blocks were retrieved from the pathology Department of the National Hepatology and Tropical Medicine Research Institute and Kasr Al-aini Hospital during the period of January 2012 till December 2019. The specimens were obtained through partial or total hepatectomy inclusion criteria included HCC cases obtained through resection type biopsy and those having no history of pre-operative cancer therapy, while cases with insufficient data, core biopsy, and marked necrosis were excluded from the study. Tumor tissue blocks were immunostained for Podoplanin and its expression was interpreted in lymphatic vessels, CAFs, and malignant hepatocytes.
RESULTS: Podoplanin expression in CAFs and malignant hepatocytes was detected in the majority of HCC cases (81.7%) and (88.3%), respectively. The malignant hepatocytes showed increased expression of Grade 1 immunostaining (36.7%). High lymphovascular density was detected over the majority of the cases (73.3%). Podoplanin expression was significantly correlated with higher mean age, male gender, presence of viral infection, cirrhosis, and higher tumor grade. Unifocal tumor mass, tumor size <5 cm, and presence of invasion showed a significant correlation with Podoplanin in malignant hepatocytes and CAFs for the formers and the later, respectively.
CONCLUSION: Podoplanin is highly expressed in HCC, which could be used as a prognostic marker for lymphangiogenesis. Furthermore, within the malignant hepatocytes and CAFs suggesting a role in hepatocellular tumorigenesis. Podoplanin targeted therapy can be investigated to slow down the tumor progression and metastasis.
Collapse
|
6
|
Olaso E, Benedicto A, Lopategi A, Cossio FP, Arteta B. A Synthetic Analog of Resveratrol Inhibits the Proangiogenic Response of Liver Sinusoidal Cells during Hepatic Metastasis. Biomol Ther (Seoul) 2021; 30:162-169. [PMID: 34873071 PMCID: PMC8902452 DOI: 10.4062/biomolther.2021.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/13/2021] [Accepted: 08/14/2021] [Indexed: 11/17/2022] Open
Abstract
We utilized Fas21, a resveratrol analog, to modulate the function of hepatic stellate cells (HSCs) and liver sinusoidal endothelial cells (LSECs) during the angiogenic phase of murine liver metastasis by B16 melanoma and 51b colorectal carcinoma. Preangiogenic micrometastases were treated with Fas21 (1 mg/kg/day) or vehicle during the development of intra-angiogenic tracts. Mice treated with Fas21 showed reduced liver tumor foci in both liver metastasis models. Micrometastases were classified immunohistochemically, as well as according to their position coordinates and connection to local microvasculature. The volume of liver occupied by sinusoidal-type foci, containing infiltrating angiogenic capillaries, decreased by ~50% in Fas21-treated mice compared to vehicle-treated ones in both tumor metastasis models. The volume of portal foci, containing peripheral neoangiogenesis within a discontinuous layer of myofibroblasts, was similar in all experimental groups in both tumor metastasis models, but displayed enhanced necrotic central areas devoid of angiogenesis following Fas21 treatment. As a result, sinusoidal tumors from mice treated with Fas21 showed a 50% reduction in desmin(+)/asma(+) HSCs and CD31(+) vessel density, and a 45% reduction in intrametastatic VEGF mRNA compared with sinusoidal tumors from vehicle-treated mice. Necrotic portal metastases increased 2-4-fold in treated mice. In vitro, Fas21 reduced VEGF secretion by HSCs and 51b cells dose-dependently. Additionally, HSCs migration in response to tumor soluble factors was dose-dependently diminished by Fas21, as was LSEC migration in response to HSCs and tumor soluble factors. Resveratrol analog Fas21 inhibits the proangiogenic response of HSCs and LSECs during the development of murine liver metastasis.
Collapse
Affiliation(s)
- Elvira Olaso
- Tumor Microenvironment Group, Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, Leioa 48940, Spain
| | - Aitor Benedicto
- Tumor Microenvironment Group, Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, Leioa 48940, Spain
| | - Aritz Lopategi
- Tumor Microenvironment Group, Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, Leioa 48940, Spain
| | - Fernando P Cossio
- Department of Organic Chemistry, School of Sciences, University of the Basque Country, Donosti 20018, Spain
| | - Beatriz Arteta
- Tumor Microenvironment Group, Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, Leioa 48940, Spain
| |
Collapse
|
7
|
Bastani S, Akbarzadeh M, Rastgar Rezaei Y, Farzane A, Nouri M, Mollapour Sisakht M, Fattahi A, Akbarzadeh M, Reiter RJ. Melatonin as a Therapeutic Agent for the Inhibition of Hypoxia-Induced Tumor Progression: A Description of Possible Mechanisms Involved. Int J Mol Sci 2021; 22:10874. [PMID: 34639215 PMCID: PMC8509383 DOI: 10.3390/ijms221910874] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/24/2021] [Accepted: 09/29/2021] [Indexed: 12/27/2022] Open
Abstract
Hypoxia has an important role in tumor progression via the up-regulation of growth factors and cellular adaptation genes. These changes promote cell survival, proliferation, invasion, metastasis, angiogenesis, and energy metabolism in favor of cancer development. Hypoxia also plays a central role in determining the resistance of tumors to chemotherapy. Hypoxia of the tumor microenvironment provides an opportunity to develop new therapeutic strategies that may selectively induce apoptosis of the hypoxic cancer cells. Melatonin is well known for its role in the regulation of circadian rhythms and seasonal reproduction. Numerous studies have also documented the anti-cancer properties of melatonin, including anti-proliferation, anti-angiogenesis, and apoptosis promotion. In this paper, we hypothesized that melatonin exerts anti-cancer effects by inhibiting hypoxia-induced pathways. Considering this action, co-administration of melatonin in combination with other therapeutic medications might increase the effectiveness of anti-cancer drugs. In this review, we discussed the possible signaling pathways by which melatonin inhibits hypoxia-induced cancer cell survival, invasion, migration, and metabolism, as well as tumor angiogenesis.
Collapse
Affiliation(s)
- Sepideh Bastani
- Research Center for Pharmaceutical Nanotechnology (RCPN), Tabriz University of Medical Sciences, Tabriz 51368, Iran;
- Stem Cell And Regenerative Medicine Institute (SCARM), Tabriz University of Medical Sciences, Tabriz 51368, Iran;
| | - Moloud Akbarzadeh
- Stem Cell And Regenerative Medicine Institute (SCARM), Tabriz University of Medical Sciences, Tabriz 51368, Iran;
- Department of Cellular and Molecular Biology, Faculty of Biological Science, Azarbaijan Shahid Madani University, Tabriz 51368, Iran
| | - Yeganeh Rastgar Rezaei
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51368, Iran;
| | - Ali Farzane
- Department of Health Information Management, School of Allied Medical Science, Tehran University of Medical Sciences, Tehran 11369, Iran;
| | - Mohammad Nouri
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51368, Iran;
| | - Mahsa Mollapour Sisakht
- Stem Cell and Regenerative Medicine Center of Excellence, Tehran University of Medical Sciences, Tehran 11369, Iran;
- Department of Biochemistry, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51368, Iran;
- Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University of Erlangen–Nürnberg, Comprehensive Cancer Center ER-EMN, 91054 Erlangen, Germany
| | - Maryam Akbarzadeh
- Department of Biochemistry, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health, Long School of Medicine, San Antonio, TX 78229, USA;
| |
Collapse
|
8
|
Wang Z, Liu J, Huang H, Ye M, Li X, Wu R, Liu H, Song Y. Metastasis-associated fibroblasts: an emerging target for metastatic cancer. Biomark Res 2021; 9:47. [PMID: 34112258 PMCID: PMC8194104 DOI: 10.1186/s40364-021-00305-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
Metastasis suggests a poor prognosis for cancer patients, and treatment strategies for metastatic cancer are still very limited. Numerous studies have shown that cancer-associated fibroblasts (CAFs), a large component of the tumor microenvironment, contribute to tumor metastasis. Stromal fibroblasts at metastatic sites are different from CAFs within primary tumors and can be termed metastasis-associated fibroblasts (MAFs), and they also make great contributions to the establishment of metastatic lesions and the therapeutic resistance of metastatic tumors. MAFs are capable of remodeling the extracellular matrix of metastatic tumors, modulating immune cells in the tumor microenvironment, promoting angiogenesis and enhancing malignant tumor phenotypes. Thus, MAFs can help establish premetastatic niches and mediate resistance to therapeutic strategies, including immunotherapy and antiangiogenic therapy. The results of preclinical studies suggest that targeting MAFs can alleviate the progression of metastatic cancer and mitigate therapeutic resistance, indicating that MAFs are a promising target for metastatic cancer. Here, we comprehensively summarize the existing evidence on MAFs and discuss their origins, generation, functions and related therapeutic strategies in an effort to provide a better understanding of MAFs and offer treatment perspectives for metastatic cancer.
Collapse
Affiliation(s)
- Zimu Wang
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China
| | - Jiaxin Liu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China
| | - Hairong Huang
- Department of Cardiothoracic Surgery, Jinling Hospital, 210002, Nanjing, China
| | - Mingxiang Ye
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China
| | - Xinying Li
- Department of Respiratory Medicine, Nanjing Drum Tower Hospital, Nanjing University School of Medicine, 210008, Nanjing, Jiangsu, China
| | - Ranpu Wu
- Department of Respiratory Medicine, Jinling Hospital, Southeast University of Medicine, 210009, Nanjing, Jiangsu, China
| | - Hongbing Liu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China.
| | - Yong Song
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China.
| |
Collapse
|
9
|
Wang J, Lou J, Fu L, Jin Q. An independent poor-prognosis subtype of hepatocellular carcinoma based on the tumor microenvironment. J Int Med Res 2021; 49:300060520980646. [PMID: 33567957 PMCID: PMC7883156 DOI: 10.1177/0300060520980646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a highly malignant tumor with a particularly poor prognosis. The tumor microenvironment (TME) is closely associated with tumorigenesis, progression, and treatment. However, the relationship between TME genes and HCC patient prognosis is poorly understood. Methods In this study, we identified two prognostic subtypes based on the TME using data from The Cancer Genome Atlas and Gene Expression Omnibus. The Microenvironment Cell Populations-counter method was used to evaluate immune cell infiltration in HCC. Differentially expressed genes between molecular subtypes were calculated with the Limma package, and clusterProfiler was used for Gene Ontology and Kyoto Encyclopedia of Genes and Genomes functional enrichment analyses to identify genes related to the independent subtypes. We also integrated mRNA expression data into our bioinformatics analysis. Results We identified 4227 TME-associated genes and 640 genes related to the prognosis of HCC. We defined two major subtypes (Clusters 1 and 2) based on the analysis of TME-associated gene expression. Cluster 1 was characterized by increased expression of immune-associated genes and a worse prognosis than Cluster 2. Conclusions The identification of these HCC subtypes based on the TME provides further insight into the molecular mechanisms and prediction of HCC prognosis.
Collapse
Affiliation(s)
- Junfeng Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Department of General Surgery, Hangzhou Mingzhou Hospital, Hangzhou, China
| | - Jianying Lou
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Fu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Department of General Surgery, Hangzhou Mingzhou Hospital, Hangzhou, China
| | - Qu Jin
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Department of General Surgery, Hangzhou Mingzhou Hospital, Hangzhou, China
| |
Collapse
|
10
|
Herrero A, Benedicto A, Romayor I, Olaso E, Arteta B. Inhibition of COX-2 Impairs Colon Cancer Liver Metastasis through Reduced Stromal Cell Reaction. Biomol Ther (Seoul) 2021; 29:342-351. [PMID: 33455946 PMCID: PMC8094073 DOI: 10.4062/biomolther.2020.160] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 12/18/2022] Open
Abstract
Liver colonization is initiated through the interplay between tumor cells and adhesion molecules present in liver sinusoidal endothelial cells (LSECs). This crosstalk stimulates tumor COX-2 upregulation and PGE2 secretion. To elucidate the role of the LSEC intercellular adhesion molecule-1 (ICAM-1) in the prometastatic response exerted by tumor and stromal COX-2, we utilized celecoxib (CLX) as a COX-2 inhibitory agent. We analyzed the in vitro proliferative and secretory responses of murine C26 colorectal cancer (CRC) cells to soluble ICAM-1 (sICAM-1), cultured alone or with LSECs, and their effect on LSEC and hepatic stellate cell (HSC) migration and in vivo liver metastasis. CLX reduced sICAM-1-stimulated COX-2 activation and PGE2 secretion in C26 cells cultured alone or cocultured with LSECs. Moreover, CLX abrogated sICAM-1-induced C26 cell proliferation and C26 secretion of promigratory factors for LSECs and HSCs. Interestingly, CLX reduced the protumoral response of HSC, reducing their migratory potential when stimulated with C26 secretomes and impairing their secretion of chemotactic factors for LSECs and C26 cells and proliferative factors for C26 cells. In vivo, CLX abrogated the prometastatic ability of sICAM-1-activated C26 cells while reducing liver metastasis. COX-2 inhibition blocked the creation of a favorable tumor microenvironment (TME) by hindering the intratumoral recruitment of activated HSCs and macrophages in addition to the accumulation of fibrillar collagen. These results point to COX-2 being a key modulator of processes initiated by host ICAM-1 during tumor cell/LSEC/HSC crosstalk, leading to the creation of a prometastatic TME in the liver.
Collapse
Affiliation(s)
- Alba Herrero
- Department of Cell Biology and Histology, University of the Basque Country, School of Medicine and Nursing, Leioa 48940, Bizkaia, Spain
| | - Aitor Benedicto
- Department of Cell Biology and Histology, University of the Basque Country, School of Medicine and Nursing, Leioa 48940, Bizkaia, Spain
| | - Irene Romayor
- Department of Cell Biology and Histology, University of the Basque Country, School of Medicine and Nursing, Leioa 48940, Bizkaia, Spain
| | - Elvira Olaso
- Department of Cell Biology and Histology, University of the Basque Country, School of Medicine and Nursing, Leioa 48940, Bizkaia, Spain
| | - Beatriz Arteta
- Department of Cell Biology and Histology, University of the Basque Country, School of Medicine and Nursing, Leioa 48940, Bizkaia, Spain
| |
Collapse
|
11
|
Benedicto A, Hernandez-Unzueta I, Sanz E, Márquez J. Ocoxin Increases the Antitumor Effect of BRAF Inhibition and Reduces Cancer Associated Fibroblast-Mediated Chemoresistance and Protumoral Activity in Metastatic Melanoma. Nutrients 2021; 13:686. [PMID: 33669949 PMCID: PMC7924874 DOI: 10.3390/nu13020686] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/11/2021] [Accepted: 02/11/2021] [Indexed: 02/07/2023] Open
Abstract
Whereas the prevalence of several cancer types is decreasing, skin malignancies are growing more common every year. Malignant melanoma is the most aggressive form of skin cancer with high metastatic capacity. In most cases, malignant melanoma shows acquired therapy resistance. We evaluated the ability of Ocoxin, a natural compound-based antioxidant and anti-inflammatory nutritional complement, to exert an antitumor effect in melanoma. To do so, the cytotoxicity of Ocoxin in a panel of BRAF-mutated murine and human melanoma cell lines was tested alone and in combination with BRAF inhibitor Vemurafenib. Our results revealed a potent cytotoxic effect of Ocoxin against melanoma cells and a synergic effect when combined with Vemurafenib, reducing viability and increasing apoptosis. Besides, Ocoxin interferes with the cell cycle, impairs the inherent and fibroblast-mediated melanoma cell migration, and reduces resistance to BRAF inhibition. Proteomic analysis revealed reduced tumor secretion of inflammatory factors Galectin-1, Osteopontin, CCL5, and CCL9 upon treatment with Ocoxin. Moreover, RNASeq showed that Ocoxin downregulated the cell cycle and proliferation-related genes. In vivo, Ocoxin reduced the number of lung metastasis of YUMM-1.7 melanoma cells. Therefore, Ocoxin arises as a good candidate for clinical trials analyzing the beneficial effects in patients suffering from this cutaneous malignancy.
Collapse
Affiliation(s)
- Aitor Benedicto
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country, 48940 Leioa, Bizkaia, Spain; (A.B.); (I.H.-U.)
| | - Iera Hernandez-Unzueta
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country, 48940 Leioa, Bizkaia, Spain; (A.B.); (I.H.-U.)
| | - Eduardo Sanz
- Research and Development, Catalysis S.L., 28016 Madrid, Spain;
| | - Joana Márquez
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country, 48940 Leioa, Bizkaia, Spain; (A.B.); (I.H.-U.)
| |
Collapse
|
12
|
Garcia-Vicién G, Mezheyeuski A, Bañuls M, Ruiz-Roig N, Molleví DG. The Tumor Microenvironment in Liver Metastases from Colorectal Carcinoma in the Context of the Histologic Growth Patterns. Int J Mol Sci 2021; 22:1544. [PMID: 33546502 PMCID: PMC7913731 DOI: 10.3390/ijms22041544] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal carcinoma (CRC) is the third most common cancer. Likewise, it is a disease that has a long survival if it is prematurely detected. However, more than 50% of patients will develop metastases, mainly in the liver (LM-CRC), throughout the evolution of their disease, which accounts for most CRC-related deaths. Treatment it is certainly a controversial issue, since it has not been shown to increase overall survival in the adjuvant setting, although it does improve disease free survival (DFS). Moreover, current chemotherapy combinations are administered based on data extrapolated from primary tumors (PT), not considering that LM-CRC present a very particular tumor microenvironment that can radically condition the effectiveness of treatments designed for a PT. The liver has a particular histology and microenvironment that can determine tumor growth and response to treatments: double blood supply, vascularization through fenestrated sinusoids and the presence of different mesenchymal cell types, among other particularities. Likewise, the liver presents a peculiar immune response against tumor cells, a fact that correlates with the poor response to immunotherapy. All these aspects will be addressed in this review, putting them in the context of the histological growth patterns of LM-CRC, a particular pathologic feature with both prognostic and predictive repercussions.
Collapse
Affiliation(s)
- Gemma Garcia-Vicién
- Tumoral and Stromal Chemoresistance Group, Molecular Mechanisms and Experimental Therapy in Oncology Program (ONCOBELL), Institut d’Investigació Biomèdica de Bellvitge—IDIBELL, 08908 L’Hospitalet de Llobregat, Spain; (G.G.-V.); (M.B.); (N.R.-R.)
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, 08908 L’Hospitalet de Llobregat, Spain
| | - Artur Mezheyeuski
- Department of Immunology, Genetics and Pathology, Uppsala University, 752 37 Uppsala, Sweden;
| | - María Bañuls
- Tumoral and Stromal Chemoresistance Group, Molecular Mechanisms and Experimental Therapy in Oncology Program (ONCOBELL), Institut d’Investigació Biomèdica de Bellvitge—IDIBELL, 08908 L’Hospitalet de Llobregat, Spain; (G.G.-V.); (M.B.); (N.R.-R.)
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, 08908 L’Hospitalet de Llobregat, Spain
| | - Núria Ruiz-Roig
- Tumoral and Stromal Chemoresistance Group, Molecular Mechanisms and Experimental Therapy in Oncology Program (ONCOBELL), Institut d’Investigació Biomèdica de Bellvitge—IDIBELL, 08908 L’Hospitalet de Llobregat, Spain; (G.G.-V.); (M.B.); (N.R.-R.)
- Department of Pathology, Hospital Universitari de Bellvitge, 08908 L’Hospitalet de Llobregat, Spain
| | - David G. Molleví
- Tumoral and Stromal Chemoresistance Group, Molecular Mechanisms and Experimental Therapy in Oncology Program (ONCOBELL), Institut d’Investigació Biomèdica de Bellvitge—IDIBELL, 08908 L’Hospitalet de Llobregat, Spain; (G.G.-V.); (M.B.); (N.R.-R.)
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, 08908 L’Hospitalet de Llobregat, Spain
| |
Collapse
|
13
|
Benedicto A, Sanz E, Márquez J. Ocoxin as a complement to first line treatments in cancer. Int J Med Sci 2021; 18:835-845. [PMID: 33437220 PMCID: PMC7797552 DOI: 10.7150/ijms.50122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/25/2020] [Indexed: 12/18/2022] Open
Abstract
Chemotherapy and radiotherapy are the most frequent treatment for patients suffering from malignant progression of cancer. Even though new treatments are now being implemented, administration of these chemotherapeutic agents remains as the first line option in many tumor types. However, the secondary effects of these compounds represent one of the main reasons cancer patients lose life quality during disease progression. Recent data suggests that Ocoxin, a plant extract and natural compound based nutritional complement rich in antioxidants and anti-inflammatory mediators exerts a positive effect in patients receiving chemotherapy and radiotherapy. This mixture attenuates the chemotherapy and radiotherapy-related side effects such as radiation-induced skin burns and mucositis, chemotherapy-related diarrhea, hepatic toxicity and blood-infection. Moreover, it has been proven to be effective as anticancer agent in different tumor models both in vitro and in vivo, potentiating the cytotoxic effect of several chemotherapy compounds such as Lapatinib, Gemcitabine, Paclitaxel, Sorafenib and Irinotecan. The aim of this review is to put some light on the potential of this nutritional mixture as an anticancer agent and complement for the standard chemotherapy routine.
Collapse
Affiliation(s)
- Aitor Benedicto
- Department of Cellular Biology and Histology, School of Medicine and Nursing, University of the Basque Country, 48940, Leioa, Bizkaia, Spain
| | | | - Joana Márquez
- Department of Cellular Biology and Histology, School of Medicine and Nursing, University of the Basque Country, 48940, Leioa, Bizkaia, Spain
| |
Collapse
|
14
|
Marvin DL, Heijboer R, ten Dijke P, Ritsma L. TGF-β signaling in liver metastasis. Clin Transl Med 2020; 10:e160. [PMID: 33252863 PMCID: PMC7701955 DOI: 10.1002/ctm2.160] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/07/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
The presence of liver metastases drastically worsens the prognosis of cancer patients. The liver is the second most prevalent metastatic site in cancer patients, but systemic therapeutic opportunities that target liver metastases are still limited. To aid the discovery of novel treatment options for metastatic liver disease, we provide insight into the cellular and molecular steps required for liver colonization. For successful colonization in the liver, adaptation of tumor cells and surrounding stroma is essential. This includes the formation of a pre-metastatic niche, the creation of a fibrotic and immune suppressive environment, angiogenesis, and adaptation of tumor cells. We illustrate that transforming growth factor β (TGF-β) is a central cytokine in all these processes. At last, we devise that future research should focus on TGF-β inhibitory strategies, especially in combination with immunotherapy. This promising systemic treatment strategy has potential to eliminate distant metastases as the efficacy of immunotherapy will be enhanced.
Collapse
Affiliation(s)
- Dieuwke L Marvin
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| | - Rosan Heijboer
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| | - Peter ten Dijke
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| | - Laila Ritsma
- Department of Cell and Chemical Biology and Oncode InstituteLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
15
|
Apraiz A, Benedicto A, Marquez J, Agüera-Lorente A, Asumendi A, Olaso E, Arteta B. Innate Lymphoid Cells in the Malignant Melanoma Microenvironment. Cancers (Basel) 2020; 12:cancers12113177. [PMID: 33138017 PMCID: PMC7692065 DOI: 10.3390/cancers12113177] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Innate lymphoid cells (ILCs) are the innate counterparts of adaptive immune cells. Emerging data indicate that they are also key players in the progression of multiple tumors. In this review we briefly describe ILCs’ functions in the skin, lungs and liver. Next, we analyze the role of ILCs in primary cutaneous melanoma and in its most frequent and deadly metastases, those in liver and lung. We focus on their dual anti– and pro-tumoral functions, depending on the cross-interactions among them and with the surrounding stromal cells that form the tumor microenvironment (TME) in each organ. Next, we detail the role of extracellular vesicles secreted to the TME by ILCs and melanoma on both cell populations. We conclude that the identification of markers and tools to allow the modulation of individual ILC subsets, in addition to the development of standardized protocols, is essential for addressing the therapeutic modulation of ILCs. Abstract The role of innate lymphoid cells (ILCs) in cancer progression has been uncovered in recent years. ILCs are classified as Type 1, Type 2, and Type 3 ILCs, which are characterized by the transcription factors necessary for their development and the cytokines and chemokines they produce. ILCs are a highly heterogeneous cell population, showing both anti– and protumoral properties and capable of adapting their phenotypes and functions depending on the signals they receive from their surrounding environment. ILCs are considered the innate counterparts of the adaptive immune cells during physiological and pathological processes, including cancer, and as such, ILC subsets reflect different types of T cells. In cancer, each ILC subset plays a crucial role, not only in innate immunity but also as regulators of the tumor microenvironment. ILCs’ interplay with other immune and stromal cells in the metastatic microenvironment further dictates and influences this dichotomy, further strengthening the seed-and-soil theory and supporting the formation of more suitable and organ-specific metastatic environments. Here, we review the present knowledge on the different ILC subsets, focusing on their interplay with components of the tumor environment during the development of primary melanoma as well as on metastatic progression to organs, such as the liver or lung.
Collapse
|
16
|
Romayor I, Badiola I, Benedicto A, Márquez J, Herrero A, Arteta B, Olaso E. Silencing of sinusoidal DDR1 reduces murine liver metastasis by colon carcinoma. Sci Rep 2020; 10:18398. [PMID: 33110221 PMCID: PMC7591579 DOI: 10.1038/s41598-020-75395-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022] Open
Abstract
Liver metastasis depends on the collagenous microenvironment generated by hepatic sinusoidal cells (SCs). DDR1 is an atypical collagen receptor linked to tumor progression, but whether SCs express DDR1 and its implication in liver metastasis remain unknown. Freshly isolated hepatic stellate cells (HSCs), Kupffer cells (KCs), and liver sinusoidal endothelial cells (LSECs), that conform the SCs, expressed functional DDR1. HSCs expressed the largest amounts. C26 colon carcinoma secretomes increased DDR1 phosphorylation in HSCs and KCs by collagen I. Inhibition of kinase activity by DDR1-IN-1 or mRNA silencing of DDR1 reduced HSCs secretion of MMP2/9 and chemoattractant and proliferative factors for LSECs and C26 cells. DDR1-IN-1 did not modify MMP2/9 in KCs or LSECs secretomes, but decreased the enhancement of C26 migration and proliferation induced by their secretomes. Gene array showed that DDR1 silencing downregulated HSCs genes for collagens, MMPs, interleukins and chemokines. Silencing of DDR1 before tumor inoculation reduced hepatic C26 metastasis in mice. Silenced livers bore less tumor foci than controls. Metastatic foci in DDR1 silenced mice were smaller and contained an altered stroma with fewer SCs, proliferating cells, collagen and MMPs than foci in control mice. In conclusion, hepatic DDR1 promotes C26 liver metastasis and favors the pro-metastatic response of SCs to the tumor.
Collapse
Affiliation(s)
- Irene Romayor
- Tumor Microenvironment Group, Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, 48940, Leioa, Spain
| | - Iker Badiola
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country, 48940, Leioa, Spain
| | - Aitor Benedicto
- Tumor Microenvironment Group, Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, 48940, Leioa, Spain
| | - Joana Márquez
- Tumor Microenvironment Group, Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, 48940, Leioa, Spain
| | - Alba Herrero
- Tumor Microenvironment Group, Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, 48940, Leioa, Spain
| | - Beatriz Arteta
- Tumor Microenvironment Group, Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, 48940, Leioa, Spain
| | - Elvira Olaso
- Tumor Microenvironment Group, Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, 48940, Leioa, Spain.
| |
Collapse
|
17
|
Targeting Cancer Associated Fibroblasts in Liver Fibrosis and Liver Cancer Using Nanocarriers. Cells 2020; 9:cells9092027. [PMID: 32899119 PMCID: PMC7563527 DOI: 10.3390/cells9092027] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer associated fibroblasts (CAF) and the extracellular matrix (ECM) produced by them have been recognized as key players in cancer biology and emerged as important targets for cancer treatment and drug discovery. Apart from their presence in stroma rich tumors, such as biliary, pancreatic and subtypes of hepatocellular cancer (HCC), both CAF and certain ECM components are also present in cancers without an overt intra-tumoral desmoplastic reaction. They support cancer development, growth, metastasis and resistance to chemo- or checkpoint inhibitor therapy by a multitude of mechanisms, including angiogenesis, ECM remodeling and active immunosuppression by secretion of tumor promoting and immune suppressive cytokines, chemokines and growth factors. CAF resemble activated hepatic stellate cells (HSC)/myofibroblasts, expressing α-smooth muscle actin and especially fibroblast activation protein (FAP). Apart from FAP, CAF also upregulate other functional cell surface proteins like platelet-derived growth factor receptor β (PDGFRβ) or the insulin-like growth factor receptor II (IGFRII). Notably, if formulated with adequate size and zeta potential, injected nanoparticles home preferentially to the liver. Several nanoparticular formulations were tested successfully to deliver dugs to activated HSC/myofibroblasts. Thus, surface modified nanocarriers with a cyclic peptide binding to the PDGFRβ or with mannose-6-phosphate binding to the IGFRII, effectively directed drug delivery to activated HSC/CAF in vivo. Even unguided nanohydrogel particles and lipoplexes loaded with siRNA demonstrated a high in vivo uptake and functional siRNA delivery in activated HSC, indicating that liver CAF/HSC are also addressed specifically by well-devised nanocarriers with optimized physicochemical properties. Therefore, CAF have become an attractive target for the development of stroma-based cancer therapies, especially in the liver.
Collapse
|
18
|
Yufei Z, Yuqi W, Binyue H, Lingchen T, Xi C, Hoffelt D, Fuliang H. Chrysin Inhibits Melanoma Tumor Metastasis via Interfering with the FOXM1/β-Catenin Signaling. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:9358-9367. [PMID: 32797754 DOI: 10.1021/acs.jafc.0c03123] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Melanoma, which features high metastasis and high lethality, is one of the toughest tumors to treat. Chrysin, which is rich in various plants, has shown a great inhibitory effect on melanoma proliferation. Here, we evaluated the metastasis suppressive effect of chrysin on melanoma in vivo and in vitro. In vitro, chrysin effectively inhibited ankios resistance from 5 μM cell migration, invasion from 10 μM, and tube formation capacity of melanoma cells from 20 μM. We discovered that chrysin interfered with the mesenchymal-epithelial transition via regulating FOXM1/β-catenin signaling, as the expression of key regulatory factors was downregulated by chrysin treatment, and overexpression of FOXM1 will attenuate the antimetastasis effect of chrysin. We also tested chrysin on lung colonization in melanoma metastasis, where we found fewer tumors were formed in the lungs of chrysin-treated mice. In addition, the expression of FOXM1 was also downregulated by chrysin in vivo. Collectively, our findings suggested the ability of chrysin treatment to lower the metastatic rate of melanoma through regulating FOXM1/β-catenin signaling, indicating the application potential of chrysin for melanoma therapy.
Collapse
Affiliation(s)
- Zheng Yufei
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- Radiation Oncology Department, University of California, San Francisco, California 94158, United States
| | - Wu Yuqi
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hu Binyue
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tao Lingchen
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chen Xi
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Dixon Hoffelt
- Radiation Oncology Department, University of California, San Francisco, California 94158, United States
| | - Hu Fuliang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
19
|
Tan F, He D, Hu K, Wang D, Zhang S, Li J, Wang Z, Tao Y. WAVE2 Enhanced Hepatic Stellate Cells Activity in Colorectal Liver Metastases. Cancer Manag Res 2020; 12:7671-7680. [PMID: 32904432 PMCID: PMC7455535 DOI: 10.2147/cmar.s259125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/05/2020] [Indexed: 12/11/2022] Open
Abstract
Background Cancer cell migration, tumor angiogenesis, and activated hepatic stellate cells (a-HSCs) promote the development of colorectal liver metastases (CLM). Wiskott–Aldrich syndrome protein family verprolin-homologous protein 2 (WAVE2) has been associated with CLM, although the underlying molecular mechanisms remain unclear. Methods In the current study, we evaluated the relationship between WAVE2 and CLM in 103 CLM patients who underwent liver resection. Immunohistochemistry (IHC) staining was performed to determine the association between WAVE2 protein expression and hepatic micro-metastasis in human CLM tissues. WAVE2 knockout was performed in hepatic stellate cells (HSC) to explore the function and signaling pathways of WAVE2 in colorectal cancer progression. Results Significantly higher levels of WAVE2 were detected in portal-associated relative to sinusoid-associated micro-metastasis. A strong correlation was identified between WAVE2 levels and microvessel density (MVD) in hepatic metastasis. Similarly, expression of WAVE2 was closely associated with activation of HSCs. Mechanistically, WAVE2 regulated the progression of human CLM acts by regulating the growth factor β (TGF-β) and Hippo pathways via effector yes-associated protein (YAP1). Conclusion Overall, our results demonstrated that WAVE2 participates in CLM tumor microenvironment, and can be a potential latent therapeutic target for CLM.
Collapse
Affiliation(s)
- Fengbo Tan
- Department of General Surgery Research, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Dongren He
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Kuan Hu
- Department of General Surgery Research, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Dong Wang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Sai Zhang
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Juanni Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Zhiming Wang
- Department of General Surgery Research, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Yiming Tao
- Department of General Surgery Research, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
20
|
Gerovska D, Larrinaga G, Solano-Iturri JD, Márquez J, García Gallastegi P, Khatib AM, Poschmann G, Stühler K, Armesto M, Lawrie CH, Badiola I, Araúzo-Bravo MJ. An Integrative Omics Approach Reveals Involvement of BRCA1 in Hepatic Metastatic Progression of Colorectal Cancer. Cancers (Basel) 2020; 12:E2380. [PMID: 32842712 PMCID: PMC7565528 DOI: 10.3390/cancers12092380] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/26/2022] Open
Abstract
(1) Background & Aims: The roles of different cells in the tumor microenvironment (TME) are critical to the metastatic process. The phenotypic transformation of the liver cells is one of the most important stages of the hepatic metastasis progression of colorectal cancer (CRC). Our aim was to identify the major molecules (i.e., genes, miRNAs and proteins) involved in this process. (2) Methods: We isolated and performed whole-genome analysis of gene, miRNA, and protein expression in three types of liver cells (Ito cells, Kupffer cells, and liver sinusoidal endothelial cells) from the TME of a murine model of CRC liver metastasis. We selected the statistically significant differentially expressed molecules using the Student's t-test with Benjamini-Hochberg correction and performed functional statistically-significant enrichment analysis of differentially expressed molecules with hypergeometric distribution using the curated collection of molecular signatures, MSigDB. To build a gene-miRNA-protein network centered in Brca1, we developed a software package (miRDiana) that collects miRNA targets from the union of the TargetScan, MicroCosm, mirTarBase, and miRWalk databases. This was used to search for miRNAs targeting Brca1. We validated the most relevant miRNAs with real-time quantitative PCR. To investigate BRCA1 protein expression, we built tissue microarrays (TMAs) from hepatic metastases of 34 CRC patients. (3) Results: Using integrated omics analyses, we observed that the Brca1 gene is among the twenty transcripts simultaneously up-regulated in all three types of TME liver cells during metastasis. Further analysis revealed that Brca1 is the last BRCA1-associated genome surveillance complex (BASC) gene activated in the TME. We confirmed this finding in human reanalyzing transcriptomics datasets from 184 patients from non-tumor colorectal tissue, primary colorectal tumor and colorectal liver metastasis of the GEO database. We found that the most probable sequence of cell activation during metastasis is Endothelial→Ito→Kupffer. Immunohistochemical analysis of human liver metastases showed the BRCA1 protein was co-localized in Ito, Kupffer, and endothelial cells in 81.8% of early or synchronous metastases. However, in the greater part of the metachronous liver metastases, this protein was not expressed in any of these TME cells. (4) Conclusions: These results suggest a possible role of the co-expression of BRCA1 in Ito, Kupffer, and sinusoidal endothelial cells in the early occurrence of CRC liver metastases, and point to BRCA1 as a potential TME biomarker.
Collapse
Affiliation(s)
- Daniela Gerovska
- Computational Biology and Systems Biomedicine Group, Biodonostia Health Research Institute, Calle Doctor Beguiristain s/n, 20014 San Sebastián, Spain;
- Computational Biomedicine Data Analysis Platform, Biodonostia Health Research Institute, Calle Doctor Beguiristain s/n, 20014 San Sebastián, Spain
| | - Gorka Larrinaga
- Department of Nursing I, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Bizkaia, Spain;
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Bizkaia, Spain
- BioCruces Health Research Institute, 48903 Barakaldo, Bizkaia, Spain;
| | - Jon Danel Solano-Iturri
- BioCruces Health Research Institute, 48903 Barakaldo, Bizkaia, Spain;
- Department of Anatomic Pathology, Cruces University Hospital, University of the Basque Country (UPV/EHU), 48903 Barakaldo, Bizkaia, Spain
| | - Joana Márquez
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), 48940 Leioa, Spain; (J.M.); (P.G.G.)
| | - Patricia García Gallastegi
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), 48940 Leioa, Spain; (J.M.); (P.G.G.)
| | - Abdel-Majid Khatib
- University of Bordeaux, Allée Geoffroy St Hilaire, 33615 Pessac, France; INSERM, LAMC, UMR 1029, Allée Geoffroy St Hilaire, 33615 Pessac, France;
| | - Gereon Poschmann
- Institute of Molecular Medicine, Proteome Research, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (G.P.); (K.S.)
| | - Kai Stühler
- Institute of Molecular Medicine, Proteome Research, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (G.P.); (K.S.)
- Molecular Proteomics Laboratory, Biologisch-Medizinisches Forschungszentrum, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - María Armesto
- Molecular Oncology Group, Biodonostia Health Research Institute, 20014 San Sebastián, Spain; (M.A.); (C.H.L.)
| | - Charles H. Lawrie
- Molecular Oncology Group, Biodonostia Health Research Institute, 20014 San Sebastián, Spain; (M.A.); (C.H.L.)
- Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- IKERBASQUE, Basque Foundation for Science, Calle María Díaz Harokoa 3, 48013 Bilbao, Spain
| | - Iker Badiola
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), 48940 Leioa, Spain; (J.M.); (P.G.G.)
| | - Marcos J. Araúzo-Bravo
- Computational Biology and Systems Biomedicine Group, Biodonostia Health Research Institute, Calle Doctor Beguiristain s/n, 20014 San Sebastián, Spain;
- Computational Biomedicine Data Analysis Platform, Biodonostia Health Research Institute, Calle Doctor Beguiristain s/n, 20014 San Sebastián, Spain
- IKERBASQUE, Basque Foundation for Science, Calle María Díaz Harokoa 3, 48013 Bilbao, Spain
- CIBER of Frailty and Healthy Aging (CIBERfes), 28029 Madrid, Spain
- Computational Biology and Bioinformatics Group, Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149 Münster, Germany
| |
Collapse
|
21
|
Vidal-Vanaclocha F, Crende O, García de Durango C, Herreros-Pomares A, López-Doménech S, González Á, Ruiz-Casares E, Vilboux T, Caruso R, Durán H, Gil A, Ielpo B, Lapuente F, Quijano Y, Vicente E, Vidal-Lartitegui L, Sotomayor EM. Liver prometastatic reaction: Stimulating factors and responsive cancer phenotypes. Semin Cancer Biol 2020; 71:122-133. [PMID: 32805395 DOI: 10.1016/j.semcancer.2020.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/04/2020] [Indexed: 02/07/2023]
Abstract
Cancer is first a localized tissue disorder, whose soluble and exosomal molecules and invasive cells induce a host response providing the stromal components of the primary tumor microenvironment (TME). Once the TME is developed, cancer-derived molecules and cells can more efficiently spread out and a whole-body response takes place, whose pathophysiological changes may result in a paraneoplastic syndrome. Remote organ-specific prometastatic reactions may also occur at this time, facilitating metastatic activities of circulating tumor cells (CTCs) through premetastatic niche development at targeted organs. However, additional signaling factors from the inter-organ communication network involved in the pathophysiology and comorbidities of cancer patients may also regulate prometastatic reaction-stimulating effects of cancer and non-cancer tissue factors. This article provides a conceptual overview of our ongoing clinical research on the liver prometastatic reaction (LPR) of patients with colorectal cancer (CRC), their portal vein- and hepatic artery-driven LPR-Stimulating Factors (LPR-SF), and their resulting LPR-derived Metastasis-Stimulating Factors (LPR-MSF) acting on liver-invading CRC cells. In addition, we also provide new insights on the molecular subtyping of LPR-responsive cancer phenotypes in patients with CRC and melanoma; and on how to investigate and interpret the prometastatic infrastructure in the real pathophysiological context of patients with cancer undergoing surgical procedures and receiving pharmacological treatments with multiple side effects, including those affecting the LPR, its stimulating factors and responsive cancer phenotypes.
Collapse
Affiliation(s)
- Fernando Vidal-Vanaclocha
- Dept. Biochemistry and Molecular Medicine, GW Cancer Center, School of Medicine and Health Sciences, George Washington University, Washington, DC, USA; Institute of Applied Molecular Medicine (IMMA), CEU-San Pablo University School of Medicine, Boadilla del Monte, Madrid, Spain; Persona Biomed Inc., Alexandria, Virginia, USA.
| | - Olatz Crende
- Dept Cell Biology and Histology, Basque Country University School of Pharmacy, Vitoria/Gasteiz, Spain
| | | | | | | | - Álvaro González
- Institute of Applied Molecular Medicine (IMMA), CEU-San Pablo University School of Medicine, Boadilla del Monte, Madrid, Spain
| | - Eva Ruiz-Casares
- Institute of Applied Molecular Medicine (IMMA), CEU-San Pablo University School of Medicine, Boadilla del Monte, Madrid, Spain
| | | | - Riccardo Caruso
- Division of General Surgery, HM-Sanchinarro University Hospital, CEU San Pablo University, Madrid, Spain
| | - Hipólito Durán
- Division of General Surgery, HM-Sanchinarro University Hospital, CEU San Pablo University, Madrid, Spain
| | - Antonio Gil
- Division of General Surgery, HM-Sanchinarro University Hospital, CEU San Pablo University, Madrid, Spain
| | - Benedetto Ielpo
- Division of General Surgery, HM-Sanchinarro University Hospital, CEU San Pablo University, Madrid, Spain
| | - Fernando Lapuente
- Department General Surgery, Bariatric and Metabolic Surgery, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Yolanda Quijano
- Division of General Surgery, HM-Sanchinarro University Hospital, CEU San Pablo University, Madrid, Spain
| | - Emilio Vicente
- Division of General Surgery, HM-Sanchinarro University Hospital, CEU San Pablo University, Madrid, Spain
| | | | - Eduardo M Sotomayor
- Department of Hematology and Oncology, George Washington University, Washington, DC, USA
| |
Collapse
|
22
|
Roife D, Sarcar B, Fleming JB. Stellate Cells in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1263:67-84. [PMID: 32588324 DOI: 10.1007/978-3-030-44518-8_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
As tumor microenvironments share many of the same qualities as chronic wounds, attention is turning to the wound-repair cells that support the growth of cancerous cells. Stellate cells are star-shaped cells that were first discovered in the perisinusoidal spaces in the liver and have been found to support wound healing by the secretion of growth factors and extracellular matrix. They have since been also found to serve a similar function in the pancreas. In both organs, the wound-healing process may become dysregulated and lead to pathological fibrosis (also known as cirrhosis in the liver). In recent years there has been increasing attention paid to the role of these cells in tumor formation and progression. They may be a factor in initiating the first steps of carcinogenesis such as with liver cirrhosis and hepatocellular carcinoma and also contribute to continued tumor growth, invasion, metastasis, evasion of the immune system, and resistance to chemotherapy, in cancers of both the liver and pancreas. In this chapter we aim to review the structure and function of hepatic and pancreatic stellate cells and their contributions to the tumor microenvironment in their respective cancers and also discuss potential new targets for cancer therapy based on our new understanding of these vital components of the tumor stroma.
Collapse
Affiliation(s)
- David Roife
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA.,Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Bhaswati Sarcar
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Jason B Fleming
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA.
| |
Collapse
|
23
|
Abstract
The liver is the largest organ in the human body and is prone for cancer metastasis. Although the metastatic pattern can differ depending on the cancer type, the liver is the organ to which cancer cells most frequently metastasize for the majority of prevalent malignancies. The liver is unique in several aspects: the vascular structure is highly permeable and has unparalleled dual blood connectivity, and the hepatic tissue microenvironment presents a natural soil for the seeding of disseminated tumor cells. Although 70% of the liver is composed of the parenchymal hepatocytes, the remaining 30% is composed of nonparenchymal cells including Kupffer cells, liver sinusoidal endothelial cells, and hepatic stellate cells. Recent discoveries show that both the parenchymal and the nonparenchymal cells can modulate each step of the hepatic metastatic cascade, including the initial seeding and colonization as well as the decision to undergo dormancy versus outgrowth. Thus, a better understanding of the molecular mechanisms orchestrating the formation of a hospitable hepatic metastatic niche and the identification of the drivers supporting this process is critical for the development of better therapies to stop or at least decrease liver metastasis. The focus of this perspective is on the bidirectional interactions between the disseminated cancer cells and the unique hepatic metastatic niche.
Collapse
Affiliation(s)
- Ainhoa Mielgo
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, L69 3GE, United Kingdom
| | - Michael C Schmid
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, L69 3GE, United Kingdom
| |
Collapse
|
24
|
Shiraha H, Iwamuro M, Okada H. Hepatic Stellate Cells in Liver Tumor. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1234:43-56. [PMID: 32040854 DOI: 10.1007/978-3-030-37184-5_4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma and intrahepatic cholangiocarcinoma are the most common types of primary liver cancers. Moreover, the liver is the second most frequently involved organ in cancer metastasis after lymph nodes. The tumor microenvironment is crucial for the development of both primary and secondary liver cancers. The hepatic microenvironment consists of multiple cell types, including liver sinusoidal endothelial cells, Kupffer cells, natural killer cells, liver-associated lymphocytes, and hepatic stellate cells (HSCs). The microenvironment of a normal liver changes to a tumor microenvironment when tumor cells exist or tumor cells migrate to and multiply in the liver. Interactions between tumor cells and non-transformed cells generate a tumor microenvironment that contributes significantly to tumor progression. HSCs play a central role in the tumor microenvironment crosstalk. As this crosstalk is crucial for liver carcinogenesis and liver-tumor development, elucidating the mechanism underlying the interaction of HSCs with the tumor microenvironment could provide potential therapeutic targets for liver cancer.
Collapse
Affiliation(s)
- Hidenori Shiraha
- Department of Gastroenterology and Hepatology, Okayama University Faculty of Medicine, Okayama, Japan.
| | - Masaya Iwamuro
- Department of Gastroenterology and Hepatology, Okayama University Faculty of Medicine, Okayama, Japan
| | - Hiroyuki Okada
- Department of Gastroenterology and Hepatology, Okayama University Faculty of Medicine, Okayama, Japan
| |
Collapse
|
25
|
Lin N, Meng L, Lin J, Chen S, Zhang P, Chen Q, Lin Y. Activated hepatic stellate cells promote angiogenesis in hepatocellular carcinoma by secreting angiopoietin-1. J Cell Biochem 2019; 121:1441-1451. [PMID: 31609020 DOI: 10.1002/jcb.29380] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 08/28/2019] [Indexed: 12/29/2022]
Abstract
Angiogenesis is the central pathological process in hepatocellular carcinoma (HCC), and its progression is affected by tumor cells and the microenvironment. Activated hepatic stellate cells (aHSCs) are the most significant stromal cells involved in HCC. This study was aimed to explore the effects and mechanisms of aHSCs on angiogenesis in HCC. We isolated primary hepatoma cells, aHSCs, and hepatic vascular endothelial cells from human HCC samples. Then, we performed a novel in vitro assay and in vivo experiment in a nude mouse HCC model to investigate the functions of aHSCs on angiogenesis in HCC. Our results demonstrated that aHSCs are the primary sources of angiopoietin-1 (Ang-1) in human HCC in vitro, and aHSCs could promote hepatic vascular endothelial cell (HVEC) growth by secreting Ang-1. Furthermore, aHSCs could induce HVEC microtubule formation, and this ability was reduced when Ang-1 expression was silenced in aHSCs. In addition, CD34 expression in a nude mouse HCC model was downregulated when Ang-1 messenger RNA was silenced in aHSCs. Our data also indicated that HSC Ang-1 expression could be inhibited by overexpressing Raf kinase inhibitor protein. Therefore, we suggest that aHSCs promote angiogenesis through secreting Ang-1, potentially providing an interesting target for antiangiogenic therapies for HCC.
Collapse
Affiliation(s)
- Nan Lin
- Department of Hepatobiliary Surgery, The People's Hospital of Kashgar, Kashgar, Xinjiang, China.,Xinjiang Medical University, Ürümqi, Xinjiang, China.,Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lili Meng
- Department of Gynecology and Obstetrics, The Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jizong Lin
- Department of General Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shuxian Chen
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Peng Zhang
- Department of General Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qilong Chen
- Xinjiang Medical University, Ürümqi, Xinjiang, China
| | - Yang Lin
- Department of Hepatobiliary Surgery, The People's Hospital of Kashgar, Kashgar, Xinjiang, China
| |
Collapse
|
26
|
Benedicto A, Herrero A, Romayor I, Marquez J, Smedsrød B, Olaso E, Arteta B. Liver sinusoidal endothelial cell ICAM-1 mediated tumor/endothelial crosstalk drives the development of liver metastasis by initiating inflammatory and angiogenic responses. Sci Rep 2019; 9:13111. [PMID: 31511625 PMCID: PMC6739321 DOI: 10.1038/s41598-019-49473-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 08/23/2019] [Indexed: 12/16/2022] Open
Abstract
The prometastatic stroma generated through tumor cells/host cells interaction is critical for metastatic growth. To elucidate the role of ICAM-1 on the crosstalk between tumor and primary liver sinusoidal endothelial cells (LSECs) and hepatic stellate cells (HSCs), implicated in tumor adhesion and angiogenesis, we performed in vitro cocultures and an in vivo model of liver metastasis of colorectal cancer (CRC). ICAM-1 blockade in the LSECs decreased the adhesion and transmigration of tumor cells through an LSEC in vitro and vivo. Cocultures of C26 cells and LSECs contained higher amounts of IL-1β, IL-6, PGE-2, TNF-α and ICAM-1 than monocultures. C26 cells incubated with sICAM-1 secreted higher amounts of PGE-2, IL-6, VEGF, and MMPs, while enhanced the migration of LSECs and HSCs. HSCs cultures activated by media from C26 cells pretreated with sICAM-1 contained the largest amounts of VEGF and MMPs. C26 cell activation with sICAM-1 enhanced their metastasizing potential in vivo, while tumor LFA-1 blockade reduced tumor burden and LSECs and HSC-derived myofibroblasts recruitment. In vivo ICAM-1 silencing produced similar results. These findings uncover LSEC ICAM-1 as a mediator of the CRC metastatic cascade in the liver and identifies it as target for the inhibition of liver colonization and metastatic progression.
Collapse
Affiliation(s)
- Aitor Benedicto
- Department of Cellular Biology and Histology, University of the Basque Country, School of Medicine and Nursing, 48940, Leioa, Bizkaia, Spain.
| | - Alba Herrero
- Department of Cellular Biology and Histology, University of the Basque Country, School of Medicine and Nursing, 48940, Leioa, Bizkaia, Spain
| | - Irene Romayor
- Department of Cellular Biology and Histology, University of the Basque Country, School of Medicine and Nursing, 48940, Leioa, Bizkaia, Spain
| | - Joana Marquez
- Department of Cellular Biology and Histology, University of the Basque Country, School of Medicine and Nursing, 48940, Leioa, Bizkaia, Spain
| | - Bård Smedsrød
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø, Tromsø, Norway
| | - Elvira Olaso
- Department of Cellular Biology and Histology, University of the Basque Country, School of Medicine and Nursing, 48940, Leioa, Bizkaia, Spain
| | - Beatriz Arteta
- Department of Cellular Biology and Histology, University of the Basque Country, School of Medicine and Nursing, 48940, Leioa, Bizkaia, Spain
| |
Collapse
|
27
|
Li Z, Guo T, Fang L, Li N, Wang X, Wang P, Zhao S, Li F, Cui Y, Shu X, Zhao L, Li J, Gu C. MACC1 overexpression in carcinoma‑associated fibroblasts induces the invasion of lung adenocarcinoma cells via paracrine signaling. Int J Oncol 2019; 54:1367-1375. [PMID: 30720137 DOI: 10.3892/ijo.2019.4702] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 11/26/2018] [Indexed: 11/05/2022] Open
Abstract
Carcinoma‑associated fibroblasts (CAFs) are essential for initiating lung cancer cell invasion and metastasis. An elevated MACC1 expression has been implicated in the progression of lung adenocarcinoma. Hitherto, the role of MACC1 in lung adenocarcinoma‑derived CAFs remains unclear. In this study, CAFs isolated from the tissues of patients with lung adenocarcinoma expressed typical CAF markers (shown by immunohistochemical and immunofluorescence analysis) and exhibited enhanced migration and invasion abilities when co‑cultured with A549 cells in a microfluidic model. MACC1‑overexpressing CAFs not only demonstrated an increased invasion, but also exerted a promoting effect on the invasion of tumor cells. The reduced expression of MACC1 impaired the invasive ability of the CAFs. Western blot analysis and RT‑qPCR analysis demonstrated that multiple paracrine pathways were activated in the MACC1‑overexpressing CAFs. Overall, this study presents a novel role of MACC1 in CAF‑induced lung adenocarcinoma cell invasion, which possibly occurs via paracrine signaling. Furthermore, MACC1 was indicated to be a potential therapeutic target for lung adenocarcinoma.
Collapse
Affiliation(s)
- Zhuoshi Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Tao Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Lei Fang
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Nan Li
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Xiaochao Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Peng Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Shilei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Fengzhou Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Yanwei Cui
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Xin Shu
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Lei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Jinxiu Li
- Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Chundong Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| |
Collapse
|
28
|
Qiu X, Chen D, Liu Y, Duan S, Zhang F, Zhang Y, Li F, Chen C, Chen Y. Relationship between stromal cells and tumor spread through air spaces in lung adenocarcinoma. Thorac Cancer 2019; 10:256-267. [PMID: 30605235 PMCID: PMC6360243 DOI: 10.1111/1759-7714.12945] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 12/28/2022] Open
Abstract
Background The mechanism underlying tumor spread through air spaces (STAS) has not been well studied. We investigated the role of tumor stromal cells in the pathogenesis of STAS from a pathological perspective and evaluated the prognostic significance of tumor stromal cells and STAS in postoperative patients with lung adenocarcinoma. Methods We retrospectively analyzed 208 postsurgical patients with stage I–IIIA lung adenocarcinoma. The presence of STAS was evaluated by hematoxylin and eosin staining. The expression of α‐smooth muscle actin (SMA)‐positive cancer‐associated fibroblasts (CAFs) and CD204‐positive tumor‐associated macrophages (TAMs) was analyzed by immunohistochemistry. A logistic regression model was applied to confirm the predictive factors of STAS. Survival analysis was performed to evaluate the effect of α‐SMA‐positive CAFs, CD204‐positive TAMs, and STAS on prognosis. A nomogram was generated to evaluate the prognosis of postoperative patients. Results Logistic regression suggested that the expression of α‐SMA‐positive CAFs (P < 0.001) and the number of CD204‐positive TAMs (P < 0.001) were related to the presence of STAS. The multivariate Cox proportional hazards model suggested that STAS (P = 0.004), α‐SMA‐positive CAFs (P < 0.001), and CD204‐positive TAMs (P < 0.001) were independent risk factors for prognosis. Harrell's c‐indexes for overall and recurrence‐free survival prediction based on nomograms were 0.84 (95% confidence interval 0.76–0.91) and 0.82 (95% confidence interval 0.76–0.89), respectively. Conclusions The presence of STAS was associated with high expression of α‐SMA and CD204 in lung adenocarcinoma. Nomograms including STAS and stromal cells as variables are recommended as practical models to evaluate the prognosis of lung adenocarcinoma patients.
Collapse
Affiliation(s)
- Xie Qiu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Department of Thoracic Surgery, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Donglai Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yangyang Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Shanzhou Duan
- Department of Thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Fuquan Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yongsheng Zhang
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Feng Li
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yongbing Chen
- Department of Thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
29
|
Schmidt KM, Dietrich P, Hackl C, Guenzle J, Bronsert P, Wagner C, Fichtner-Feigl S, Schlitt HJ, Geissler EK, Hellerbrand C, Lang SA. Inhibition of mTORC2/RICTOR Impairs Melanoma Hepatic Metastasis. Neoplasia 2018; 20:1198-1208. [PMID: 30404068 PMCID: PMC6224335 DOI: 10.1016/j.neo.2018.10.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/09/2018] [Accepted: 10/11/2018] [Indexed: 02/07/2023] Open
Abstract
Mammalian target of rapamycin complex 2 (mTORC2) with its pivotal component rapamycin-insensitive companion of mTOR (RICTOR) is the major regulator of AKT phosphorylation and is increasingly implicated in tumor growth and progression. In cutaneous melanoma, an extremely aggressive and highly metastatic disease, RICTOR overexpression is involved in tumor development and invasiveness. Therefore, we investigated the impact of RICTOR inhibition in melanoma cells in vitro and in vivo with special emphasis on hepatic metastasis. Moreover, our study focused on the interaction of tumor cells and hepatic stellate cells (HSC) which play a crucial role in the hepatic microenvironment. In silico analysis revealed increased RICTOR expression in melanoma cells and tissues and indicated higher expression in advanced melanoma stages and metastases. In vitro, transient RICTOR knock-down via siRNA caused a significant reduction of tumor cell motility. Using a syngeneic murine splenic injection model, a significant decrease in liver metastasis burden was detected in vivo. Moreover, stimulation of melanoma cells with conditioned medium (CM) from activated HSC or hepatocyte growth factor (HGF) led to a significant induction of AKT phosphorylation and tumor cell motility. Blocking of RICTOR expression in cancer cells diminished constitutive and HGF-induced AKT phosphorylation as well as cell motility. Interestingly, RICTOR blockade also led to an abrogation of CM-induced effects on AKT phosphorylation and motility in melanoma cells. In conclusion, these results provide first evidence for a critical role of mTORC2/RICTOR in melanoma liver metastasis via cancer cell/HSC interactions.
Collapse
Affiliation(s)
- Katharina M Schmidt
- Department of Surgery, Regensburg University Hospital, Franz-Josef-Strauss Allee 9, Regensburg, Germany.
| | - Peter Dietrich
- Institute of Biochemistry, Friedrich-Alexander University Erlangen-Nürnberg, Fahrstrasse 17, Germany; Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany.
| | - Christina Hackl
- Department of Surgery, Regensburg University Hospital, Franz-Josef-Strauss Allee 9, Regensburg, Germany.
| | - Jessica Guenzle
- Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, Germany.
| | - Peter Bronsert
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Strasse 115a, Germany; Tumorbank Comprehensive Cancer Center Freiburg, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Strasse 115a, Germany.
| | - Christine Wagner
- Department of Surgery, Regensburg University Hospital, Franz-Josef-Strauss Allee 9, Regensburg, Germany.
| | - Stefan Fichtner-Feigl
- Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, Germany.
| | - Hans J Schlitt
- Department of Surgery, Regensburg University Hospital, Franz-Josef-Strauss Allee 9, Regensburg, Germany.
| | - Edward K Geissler
- Department of Surgery, Regensburg University Hospital, Franz-Josef-Strauss Allee 9, Regensburg, Germany.
| | - Claus Hellerbrand
- Institute of Biochemistry, Friedrich-Alexander University Erlangen-Nürnberg, Fahrstrasse 17, Germany.
| | - Sven A Lang
- Department of Surgery, Regensburg University Hospital, Franz-Josef-Strauss Allee 9, Regensburg, Germany; Department of General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, Germany.
| |
Collapse
|
30
|
Abdul-Wahid A, Cydzik M, Fischer NW, Prodeus A, Shively JE, Martel A, Alminawi S, Ghorab Z, Berinstein NL, Gariépy J. Serum-derived carcinoembryonic antigen (CEA) activates fibroblasts to induce a local re-modeling of the extracellular matrix that favors the engraftment of CEA-expressing tumor cells. Int J Cancer 2018; 143:1963-1977. [PMID: 29756328 DOI: 10.1002/ijc.31586] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 04/04/2018] [Accepted: 04/18/2018] [Indexed: 12/21/2022]
Abstract
Elevated levels of the carcinoembryonic antigen (CEA; CEACAM5) in the serum of colorectal cancer (CRC) patients represent a clinical biomarker that correlates with disease recurrence. However, a mechanistic role for soluble CEA (sCEA) in tumor progression and metastasis remains to be established. In our study, we report that sCEA acts as a paracrine factor, activating human fibroblasts by signaling through both the STAT3 and AKT1-mTORC1 pathways, promoting their transition to a cancer-associated fibroblast (CaF) phenotype. sCEA-activated fibroblasts express and secrete higher levels of fibronectin, including cellular EDA+ -fibronectin (Fn-EDA) that selectively promote the implantation and adherence of CEA-expressing cancer cells. Immunohistochemical analyses of liver tissues derived from CRC patients with elevated levels of sCEA reveal that the expression of cellular Fn-EDA co-registers with CEA-expressing liver metastases. Taken together, these findings indicate a direct role for sCEA as a human fibroblast activation factor, in priming target tissues for the engraftment of CEA-expressing cancer cells, through the differentiation of tissue-resident fibroblasts, resulting in a local change in composition of the extracellular matrix.
Collapse
Affiliation(s)
- Aws Abdul-Wahid
- Departments of Medical Biophysics and Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Marzena Cydzik
- Departments of Medical Biophysics and Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Nicholas W Fischer
- Departments of Medical Biophysics and Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Aaron Prodeus
- Departments of Medical Biophysics and Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - John E Shively
- Department of Immunology, Beckman Research Institute, City of Hope, Duarte, CA
| | - Anne Martel
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Samira Alminawi
- Department of Anatomic Pathology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, CANADA
| | - Zeina Ghorab
- Department of Anatomic Pathology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, CANADA
| | | | - Jean Gariépy
- Departments of Medical Biophysics and Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|
31
|
Marquez J, Fernandez-Piñeiro I, Araúzo-Bravo MJ, Poschmann G, Stühler K, Khatib AM, Sanchez A, Unda F, Ibarretxe G, Bernales I, Badiola I. Targeting liver sinusoidal endothelial cells with miR-20a-loaded nanoparticles reduces murine colon cancer metastasis to the liver. Int J Cancer 2018; 143:709-719. [PMID: 29492958 DOI: 10.1002/ijc.31343] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 02/02/2018] [Accepted: 02/19/2018] [Indexed: 12/15/2022]
Abstract
Phenotypic transformation of liver sinusoidal endothelial cells is one of the most important stages of liver metastasis progression. The miRNA effects on liver sinusoidal endothelial cells during liver metastasis have not yet been studied. Herein, whole genome analysis of miRNA expression in these cells during colorectal liver metastasis revealed repressed expression of microRNA-20a. Importantly, downregulation of miR-20a occurs in parallel with upregulation of its known protein targets. To restore normal miR-20a levels in liver sinusoidal endothelial cells, we developed chondroitin sulfate-sorbitan ester nanoparticles conjugated with miR-20a in a delivery system that specifically targets liver sinusoidal endothelial cells. The restoration of normal mir-20a levels in these cells induced downregulation of the expression of its protein targets, and this also resulted in a reduction of in vitro LSEC migration and a reduction of in vivo activation and tumor-infiltrating capacity and ability of the tumor decreased by ∼80% in a murine liver metastasis model.
Collapse
Affiliation(s)
- Joana Marquez
- Department of Cell Biology and Histology, Faculty of Medicine and Nursery, University of Basque Country, UPV/EHU, Leioa, Spain
| | - Ines Fernandez-Piñeiro
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Marcos J Araúzo-Bravo
- Computational Biology and Systems Biomedicine Research Group.Computational Biology Data Analysis Platform. Biodonostia Health Research Institute, San Sebastián, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Gereon Poschmann
- Molecular Proteomics Laboratory (MPL), Biologisch-Medizinisches Forschungszentrum (BMFZ),Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory (MPL), Biologisch-Medizinisches Forschungszentrum (BMFZ),Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Abdel-Majid Khatib
- Université Bordeaux, Pessac, France.,INSERM, LAMC, UMR 1029, Pessac, France
| | - Alejandro Sanchez
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, Spain.,Genetics and Biology of the Development of Kidney Diseases Unit, Sanitary Research Institute (IDIS) of the University Hospital Complex of Santiago de Compostela, Santiago de Compostela, Spain
| | - Fernando Unda
- Department of Cell Biology and Histology, Faculty of Medicine and Nursery, University of Basque Country, UPV/EHU, Leioa, Spain
| | - Gaskon Ibarretxe
- Department of Cell Biology and Histology, Faculty of Medicine and Nursery, University of Basque Country, UPV/EHU, Leioa, Spain
| | - Irantzu Bernales
- Gene Expression Unit, Genomics Facility of General Research Services (SGIker), University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Iker Badiola
- Department of Cell Biology and Histology, Faculty of Medicine and Nursery, University of Basque Country, UPV/EHU, Leioa, Spain
| |
Collapse
|
32
|
Wang X, Hassan W, Jabeen Q, Khan GJ, Iqbal F. Interdependent and independent multidimensional role of tumor microenvironment on hepatocellular carcinoma. Cytokine 2017; 103:150-159. [PMID: 29029799 DOI: 10.1016/j.cyto.2017.09.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 09/03/2017] [Accepted: 09/22/2017] [Indexed: 02/07/2023]
Abstract
The novelty of an effective therapeutic targeting for hepatocellular carcinoma (HCC) is based on improved understanding of each component of tumor microenvironment (TME) and its correspondent interactions at biological and molecular levels. In this context, new expansions for the treatment against TME and its communication with HCC are under exploration. Despite of the fact that blockage of growth factor receptors has become a treatment of choice in late phases of HCC in clinical practice, still a precise targeted treatment should address all the components of TME. Targeting one specific element out of cellular (cancer associated fibroblasts, endothelial cells, hepatic stellate cells, Kupffer cells and lymphocytes) or non-cellular (extracellular matrix, growth factors, inflammatory cytokines, proteolytic enzymes) parts of TME may not be a successful remedy for the disease because of well-designed hindrances of each component and their functional alternativeness. Meanwhile there are some elements of TME like epithelial-mesenchymal transition and CAF, which are considerably important and need thorough investigations. Ascertaining the potential role of these elements, and a single or combinational drug therapy targeting these elements of TME simultaneously, may provide the appreciable considerations to eventually improve in clinical practices and may also minimize the chances of reoccurrence of HCC.
Collapse
Affiliation(s)
- Xue Wang
- Jiangnan University, Wuxi Medical School, Wuxi 214122, China; China Pharmaceutical University, Department of Pharmacology, Nanjing 210009, China.
| | - Waseem Hassan
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan; Department of Pharmacy, The University of Lahore, Pakistan.
| | - Qaiser Jabeen
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan.
| | - Ghulam Jilany Khan
- China Pharmaceutical University, Department of Pharmacology, Nanjing 210009, China.
| | - Furqan Iqbal
- Department of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan.
| |
Collapse
|
33
|
Benedicto A, Romayor I, Arteta B. Role of liver ICAM-1 in metastasis. Oncol Lett 2017; 14:3883-3892. [PMID: 28943897 DOI: 10.3892/ol.2017.6700] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 07/07/2017] [Indexed: 12/15/2022] Open
Abstract
Intercellular adhesion molecule (ICAM)-1, is a transmembrane glycoprotein of the immunoglobulin (Ig)-like superfamily, consisting of five extracellular Ig-like domains, a transmembrane domain and a short cytoplasmic tail. ICAM-1 is expressed in various cell types, including endothelial cells and leukocytes, and is involved in several physiological processes. Furthermore, it has additionally been reported to be expressed in various cancer cells, including melanoma, colorectal cancer and lymphoma. The majority of studies to date have focused on the expression of the ICAM-1 on the surface of tumor cells, without research into ICAM-1 expression at sites of metastasis. Cancer cells frequently metastasize to the liver, due to its unique physiology and specialized liver sinusoid capillary network. Liver sinusoidal endothelial cells constitutively express ICAM-1, which is upregulated under inflammatory conditions. Furthermore, liver ICAM-1 may be important during the development of liver metastasis. Therefore, it is necessary to improve the understanding of the mechanisms mediated by this adhesion molecule in order to develop host-directed anticancer therapies.
Collapse
Affiliation(s)
- Aitor Benedicto
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of The Basque Country, UPV/EHU, Leioa, E-48940 Vizcaya, Spain
| | - Irene Romayor
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of The Basque Country, UPV/EHU, Leioa, E-48940 Vizcaya, Spain
| | - Beatriz Arteta
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of The Basque Country, UPV/EHU, Leioa, E-48940 Vizcaya, Spain
| |
Collapse
|
34
|
Meyer T, Koch A, Ebert EV, Czech B, Mueller M, Bosserhoff A, Lang SA, Hellerbrand C. Effect of melanoma cells on proliferation and migration of activated hepatic stellate cells in vitro. Pathol Res Pract 2017; 213:400-404. [DOI: 10.1016/j.prp.2016.12.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 12/16/2016] [Indexed: 12/28/2022]
|
35
|
Xu Y, Zhao W, Xu J, Li J, Hong Z, Yin Z, Wang X. Activated hepatic stellate cells promote liver cancer by induction of myeloid-derived suppressor cells through cyclooxygenase-2. Oncotarget 2017; 7:8866-78. [PMID: 26758420 PMCID: PMC4891010 DOI: 10.18632/oncotarget.6839] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 12/12/2015] [Indexed: 01/01/2023] Open
Abstract
Hepatic stellate cells (HSCs) are critical mediators of immunosuppression and the pathogenesis of hepatocellular carcinoma (HCC). Our previous work indicates that HSCs promote HCC progression by enhancing immunosuppressive cell populations including myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs). MDSCs are induced by inflammatory cytokines (e.g., prostaglandins) and are important in immune suppression. However, how HSCs mediate expansion of MDSCs is uncertain. Thus, we studied activated HSCs that could induce MDSCs from bone marrow cells and noted that HSC-induced MDSCs up-regulated immunosuppressive activity via iNOS, Arg-1, and IL-4Rα. After treating cells with a COX-2 inhibitor or an EP4 antagonist, we established that HSC-induced MDSC accumulation was mediated by the COX2-PGE2-EP4 signaling. Furthermore, in vivo animal studies confirmed that inhibition of HSC-derived PGE2 could inhibit HSC-induced MDSC accumulation and HCC growth. Thus, our data show that HSCs are required for MDSC accumulation mediated by the COX2-PGE2-EP4 pathway, and these data are the first to link HSC and MDSC subsets in HCC immune microenvironment and provide a rationale for targeting PGE2 signaling for HCC therapy.
Collapse
Affiliation(s)
- Yaping Xu
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma (Xiamen University Affiliated Zhongshan Hospital), Xiamen, Fujian, China.,Department of Basic Medicine, Xiamen Medicine College, Fujian, China
| | - Wenxiu Zhao
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma (Xiamen University Affiliated Zhongshan Hospital), Xiamen, Fujian, China
| | - Jianfeng Xu
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma (Xiamen University Affiliated Zhongshan Hospital), Xiamen, Fujian, China
| | - Jie Li
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma (Xiamen University Affiliated Zhongshan Hospital), Xiamen, Fujian, China
| | - Zaifa Hong
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma (Xiamen University Affiliated Zhongshan Hospital), Xiamen, Fujian, China
| | - Zhenyu Yin
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma (Xiamen University Affiliated Zhongshan Hospital), Xiamen, Fujian, China
| | - Xiaomin Wang
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma (Xiamen University Affiliated Zhongshan Hospital), Xiamen, Fujian, China
| |
Collapse
|
36
|
Affo S, Yu LX, Schwabe RF. The Role of Cancer-Associated Fibroblasts and Fibrosis in Liver Cancer. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2016; 12:153-186. [PMID: 27959632 DOI: 10.1146/annurev-pathol-052016-100322] [Citation(s) in RCA: 490] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Liver cancer is the second leading cause of cancer mortality worldwide, causing more than 700,000 deaths annually. Because of the wide landscape of genomic alterations and limited therapeutic success of targeting tumor cells, a recent focus has been on better understanding and possibly targeting the microenvironment in which liver tumors develop. A unique feature of liver cancer is its close association with liver fibrosis. More than 80% of hepatocellular carcinomas (HCCs) develop in fibrotic or cirrhotic livers, suggesting an important role of liver fibrosis in the premalignant environment (PME) of the liver. Cholangiocarcinoma (CCA), in contrast, is characterized by a strong desmoplasia that typically occurs in response to the tumor, suggesting a key role of cancer-associated fibroblasts (CAFs) and fibrosis in its tumor microenvironment (TME). Here, we discuss the functional contributions of myofibroblasts, CAFs, and fibrosis to the development of HCC and CCA in the hepatic PME and TME, focusing on myofibroblast- and extracellular matrix-associated growth factors, fibrosis-associated immunosuppressive pathways, as well as mechanosensitive signaling cascades that are activated by increased tissue stiffness. Better understanding of the role of myofibroblasts in HCC and CCA development and progression may provide the basis to target these cells for tumor prevention or therapy.
Collapse
Affiliation(s)
- Silvia Affo
- Department of Medicine, Columbia University, New York, NY 10032;
| | - Le-Xing Yu
- Department of Medicine, Columbia University, New York, NY 10032;
| | - Robert F Schwabe
- Department of Medicine, Columbia University, New York, NY 10032;
| |
Collapse
|
37
|
Brodt P. Role of the Microenvironment in Liver Metastasis: From Pre- to Prometastatic Niches. Clin Cancer Res 2016; 22:5971-5982. [PMID: 27797969 DOI: 10.1158/1078-0432.ccr-16-0460] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 09/19/2016] [Accepted: 09/22/2016] [Indexed: 01/14/2023]
Abstract
Liver metastases remain a major barrier to successful management of malignant disease, particularly for cancers of the gastrointestinal tract but also for other malignancies, such as breast carcinoma and melanoma. The ability of metastatic cells to survive and proliferate in the liver is determined by the outcome of complex, reciprocal interactions between tumor cells and different local resident subpopulations, including the sinusoidal endothelium, stellate, Kupffer, and inflammatory cells that are mediated through cell-cell and cell-extracellular matrix adhesion and the release of soluble factors. Cross-communication between different hepatic resident cells in response to local tissue damage and inflammation and the recruitment of bone marrow cells further enhance this intercellular communication network. Both resident and recruited cells can play opposing roles in the progression of metastasis, and the balance of these divergent effects determines whether the tumor cells will die, proliferate, and colonize the new site or enter a state of dormancy. Moreover, this delicate balance can be tilted in favor of metastasis, if factors produced by the primary tumor precondition the microenvironment to form niches of activated resident cells that promote tumor expansion. This review aims to summarize current knowledge on these diverse interactions and the impact they can have on the clinical management of hepatic metastases. Clin Cancer Res; 22(24); 5971-82. ©2016 AACR.
Collapse
Affiliation(s)
- Pnina Brodt
- Departments of Surgery, Medicine, and Oncology, McGill University and the McGill University Health Centre, Montreal, Quebec, Canada.
| |
Collapse
|
38
|
Wang F, Wang FM, Lv HM, Han T. Role of β2 adrenergic receptor signaling pathway in hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2016; 24:3598-3606. [DOI: 10.11569/wcjd.v24.i24.3598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies characterized by insidious onset and poor prognosis. Studies have shown that β adrenergic receptor signaling, especially β2 adrenergic receptor (β2-AR) signaling, regulates multiple cellular processes that contribute to the initiation and progression of cancer, including differentiation, proliferation and apoptosis. β2-AR signaling is also involved in tumor angiogenesis, progression and metastasis in HCC. Therefore, understanding of the role of the β2 adrenergic receptor signaling pathway in HCC progression and metastasis will be of great value in developing therapeutic strategies for this maliganancy. In this paper, we will discuss the role of β2 adrenergic receptor signaling pathway in HCC.
Collapse
|
39
|
Clark AM, Ma B, Taylor DL, Griffith L, Wells A. Liver metastases: Microenvironments and ex-vivo models. Exp Biol Med (Maywood) 2016; 241:1639-52. [PMID: 27390264 DOI: 10.1177/1535370216658144] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The liver is a highly metastasis-permissive organ, tumor seeding of which usually portends mortality. Its unique and diverse architectural and cellular composition enable the liver to undertake numerous specialized functions, however, this distinctive biology, notably its hemodynamic features and unique microenvironment, renders the liver intrinsically hospitable to disseminated tumor cells. The particular focus for this perspective is the bidirectional interactions between the disseminated tumor cells and the unique resident cell populations of the liver; notably, parenchymal hepatocytes and non-parenchymal liver sinusoidal endothelial, Kupffer, and hepatic stellate cells. Understanding the early steps in the metastatic seeding, including the decision to undergo dormancy versus outgrowth, has been difficult to study in 2D culture systems and animals due to numerous limitations. In response, tissue-engineered biomimetic systems have emerged. At the cutting-edge of these developments are ex vivo 'microphysiological systems' (MPS) which are cellular constructs designed to faithfully recapitulate the structure and function of a human organ or organ regions on a milli- to micro-scale level and can be made all human to maintain species-specific interactions. Hepatic MPSs are particularly attractive for studying metastases as in addition to the liver being a main site of metastatic seeding, it is also the principal site of drug metabolism and therapy-limiting toxicities. Thus, using these hepatic MPSs will enable not only an enhanced understanding of the fundamental aspects of metastasis but also allow for therapeutic agents to be fully studied for efficacy while also monitoring pharmacologic aspects and predicting toxicities. The review discusses some of the hepatic MPS models currently available and although only one MPS has been validated to relevantly modeling metastasis, it is anticipated that the adaptation of the other hepatic models to include tumors will not be long in coming.
Collapse
Affiliation(s)
- Amanda M Clark
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Bo Ma
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - D Lansing Taylor
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA University of Pittsburgh Cancer Institute, University of Pittsburgh, PA 15213, USA
| | - Linda Griffith
- Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA Pittsburgh VA Medical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| |
Collapse
|
40
|
Lin JZ, Meng LL, Li YZ, Chen SX, Xu JL, Tang YJ, Lin N. Importance of activated hepatic stellate cells and angiopoietin-1 in the pathogenesis of hepatocellular carcinoma. Mol Med Rep 2016; 14:1721-5. [DOI: 10.3892/mmr.2016.5418] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 05/23/2016] [Indexed: 11/05/2022] Open
|
41
|
Shi H, Li J, Fu D. Process of hepatic metastasis from pancreatic cancer: biology with clinical significance. J Cancer Res Clin Oncol 2016; 142:1137-1161. [PMID: 26250876 DOI: 10.1007/s00432-015-2024-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 07/23/2015] [Indexed: 12/14/2022]
Abstract
PURPOSE Pancreatic cancer shows a remarkable preference for the liver to establish secondary tumors. Selective metastasis to the liver is attributed to the development of potential microenvironment for the survival of pancreatic cancer cells. This review aims to provide a full understanding of the hepatic metastatic process from circulating pancreatic cancer cells to their settlement in the liver, serving as a basic theory for efficient prediction and treatment of metastatic diseases. METHODS A systematic search of relevant original articles and reviews was performed on PubMed, EMBASE and Cochrane Library for the purpose of this review. RESULTS Three interrelated phases are delineated as the contributions of the interaction between pancreatic cancer cells and the liver to hepatic metastasis process. Chemotaxis of disseminated pancreatic cancer cells and simultaneous defensive formation of platelets or neutrophils facilitate specific metastasis toward the liver. Remodeling of extracellular matrix and stromal cells in hepatic lobules and angiogenesis induced by proangiogenic factors support the survival and growth of clinical micrometastasis colonizing the liver. The bimodal role of the immune system or prevalence of cancer cells over the immune system makes metastatic progression successfully proceed from micrometastasis to macrometastasis. CONCLUSIONS Pancreatic cancer is an appropriate research object of cancer metastasis representing more than a straight cascade. If any of the successive or simultaneous phases, especially tumor-induced immunosuppression, is totally disrupted, hepatic metastasis will be temporarily under control or even cancelled forever. To shrink cancers on multiple fronts and prolong survival for patients, novel oral or intravenous anti-cancer agents covering one or different phases of metastatic pancreatic cancer are expected to be integrated into innovative strategies on the premise of safety and efficacious biostability.
Collapse
Affiliation(s)
- Haojun Shi
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai, 200040, China
| | - Ji Li
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai, 200040, China
| | - Deliang Fu
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai, 200040, China.
| |
Collapse
|
42
|
Wu MP, Wu LW, Chou CY. The anticancer potential of thrombospondin-1 by inhibiting angiogenesis and stroma reaction during cervical carcinogenesis. Gynecol Minim Invasive Ther 2016. [DOI: 10.1016/j.gmit.2015.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
43
|
Li J, Jia Z, Kong J, Zhang F, Fang S, Li X, Li W, Yang X, Luo Y, Lin B, Liu T. Carcinoma-Associated Fibroblasts Lead the Invasion of Salivary Gland Adenoid Cystic Carcinoma Cells by Creating an Invasive Track. PLoS One 2016; 11:e0150247. [PMID: 26954362 PMCID: PMC4782997 DOI: 10.1371/journal.pone.0150247] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 02/11/2016] [Indexed: 02/04/2023] Open
Abstract
Carcinoma-associated fibroblasts (CAFs) are critical in determining tumor invasion and metastasis. However the role of CAFs in the invasion of salivary gland adenoid cystic carcinoma (ACC) is poorly understood. In this study, we isolated primary CAFs from two ACC patients. ACC-derived CAFs expressed typical CAF biomarkers and showed increased migration and invasion activity. Conditioned medium collected from CAFs significantly promoted ACC cell migration and invasion. Co-culture of CAFs with ACC cells in a microfluidic device further revealed that CAFs localized at the invasion front and ACC cells followed the track behind the CAFs. Interfering of both matrix metalloproteinase and CXCL12/CXCR4 pathway inhibited ACC invasion promoted by CAFs. Overall, our study demonstrates that ACC-derived CAFs exhibit the most important defining feature of CAFs by promoting cancer invasion. In addition to secretion of soluble factors, CAFs also lead ACC invasion by creating an invasive track in the ECM.
Collapse
Affiliation(s)
- Jiao Li
- College of Stomatology, Dalian Medical University, Dalian, China
| | - Zhuqiang Jia
- Department of Oral Surgery, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jing Kong
- College of Stomatology, Dalian Medical University, Dalian, China
| | - Fuyin Zhang
- Department of Oral Surgery, the Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Shimeng Fang
- College of Stomatology, Dalian Medical University, Dalian, China
| | - Xiaojie Li
- College of Stomatology, Dalian Medical University, Dalian, China
| | - Wuwei Li
- College of Stomatology, Dalian Medical University, Dalian, China
| | - Xuesong Yang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, China
| | - Yong Luo
- Faculty of Chemical, Environmental and Biological Science and Technology, Dalian Technology University, Dalian, China
| | - Bingcheng Lin
- Faculty of Chemical, Environmental and Biological Science and Technology, Dalian Technology University, Dalian, China
- Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Tingjiao Liu
- College of Stomatology, Dalian Medical University, Dalian, China
- * E-mail:
| |
Collapse
|
44
|
Márquez J, Mena J, Hernandez-Unzueta I, Benedicto A, Sanz E, Arteta B, Olaso E. Ocoxin® oral solution slows down tumor growth in an experimental model of colorectal cancer metastasis to the liver in Balb/c mice. Oncol Rep 2015; 35:1265-72. [PMID: 26676882 PMCID: PMC4750781 DOI: 10.3892/or.2015.4486] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 11/06/2015] [Indexed: 12/12/2022] Open
Abstract
Liver metastatic disease is the main cause of death in colorectal cancer (CRC) patients. During metastatic spread of the disease an imbalance in the oxidative stress and inflammation plays a crucial role in tumor progression. In order to improve the efficacy of current therapies, new complementary therapeutic approaches are being analyzed including biologically active compounds with low side effects. The anti-inflammatory and anti-oxidant properties of Ocoxin® oral solution (OOS) prompt us to analyze its effect on the metastatic development of CRC to the liver. First, in vitro effect of OOS in tumor cell viability and migration was analyzed. Second, in vivo effect of different dosage patterns and concentrations in the development of hepatic metastasis was analyzed by intrasplenic inoculation of C26 colon carcinoma cells in Balb/c mice. Third, the expression of alpha smooth muscle actin, caspase-3 and Ki-67 expression was quantified by immunohistochemistry, then gene expression levels of inflammatory factors were measured by quantitative RT-PCR. According to our results, OOS reduced tumor cell viability and migration in vitro. Moreover, in vivo daily administration of OOS from the 7th day after tumor cell inoculation decreased the total area and size of metastatic foci in the liver. Furthermore, cell proliferation and fibroblast recruitment was decreased in tumor foci while a higher number of apoptotic cells were observed. Finally, RNA levels for the inflammatory mediators COX-2, IFNγ, IL1β, IL6 and TNFα were reduced in total liver. In conclusion, OOS reduced the metastatic development of colorectal cancer to the liver by increasing apoptosis, and decreasing tumor cell proliferation and fibroblast recruitment in the tumor foci, as well as the expression of inflammatory mediators in total liver. These results point out OOS as a potential supplement to be applied as complementary therapy for the treatment of liver metastasis from colorectal cancer.
Collapse
Affiliation(s)
- Joana Márquez
- Department of Cellular Biology and Histology, School of Medicine and Dentistry, Basque Country University, Leioa, Bizkaia E-48940, Spain
| | - Jorge Mena
- Department of Cellular Biology and Histology, School of Medicine and Dentistry, Basque Country University, Leioa, Bizkaia E-48940, Spain
| | - Iera Hernandez-Unzueta
- Department of Cellular Biology and Histology, School of Medicine and Dentistry, Basque Country University, Leioa, Bizkaia E-48940, Spain
| | - Aitor Benedicto
- Department of Cellular Biology and Histology, School of Medicine and Dentistry, Basque Country University, Leioa, Bizkaia E-48940, Spain
| | | | - Beatriz Arteta
- Department of Cellular Biology and Histology, School of Medicine and Dentistry, Basque Country University, Leioa, Bizkaia E-48940, Spain
| | - Elvira Olaso
- Department of Cellular Biology and Histology, School of Medicine and Dentistry, Basque Country University, Leioa, Bizkaia E-48940, Spain
| |
Collapse
|
45
|
Geng ZM, Jha RK, Li B, Chen C, Li WZ, Zheng JB, Wang L, Huanchen S. Sorafenib inhibition of hepatic stellate cell proliferation in tumor microenvironment of hepatocellular carcinoma: a study of the sorafenib mechanisms. Cell Biochem Biophys 2015; 69:717-24. [PMID: 24633454 DOI: 10.1007/s12013-014-9858-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We investigated the mechanism and effects of sorafenib on hepatic stellate cell (HSC) viability and in the liver tumor microenvironment. The expression of α-smooth muscle actin (α-SMA) was measured immunocytochemically in the LX2 cells treated with differing concentrations of sorafenib. Changes in the platelet-derived growth factor (PDGF)-BB and tumor growth factor (TGF)-β1 concentrations were detected in the LX2 supernatant using an enzyme-linked immunosorbent assay (ELISA). Expressions of the extracellular signal-regulated kinase 1 (ERK1), ERK2, and Akt signaling pathways were measured using a western blot assay. The LX2 cells were cocultured with HepG2 cells for 24 h to observe their effects on HepG2 cell invasive ability. (1) After treatment with various concentrations of sorafenib for 12, 24, 36, or 48 h, MTT assay showed that the viability of the treated LX2 cells was lower than in the controls. (2) As sorafenib concentration and time of exposure increased, α-SMA expression became weaker in the treated cells. (3) The PDGF-BB and TGF-β1 concentrations decreased with higher concentration, and longer exposures under the same sorafenib concentration. (4) The ERK1, ERK2, and Akt expressions were identical between the treated and the control groups, but their phosphorylated expression decreased with increased concentrations of sorafenib. (5) The invasive ability of the HepG2 cells induced by the LX2 gradually decreased as sorafenib concentrations increased. Sorafenib suppressed α-SMA expression, inhibited PDGF-dependent signaling pathways in HSCs, downregulated the PDGF-BB and TGF-β1 expression in the HSCs supernatant, and restrained viability of the HSCs, resulting in suppressed proliferation and invasion in the HepG2 cells.
Collapse
Affiliation(s)
- Zhi-min Geng
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Geng ZM, Li QH, Li WZ, Zheng JB, Shah V. Activated human hepatic stellate cells promote growth of human hepatocellular carcinoma in a subcutaneous xenograft nude mouse model. Cell Biochem Biophys 2015; 70:337-47. [PMID: 24676678 DOI: 10.1007/s12013-014-9918-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tumor cell microenvironment defines cancer development, also in hepatocellular carcinoma (HCC). Hepatic stellate cells (HSCs) are believed to be the key contributors to tumor microenvironment in HCC, yet their precise role in cancer progression is still unclear. The aim of this study was to determine the effect of human HSCs on progression of HCC using a subcutaneous xenograft nude mouse model. Nude mice were stratified to receive subcutaneous injections of human HCC cell line HepG2 and human HSC line LX-2 (HepG2 + LX-2), HepG2 alone, LX-2 alone, or phosphate-buffered saline. Tumor growth was assessed by measuring tumor size. After 30 days, final tumor size, weight, and histology were assessed. Compared with mice that were only injected HepG2 cells, mice injected with HepG2 + LX-2 exhibited more rapid tumor growth, increased tumor size and weight, higher tumor cell numbers due to increased proliferation and reduced apoptosis, increased fibrotic bands containing LX-2 cells, and increased tumor angiogenesis. In conclusion, HSCs play a significant role in promotion of HCC growth.
Collapse
Affiliation(s)
- Zhi-min Geng
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, 277 Yan Ta Xi Lu, Xi'an, 710061, People's Republic of China,
| | | | | | | | | |
Collapse
|
47
|
Agrawal V, Woo JH, Mauldin JP, Stone EM, Meininger CJ, Jo C, Kleypas K, Frenkel EP, Frankel AE. In-vivo evaluation of human recombinant Co-arginase against A375 melanoma xenografts. Melanoma Res 2015; 24:556-67. [PMID: 25304236 DOI: 10.1097/cmr.0000000000000119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Metastatic melanoma is a deadly form of cancer with few therapeutic options and the cause of more than 9480 deaths annually in the USA alone. Novel treatment options for this disease are urgently needed. Here we test the efficacy of a novel melanoma drug, the human recombinant Co-arginase (CoArgIPEG), against an aggressive A375 melanoma mouse model. CoArgIPEG is a modification of the naturally occurring human enzyme with improved stability, catalytic activity, and potentially lower immunogenicity compared with current amino acid-depleting drugs. Marked tumor growth reductions (mean P=0.0057) with apoptosis induction and proliferation inhibition are noted with CoArgIPEG treatment, both in the presence and in the absence of supplemental citrulline. Further, improved therapeutic efficacy has been noted against A375 xenografts relative to the naturally occurring human recombinant arginase enzyme at lower doses of CoArgIPEG. Unfortunately, after 1 month, half of the relapsing tumors showed argininosuccinate synthase induction, which correlated with Ser62-phosphorylated cMyc. Although argininosuccinate synthase induction could not be induced in vitro, a drug targeting pathway previously demonstrated to be associated with Ser62 cMyc phosphorylation - U0126 - in combination with CoArgIPEG demonstrated an in-vitro synergistic response (combination indices 0.13±0.10 and 0.14±0.10 with or without citrulline, respectively). Overall, favorable efficacy and potential synergy with other antimelanoma drugs support CoArgIPEG as a potent, novel cancer therapeutic.
Collapse
Affiliation(s)
- Vaidehi Agrawal
- aScott & White Cancer Research Institute, Baylor-Scott & White Health bDepartment of Medical Physiology, Texas A&M Health Science Center, Temple cDepartment of Chemical Engineering, University of Texas, Austin dDepartment of Internal Medicine University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Banerjee D, Hernandez SL, Garcia A, Kangsamaksin T, Sbiroli E, Andrews J, Forrester LA, Wei N, Kadenhe-Chiweshe A, Shawber CJ, Kitajewski JK, Kandel JJ, Yamashiro DJ. Notch suppresses angiogenesis and progression of hepatic metastases. Cancer Res 2015; 75:1592-602. [PMID: 25744722 DOI: 10.1158/0008-5472.can-14-1493] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 01/29/2015] [Indexed: 12/12/2022]
Abstract
The Notch pathway plays multiple key roles in tumorigenesis, and its signaling components have therefore aroused great interest as targets for emerging therapies. Here, we show that inhibition of Notch, using a soluble receptor Notch1 decoy, unexpectedly caused a remarkable increase in liver metastases from neuroblastoma and breast cancer cells. Increased liver metastases were also seen after treatment with the γ-secretase inhibitor PF-03084014. Transgenic mice with heterozygous loss of Notch1 demonstrated a marked increase in hepatic metastases, indicating that Notch1 signaling acts as metastatic suppressor in the liver microenvironment. Inhibition of DLL1/4 with ligand-specific Notch1 decoys increased sprouting of sinusoidal endothelial cells into micrometastases, thereby supporting early metastatic angiogenic growth. Inhibition of tumor-derived JAG1 signaling activated hepatic stellate cells, increasing their recruitment to vasculature of micrometastases, thereby supporting progression to macrometastases. These results demonstrate that inhibition of Notch causes pathologic activation of liver stromal cells, promoting angiogenesis and growth of hepatic metastases. Our findings have potentially serious implications for Notch inhibition therapy.
Collapse
Affiliation(s)
- Debarshi Banerjee
- Department of Pediatrics, Columbia University Medical Center, New York, New York
| | - Sonia L Hernandez
- Department of Pediatrics, Columbia University Medical Center, New York, New York
| | - Alejandro Garcia
- Department of Surgery, Columbia University Medical Center, New York, New York
| | - Thaned Kangsamaksin
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, New York
| | - Emily Sbiroli
- Department of Surgery, Columbia University Medical Center, New York, New York
| | - John Andrews
- Department of Surgery, Columbia University Medical Center, New York, New York
| | - Lynn Ann Forrester
- Department of Surgery, Columbia University Medical Center, New York, New York
| | - Na Wei
- Department of Pediatrics, Columbia University Medical Center, New York, New York
| | | | - Carrie J Shawber
- Department of Surgery, Columbia University Medical Center, New York, New York. Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, New York
| | - Jan K Kitajewski
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, New York. Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Jessica J Kandel
- Department of Surgery, Columbia University Medical Center, New York, New York
| | - Darrell J Yamashiro
- Department of Pediatrics, Columbia University Medical Center, New York, New York. Department of Surgery, Columbia University Medical Center, New York, New York. Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York.
| |
Collapse
|
49
|
Song J, Ge Z, Yang X, Luo Q, Wang C, You H, Ge T, Deng Y, Lin H, Cui Y, Chu W, Yao M, Zhang Z, Gu J, Fan J, Qin W. Hepatic stellate cells activated by acidic tumor microenvironment promote the metastasis of hepatocellular carcinoma via osteopontin. Cancer Lett 2015; 356:713-720. [PMID: 25449435 DOI: 10.1016/j.canlet.2014.10.021] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Revised: 10/21/2014] [Accepted: 10/21/2014] [Indexed: 12/12/2022]
Abstract
Extracellular pH of solid tumor is generally acidic due to excessive glycolysis and poor perfusion. But whether acidic tumor microenvironment influenced the stromal cells infiltrating in tumor remains unknown. As the predominant progenitor of stromal cells in liver, the number of activated hepatic stellate cells (HSCs) was found positively correlated to the acidification level in the tumor tissues of HCC patients in our study. Whereas, in vitro acidic culture condition and in vivo co-implanting xenograft model were adopted to study the response of HSCs and its influence on HCC progression. HSCs were activated under acidic culture condition depending on the phosphorylation of cellular signal-regulated kinase (ERK). Acidity-activated HSCs promoted HCC metastasis in vitro and in vivo. Osteopontin (OPN) excretion from HSCs was increased under acidic condition and proved to promote the migration of HCC cells. Furthermore, the expression level of OPN was significantly associated with myofibroblasts and the combination of α-SMA with OPN was a powerful predictor for poor prognosis of HCC patients. Activation of HSCs in acidic tumor microenvironment represents a novel mechanism for HCC metastasis and provides a potential therapeutic strategy for HCC.
Collapse
MESH Headings
- Acids/chemistry
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/mortality
- Carcinoma, Hepatocellular/secondary
- Cell Movement
- Cell Proliferation
- Culture Media, Conditioned/pharmacology
- Enzyme-Linked Immunosorbent Assay
- Female
- Fluorescent Antibody Technique
- Gene Expression Profiling
- Hepatic Stellate Cells/metabolism
- Hepatic Stellate Cells/pathology
- Humans
- Hydrogen-Ion Concentration
- Immunoenzyme Techniques
- Liver Neoplasms/metabolism
- Liver Neoplasms/mortality
- Liver Neoplasms/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Myofibroblasts/metabolism
- Myofibroblasts/pathology
- Osteopontin/genetics
- Osteopontin/metabolism
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Cells, Cultured
- Tumor Microenvironment
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jin Song
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 25/Ln 2200 Xie-Tu Road, Shanghai 200032, China
| | - Zhouhong Ge
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 25/Ln 2200 Xie-Tu Road, Shanghai 200032, China
| | - Xinrong Yang
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical College, Fudan University, Key Laboratory for Carcinogenesis & Cancer Invasion, the Chinese Ministry of Education, Shanghai 200032, China
| | - Qin Luo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 25/Ln 2200 Xie-Tu Road, Shanghai 200032, China
| | - Cun Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 25/Ln 2200 Xie-Tu Road, Shanghai 200032, China
| | - Haiyan You
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 25/Ln 2200 Xie-Tu Road, Shanghai 200032, China
| | - Tianxiang Ge
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 25/Ln 2200 Xie-Tu Road, Shanghai 200032, China
| | - Yun Deng
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 25/Ln 2200 Xie-Tu Road, Shanghai 200032, China
| | - Hechun Lin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 25/Ln 2200 Xie-Tu Road, Shanghai 200032, China
| | - Yongqi Cui
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 25/Ln 2200 Xie-Tu Road, Shanghai 200032, China
| | - Wei Chu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 25/Ln 2200 Xie-Tu Road, Shanghai 200032, China
| | - Ming Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 25/Ln 2200 Xie-Tu Road, Shanghai 200032, China
| | - Zhigang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 25/Ln 2200 Xie-Tu Road, Shanghai 200032, China
| | - Jianren Gu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 25/Ln 2200 Xie-Tu Road, Shanghai 200032, China
| | - Jia Fan
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical College, Fudan University, Key Laboratory for Carcinogenesis & Cancer Invasion, the Chinese Ministry of Education, Shanghai 200032, China.
| | - Wenxin Qin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 25/Ln 2200 Xie-Tu Road, Shanghai 200032, China.
| |
Collapse
|
50
|
Illemann M, Eefsen RHL, Bird NC, Majeed A, Osterlind K, Laerum OD, Alpízar-Alpízar W, Lund IK, Høyer-Hansen G. Tissue inhibitor of matrix metalloproteinase-1 expression in colorectal cancer liver metastases is associated with vascular structures. Mol Carcinog 2015; 55:193-208. [PMID: 25594187 PMCID: PMC6680289 DOI: 10.1002/mc.22269] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 10/30/2014] [Accepted: 11/26/2014] [Indexed: 01/22/2023]
Abstract
Metastatic growth by colorectal cancer cells in the liver requires the ability of the cancer cells to interact with the new microenvironment. This interaction results in three histological growth patterns of liver metastases: desmoplastic, pushing, and replacement. In primary colorectal cancer several proteases, involved in the degradation of extracellular matrix components, are up‐regulated. In liver metastases, their expression is growth pattern dependent. Tissue inhibitor of matrix metalloproteinase‐1 (TIMP‐1) is a strong prognostic marker in plasma from colorectal cancer patients, with significant higher levels in patients with metastatic disease. We therefore wanted to determine the expression pattern of TIMP‐1 in primary colorectal cancers and their matching liver metastases. TIMP‐1 mRNA was primarily seen in α‐smooth‐muscle actin (α‐SMA)‐positive cells. In all primary tumors and liver metastases with desmoplastic growth pattern, TIMP‐1 mRNA was primarily found in α‐SMA‐positive myofibroblasts located at the invasive front. Some α‐SMA‐positive cells with TIMP‐1 mRNA were located adjacent to CD34‐positive endothelial cells, identifying them as pericytes. This indicates that TIMP‐1 in primary tumors and liver metastases with desmoplastic growth pattern has dual functions; being an MMP‐inhibitor at the cancer periphery and involved in tumor‐induced angiogenesis in the pericytes. In the liver metastases with pushing or replacement growth patterns, TIMP‐1 was primarily expressed by activated hepatic stellate cells at the metastasis/liver parenchyma interface. These cells were located adjacent to CD34‐positive endothelial cells, suggesting a function in tumor‐induced angiogenesis. We therefore conclude that TIMP‐1 expression is growth pattern dependent in colorectal cancer liver metastases. © 2015 The Authors. Molecular Carcinogenesis published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Martin Illemann
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Rikke Helene Løvendahl Eefsen
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark.,Department of Oncology, Rigshospitalet, Copenhagen, Denmark
| | | | - Ali Majeed
- Academic Surgical Unit, University of Sheffield, Sheffield, England
| | - Kell Osterlind
- Department of Oncology, Rigshospitalet, Copenhagen, Denmark
| | - Ole Didrik Laerum
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Warner Alpízar-Alpízar
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark.,Center for Research on Microscopic Structures, University of Costa Rica, San José, Costa Rica
| | - Ida Katrine Lund
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Gunilla Høyer-Hansen
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark.,Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|