1
|
Shuai Q, Xu X, Liang Y, Halbiyat Z, Lu X, Hu Z, Peng Z, An J, Feng Z, Huang T, Zhao H, Liu Z, Xu J, Xie J. Engineered in vivo and in vitro tumor model recapitulates vasculogenic mimicry signatures in melanoma. Bioeng Transl Med 2024; 9:e10648. [PMID: 39036079 PMCID: PMC11256191 DOI: 10.1002/btm2.10648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 12/21/2023] [Accepted: 01/11/2024] [Indexed: 07/23/2024] Open
Abstract
Vasculogenic mimicry (VM) describes a process by which tumor cells formed a novel microcirculation pattern in an endothelial cell-free manner. Clinically, VM is associated with aggressive phenotype and poor patient survival. However, the current models for investigating VM include 2D monolayer cultures, Matrigel-based cultures, and animal models, each of which has limitations. Matrigel-based models often exhibit batch-to-batch variations, while in vivo tumor models currently produce insufficient amounts of VM. There is currently no suitable tumor model to discover new therapeutic targets against VM. Herein, we establish an extracellular matrix (ECM)-based engineered tumor model in vivo and in vitro. In this study, we demonstrate that matrix proteins enhanced the VM formation in the engineered xenograft model. Furthermore, we also investigated the role of collagen/fibronectin (FN) in melanoma progression and VM formation. Compared with cells cultured on TCPS plates, the B16F10 cells cultured on collagen/FN coated plates showed increased proliferation and stemness, and significantly enhanced invasion and formation of VM networks. Molecular mechanism analysis showed that Integrin/VE-cadherin/EphA2/PI3K/MMP-2 signaling pathways are responsible for VM formation. Our results indicate that collagen/FN matrix plays an important role in VM formation in melanoma, suggesting that ECM protein is a potential therapeutic target for anti-VM therapy for melanoma.
Collapse
Affiliation(s)
- Qizhi Shuai
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuanChina
| | - Xinrui Xu
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuanChina
- Laboratory of Ethnopharmacology, Tissue‐Orientated Property of Chinese Medicine Key Laboratory of Sichuan ProvinceWest China School of Medicine, West China Hospital, Sichuan UniversityChengduChina
| | - Yuxiang Liang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuanChina
- Experimental Animal Center of Shanxi Medical UniversityShanxi Key Laboratory of Human Disease and Animal ModelsTaiyuanChina
| | - Zulala Halbiyat
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuanChina
| | - Xin Lu
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuanChina
| | - Zixuan Hu
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuanChina
| | - Zhiwei Peng
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuanChina
| | - Jie An
- Department of Nuclear MedicineThe First Hospital of Shanxi Medical University, Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical UniversityTaiyuanChina
| | - Zhiwei Feng
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuanChina
| | - Tingjuan Huang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuanChina
| | - Hong Zhao
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuanChina
| | - Zhizhen Liu
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuanChina
| | - Jun Xu
- Department of Hepatopancreatobiliary SurgeryThe First Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuanChina
| |
Collapse
|
2
|
Wu Z, Bian Y, Chu T, Wang Y, Man S, Song Y, Wang Z. The role of angiogenesis in melanoma: Clinical treatments and future expectations. Front Pharmacol 2022; 13:1028647. [PMID: 36588679 PMCID: PMC9797529 DOI: 10.3389/fphar.2022.1028647] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
The incidence of melanoma has increased rapidly over the past few decades, with mortality accounting for more than 75% of all skin cancers. The high metastatic potential of Melanoma is an essential factor in its high mortality. Vascular angiogenic system has been proved to be crucial for the metastasis of melanoma. An in-depth understanding of angiogenesis will be of great benefit to melanoma treatment and may promote the development of melanoma therapies. This review summarizes the recent advances and challenges of anti-angiogenic agents, including monoclonal antibodies, tyrosine kinase inhibitors, human recombinant Endostatin, and traditional Chinese herbal medicine. We hope to provide a better understanding of the mechanisms, clinical research progress, and future research directions of melanoma.
Collapse
Affiliation(s)
- Zhuzhu Wu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China,Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yifei Bian
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tianjiao Chu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuman Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuai Man
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China,Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China,Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, China,*Correspondence: Shuai Man, ; Yongmei Song, ; Zhenguo Wang,
| | - Yongmei Song
- Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China,*Correspondence: Shuai Man, ; Yongmei Song, ; Zhenguo Wang,
| | - Zhenguo Wang
- Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China,Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China,*Correspondence: Shuai Man, ; Yongmei Song, ; Zhenguo Wang,
| |
Collapse
|
3
|
Hosen SMZ, Uddin MN, Xu Z, Buckley BJ, Perera C, Pang TCY, Mekapogu AR, Moni MA, Notta F, Gallinger S, Pirola R, Wilson J, Ranson M, Goldstein D, Apte M. Metastatic phenotype and immunosuppressive tumour microenvironment in pancreatic ductal adenocarcinoma: Key role of the urokinase plasminogen activator (PLAU). Front Immunol 2022; 13:1060957. [PMID: 36591282 PMCID: PMC9794594 DOI: 10.3389/fimmu.2022.1060957] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022] Open
Abstract
Background Previous studies have revealed the role of dysregulated urokinase plasminogen activator (encoded by PLAU) expression and activity in several pathways associated with cancer progression. However, systematic investigation into the association of PLAU expression with factors that modulate PDAC (pancreatic ductal adenocarcinoma) progression is lacking, such as those affecting stromal (pancreatic stellate cell, PSC)-cancer cell interactions, tumour immunity, PDAC subtypes and clinical outcomes from potential PLAU inhibition. Methods This study used an integrated bioinformatics approach to identify prognostic markers correlated with PLAU expression using different transcriptomics, proteomics, and clinical data sets. We then determined the association of dysregulated PLAU and correlated signatures with oncogenic pathways, metastatic phenotypes, stroma, immunosuppressive tumour microenvironment (TME) and clinical outcome. Finally, using an in vivo orthotopic model of pancreatic cancer, we confirmed the predicted effect of inhibiting PLAU on tumour growth and metastasis. Results Our analyses revealed that PLAU upregulation is not only associated with numerous other prognostic markers but also associated with the activation of various oncogenic signalling pathways, aggressive phenotypes relevant to PDAC growth and metastasis, such as proliferation, epithelial-mesenchymal transition (EMT), stemness, hypoxia, extracellular cell matrix (ECM) degradation, upregulation of stromal signatures, and immune suppression in the tumour microenvironment (TME). Moreover, the upregulation of PLAU was directly connected with signalling pathways known to mediate PSC-cancer cell interactions. Furthermore, PLAU upregulation was associated with the aggressive basal/squamous phenotype of PDAC and significantly reduced overall survival, indicating that this subset of patients may benefit from therapeutic interventions to inhibit PLAU activity. Our studies with a clinically relevant orthotopic pancreatic model showed that even short-term PLAU inhibition is sufficient to significantly halt tumour growth and, importantly, eliminate visible metastasis. Conclusion Elevated PLAU correlates with increased aggressive phenotypes, stromal score, and immune suppression in PDAC. PLAU upregulation is also closely associated with the basal subtype type of PDAC; patients with this subtype are at high risk of mortality from the disease and may benefit from therapeutic targeting of PLAU.
Collapse
Affiliation(s)
- S. M. Zahid Hosen
- Pancreatic Research Group, SWS Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia,Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia
| | - Md. Nazim Uddin
- Institute of Food Science and Technology, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh
| | - Zhihong Xu
- Pancreatic Research Group, SWS Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia,Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia
| | - Benjamin J. Buckley
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia,Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
| | - Chamini Perera
- Pancreatic Research Group, SWS Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia,Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia
| | - Tony C. Y. Pang
- Pancreatic Research Group, SWS Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia,Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, The University of Sydney, Sydney, NSW, Australia
| | - Alpha Raj Mekapogu
- Pancreatic Research Group, SWS Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia,Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia
| | - Mohammad Ali Moni
- School of Health and Rehabilitation Sciences, Faculty of Health and Behavioural Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - Faiyaz Notta
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Steven Gallinger
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Ron Pirola
- Pancreatic Research Group, SWS Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Jeremy Wilson
- Pancreatic Research Group, SWS Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Marie Ranson
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia,Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
| | - David Goldstein
- Prince of Wales Clinical School, University of New South Wales, Sydney, NSW, Australia,Department of Medical Oncology, Prince of Wales Hospital, Randwick, NSW, Australia
| | - Minoti Apte
- Pancreatic Research Group, SWS Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia,Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia,*Correspondence: Minoti Apte,
| |
Collapse
|
4
|
Andreucci E, Peppicelli S, Ruzzolini J, Bianchini F, Calorini L. Physicochemical aspects of the tumour microenvironment as drivers of vasculogenic mimicry. Cancer Metastasis Rev 2022; 41:935-951. [PMID: 36224457 PMCID: PMC9758104 DOI: 10.1007/s10555-022-10067-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/04/2022] [Indexed: 01/25/2023]
Abstract
Tumour vascularisation is vital for cancer sustainment representing not only the main source of nutrients and oxygen supply but also an escape route for single or clustered cancer cells that, once detached from the primary mass, enter the blood circulation and disseminate to distant organs. Among the mechanisms identified to contribute to tumour vascularisation, vasculogenic mimicry (VM) is gaining increasing interest in the scientific community representing an intriguing target for cancer treatment. VM indeed associates with highly aggressive tumour phenotypes and strongly impairs patient outcomes. Differently from vessels of healthy tissues, tumour vasculature is extremely heterogeneous and tortuous, impeding efficient chemotherapy delivery, and at the meantime hyperpermeable and thus extremely accessible to metastasising cancer cells. Moreover, tumour vessel disorganisation creates a self-reinforcing vicious circle fuelling cancer malignancy and progression. Because of the inefficient oxygen delivery and metabolic waste removal from tumour vessels, many cells within the tumour mass indeed experience hypoxia and acidosis, now considered hallmarks of cancer. Being strong inducers of vascularisation, therapy resistance, inflammation and metastasis, hypoxia and acidosis create a permissive microenvironment for cancer progression and dissemination. Along with these considerations, we decided to focus our attention on the relationship between hypoxia/acidosis and VM. Indeed, besides tumour angiogenesis, VM is strongly influenced by both hypoxia and acidosis, which could potentiate each other and fuel this vicious circle. Thus, targeting hypoxia and acidosis may represent a potential target to treat VM to impair tumour perfusion and cancer cell sustainment.
Collapse
Affiliation(s)
- Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.
| | - Jessica Ruzzolini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| |
Collapse
|
5
|
Andreucci E, Ruzzolini J, Bianchini F, Versienti G, Biagioni A, Lulli M, Guasti D, Nardini P, Serratì S, Margheri F, Laurenzana A, Nediani C, Peppicelli S, Calorini L. miR-214-Enriched Extracellular Vesicles Released by Acid-Adapted Melanoma Cells Promote Inflammatory Macrophage-Dependent Tumor Trans-Endothelial Migration. Cancers (Basel) 2022; 14:cancers14205090. [PMID: 36291876 PMCID: PMC9599952 DOI: 10.3390/cancers14205090] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 11/22/2022] Open
Abstract
Simple Summary Cutaneous melanoma is the most aggressive form of skin cancer with high-metastatic ability. Despite the recent advancements in melanoma treatments, the prognosis of metastatic patients remains very poor. A better understanding of the molecular mechanisms leading to melanoma dissemination is urgently needed in order to develop novel therapeutical strategies to ameliorate patients’ outcomes. Extracellular vesicles (EV) released by tumor cells are key players in metastasis development: by conveying bioactive molecules with oncogenic activity, they can modulate the surrounding—and even the distant—microenvironment and reprogram recipient cells to facilitate the metastatic cascade. Here, we show that melanoma cells release a higher amount of miR-214-enriched EV when adapted to extracellular acidosis, which promote a macrophage activation state, capable of facilitating the trans-endothelial migration of melanoma cells. Thus, we disclose a new molecular mechanism to prevent melanoma intravasation based on miR-214 targeting. Abstract The understanding of the molecular mechanisms leading to melanoma dissemination is urgently needed in view of the identification of new targets and the development of innovative strategies to improve patients’ outcomes. Within the complexity of tumor intercellular communications leading to metastatic dissemination, extracellular vesicles (EV) released by tumor cells are central players. Indeed, the ability to travel through the circulatory system conveying oncogenic bioactive molecules even at distant sites makes EV capable of modulating recipient cells to facilitate metastatic dissemination. The dynamic remodeling of the tumor microenvironment might influence, along with a number of other events, tumoral EV release. We observed that, in melanoma, extracellular acidosis increases the release of EV enriched in miR-214, an onco-miRNA involved in melanoma metastasis. Then, miR-214-enriched EV were found to induce a state of macrophage activation, leading to an overproduction of proinflammatory cytokines and nitric oxide. Such an inflammatory microenvironment was able to alter the endothelial cell permeability, thereby facilitating the trans-endothelial migration of melanoma cells, a crucial step in the metastatic cascade. The use of synthetic miR-214 inhibitors and miR-214 overexpression allowed us to demonstrate the key role of miR-214 in the EV-dependent induction of macrophage activation. Overall, our in vitro study reveals that the release of tumor miR-214-enriched EV, potentiated by adapting tumor cells to extracellular acidosis, drives a macrophage-dependent trans-endothelial migration of melanoma cells. This finding points to miR-214 as a potential new therapeutic target to prevent melanoma intravasation.
Collapse
Affiliation(s)
- Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
- Correspondence: (E.A.); (L.C.)
| | - Jessica Ruzzolini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Giampaolo Versienti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Daniele Guasti
- Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Patrizia Nardini
- Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Simona Serratì
- Laboratory of Nanotecnology, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Chiara Nediani
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, 50134 Florence, Italy
- Correspondence: (E.A.); (L.C.)
| |
Collapse
|
6
|
Biagioni A, Peri S, Schiavone N, Giommoni E, Papucci L. Editorial: Redox Potential and Metabolic Behavior in Gastrointestinal Cancers. Front Oncol 2022; 12:882237. [PMID: 35574349 PMCID: PMC9092526 DOI: 10.3389/fonc.2022.882237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/05/2022] [Indexed: 11/21/2022] Open
Affiliation(s)
- Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Firenze, Italy
| | - Sara Peri
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
| | - Nicola Schiavone
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Firenze, Italy
| | - Elisa Giommoni
- Medical Oncology Unit, Department of Oncology and Robotic Surgery, AOU Careggi, Firenze, Italy
| | - Laura Papucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Firenze, Italy
| |
Collapse
|
7
|
Del Bufalo D, Di Martile M, Valentini E, Manni I, Masi I, D'Amore A, Filippini A, Nicoletti C, Zaccarini M, Cota C, Castro MV, Quezada MJ, Rosanò L, Lopez-Bergami P, D'Aguanno S. Bcl-2-like protein-10 increases aggressive features of melanoma cells. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:11-26. [PMID: 36046354 PMCID: PMC9400776 DOI: 10.37349/etat.2022.00068] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/20/2021] [Indexed: 11/29/2022] Open
Abstract
Aim: B-cell lymphoma-2 (Bcl-2)-like protein-10 (Bcl2L10) is the less studied
member of Bcl-2 family proteins, with the controversial role in different
cancer histotypes. Very recently, Bcl2L10 expression in melanoma tumor
specimens and its role in melanoma response to therapy have been
demonstrated. Here, the involvement of Bcl2L10 on the in
vitro and in vivo properties associated with
melanoma aggressive features has been investigated. Methods: Endogenous Bcl2L10 protein expression was detected by western blotting
analysis in a panel of patient-derived and commercially available human
melanoma cells. In vitro assays to evaluate clonogenicity,
cell proliferation, cell migration, cell invasion, and in
vitro capillary-like structure formation [vasculogenic
mimicry (VM)] have been performed by using human melanoma cells
stably overexpressing Bcl2L10 or transiently transfected for loss/gain
function of Bcl2L10, grown under two- or three-dimensional (3D) conditions
Xenograft melanoma model was employed to evaluate in vivo
tumor growth and angiogenesis. Results: Results demonstrated that Bcl2L10 acts as an inducer of in
vitro cell migration, invasion, and VM, while in
vitro cell proliferation, in vivo tumor
growth, as well as colony formation properties were not affected. Dissecting
different signaling pathways, it was found that Bcl2L10 positively affects
the phosphorylation of extracellular-signal-regulated kinase (ERK) and the
expression of markers of cell invasion, such as urokinase plasminogen
activator receptor (uPAR) and matrix metalloproteinases (MMPs). Of note,
Bcl2L10-dependent in vitro migration, invasion, and VM are
linked to uPAR. Bcl2L10 also negatively regulates the intracellular calcium
level. Finally, reduced invasion capability in 3D spheroid invasion assay of
melanoma cells transiently overexpressing Bcl2L10 was observed after
treatment with inhibitors of MMPs and uPAR. Conclusions: Overall, data reported in this paper provide evidence supporting a positive
role of Bcl2L10 in melanoma aggressive features.
Collapse
Affiliation(s)
- Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Marta Di Martile
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Elisabetta Valentini
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Isabella Manni
- SAFU Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Ilenia Masi
- Institute of Molecular Biology and Pathology, National Research Council, 00161 Rome, Italy
| | - Antonella D'Amore
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University, 00161 Rome, Italy
| | - Antonio Filippini
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University, 00161 Rome, Italy
| | - Carmine Nicoletti
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University, 00161 Rome, Italy
| | - Marco Zaccarini
- Genetic Research, Dermatological Molecular Biology and Dermatopathology Unit, IRCCS San Gallicano Dermatological Institute, 00144 Rome, Italy
| | - Carlo Cota
- Genetic Research, Dermatological Molecular Biology and Dermatopathology Unit, IRCCS San Gallicano Dermatological Institute, 00144 Rome, Italy
| | - Maria Victoria Castro
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo, Universidad Maimónides, Buenos Aires C1405BCK, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires C1405BCK, Argentina
| | - María Josefina Quezada
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo, Universidad Maimónides, Buenos Aires C1405BCK, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires C1405BCK, Argentina
| | - Laura Rosanò
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; Institute of Molecular Biology and Pathology, National Research Council, 00161 Rome, Italy
| | - Pablo Lopez-Bergami
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo, Universidad Maimónides, Buenos Aires C1405BCK, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires C1405BCK, Argentina
| | - Simona D'Aguanno
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| |
Collapse
|
8
|
Therapeutic Strategies Targeting Urokinase and Its Receptor in Cancer. Cancers (Basel) 2022; 14:cancers14030498. [PMID: 35158766 PMCID: PMC8833673 DOI: 10.3390/cancers14030498] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/11/2022] [Accepted: 01/15/2022] [Indexed: 01/19/2023] Open
Abstract
Several studies have ascertained that uPA and uPAR do participate in tumor progression and metastasis and are involved in cell adhesion, migration, invasion and survival, as well as angiogenesis. Increased levels of uPA and uPAR in tumor tissues, stroma and biological fluids correlate with adverse clinic-pathologic features and poor patient outcomes. After binding to uPAR, uPA activates plasminogen to plasmin, a broad-spectrum matrix- and fibrin-degrading enzyme able to facilitate tumor cell invasion and dissemination to distant sites. Moreover, uPAR activated by uPA regulates most cancer cell activities by interacting with a broad range of cell membrane receptors. These findings make uPA and uPAR not only promising diagnostic and prognostic markers but also attractive targets for developing anticancer therapies. In this review, we debate the uPA/uPAR structure-function relationship as well as give an update on the molecules that interfere with or inhibit uPA/uPAR functions. Additionally, the possible clinical development of these compounds is discussed.
Collapse
|
9
|
Biagioni A, Andreucci E. Immunohistochemistry for VM Markers. Methods Mol Biol 2022; 2514:141-152. [PMID: 35771426 DOI: 10.1007/978-1-0716-2403-6_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Vasculogenic mimicry (VM) is the biological process by which aggressive cancer cells are able to organize themselves-independently from endothelial cells-into new vessel-like structures to sustain fast tumor perfusion and thus an efficient supply of oxygen and nutrients, required for rapid cancer growth and dissemination. In the last two decades, the molecular mechanisms and key regulators of VM have been identified. Several methods are currently available to detect VM both in vitro and in vivo, but the gold standard is still the immunohistochemical staining of specific antigens. Even though many markers are debated if belong to the angiogenic process or VM exclusively, the immunohistochemistry of CD31 and the PAS reaction often clarify in frozen or paraffin sections the pathologic status and the vasculature grade of a tumor mass.
Collapse
Affiliation(s)
- Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.
| | - Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.
| |
Collapse
|