1
|
Cen K, Huang Y, Xie Y, Liu Y. The guardian of intracranial vessels: Why the pericyte? Biomed Pharmacother 2024; 176:116870. [PMID: 38850658 DOI: 10.1016/j.biopha.2024.116870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/27/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
Intracranial atherosclerotic stenosis (ICAS) is a pathological condition characterized by progressive narrowing or complete blockage of intracranial blood vessels caused by plaque formation. This condition leads to reduced blood flow to the brain, resulting in cerebral ischemia and hypoxia. Ischemic stroke (IS) resulting from ICAS poses a significant global public health challenge, especially among East Asian populations. However, the underlying causes of the notable variations in prevalence among diverse populations, as well as the most effective strategies for preventing and treating the rupture and blockage of intracranial plaques, remain incompletely comprehended. Rupture of plaques, bleeding, and thrombosis serve as precipitating factors in the pathogenesis of luminal obstruction in intracranial arteries. Pericytes play a crucial role in the structure and function of blood vessels and face significant challenges in regulating the Vasa Vasorum (VV)and preventing intraplaque hemorrhage (IPH). This review aims to explore innovative therapeutic strategies that target the pathophysiological mechanisms of vulnerable plaques by modulating pericyte biological function. It also discusses the potential applications of pericytes in central nervous system (CNS) diseases and their prospects as a therapeutic intervention in the field of biological tissue engineering regeneration.
Collapse
Affiliation(s)
- Kuan Cen
- Department of Neurology, Zhongnan Hospital Affiliated to Wuhan University, Wuhan 430000, China
| | - YinFei Huang
- Department of Neurology, Zhongnan Hospital Affiliated to Wuhan University, Wuhan 430000, China
| | - Yu Xie
- Department of Neurology, Zhongnan Hospital Affiliated to Wuhan University, Wuhan 430000, China
| | - YuMin Liu
- Department of Neurology, Zhongnan Hospital Affiliated to Wuhan University, Wuhan 430000, China.
| |
Collapse
|
2
|
Mansur A, Radovanovic I. Defining the Role of Oral Pathway Inhibitors as Targeted Therapeutics in Arteriovenous Malformation Care. Biomedicines 2024; 12:1289. [PMID: 38927496 PMCID: PMC11201820 DOI: 10.3390/biomedicines12061289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Arteriovenous malformations (AVMs) are vascular malformations that are prone to rupturing and can cause significant morbidity and mortality in relatively young patients. Conventional treatment options such as surgery and endovascular therapy often are insufficient for cure. There is a growing body of knowledge on the genetic and molecular underpinnings of AVM development and maintenance, making the future of precision medicine a real possibility for AVM management. Here, we review the pathophysiology of AVM development across various cell types, with a focus on current and potential druggable targets and their therapeutic potentials in both sporadic and familial AVM populations.
Collapse
Affiliation(s)
- Ann Mansur
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Laboratory Medicine and Pathobiology, School of Graduate Studies, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ivan Radovanovic
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Laboratory Medicine and Pathobiology, School of Graduate Studies, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada
| |
Collapse
|
3
|
De Palma M, Hanahan D. Milestones in tumor vascularization and its therapeutic targeting. NATURE CANCER 2024; 5:827-843. [PMID: 38918437 DOI: 10.1038/s43018-024-00780-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 04/22/2024] [Indexed: 06/27/2024]
Abstract
Research into the mechanisms and manifestations of solid tumor vascularization was launched more than 50 years ago with the proposition and experimental demonstrations that angiogenesis is instrumental for tumor growth and was, therefore, a promising therapeutic target. The biological knowledge and therapeutic insights forthcoming have been remarkable, punctuated by new concepts, many of which were not foreseen in the early decades. This article presents a perspective on tumor vascularization and its therapeutic targeting but does not portray a historical timeline. Rather, we highlight eight conceptual milestones, integrating initial discoveries and recent progress and posing open questions for the future.
Collapse
Affiliation(s)
- Michele De Palma
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, Switzerland.
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland.
| | - Douglas Hanahan
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, Switzerland.
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.
| |
Collapse
|
4
|
AL-Eitan L, Abusirdaneh R. The synthetic cannabinoid 5-fluoro ABICA upregulates angiogenic markers and stimulates tube formation in human brain microvascular endothelial cells. J Taibah Univ Med Sci 2024; 19:359-371. [PMID: 38357583 PMCID: PMC10864802 DOI: 10.1016/j.jtumed.2024.01.002] [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: 07/20/2023] [Revised: 12/11/2023] [Accepted: 01/21/2024] [Indexed: 02/16/2024] Open
Abstract
Objective Synthetic cannabinoids (SCs), a class of psychoactive compounds emulating the effects of natural cannabis, have prompted addiction and psychosis concerns. However, recent research has suggested potential pharmacological applications, particularly in brain angiogenesis-an essential physiological process for growth, repair, and tissue maintenance, in which new blood vasculature is formed from existing vasculature. This study explored the in vitro ability of the SC 5-fluoro ABICA to enhance new blood formation processes in human brain microvascular endothelial cells (HBMECs). Methods HBMECs were treated with various concentrations of 5-fluoro ABICA (1 μM, 0.1 μM, 0.01 μM, 0.001 μM, and 0.0001 μM). A comprehensive analysis was conducted, including MTT assays indicating cell viability, wound healing assays indicating migration ability, and tube formation assays indicating the angiogenesis potential of endothelial cells. Additionally, mRNA expression and protein levels of specific pro-angiogenic factors were measured, and the phosphorylation levels of glycogen synthase kinase-3β were detected in treated HBMECs through ELISA, real-time PCR, and western blotting. Results Treatment with 5-fluoro ABICA effectively stimulated proliferation, migration, and tube formation in HBMECs in a dose-dependent manner; markedly increased the expression of pro-angiogenic factors; and upregulated levels of phosphorylated-GSK-3β. Conclusion Our findings demonstrate that 5-fluoro ABICA stimulates angiogenesis in endothelial cells, thus potentially offering therapeutic options for diseases associated with angiogenesis. However, further research is needed to fully understand the molecular mechanism of 5-fluoro ABICA in angiogenesis, including ethical considerations regarding its use in medical research.
Collapse
Affiliation(s)
- Laith AL-Eitan
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, Jordan
| | - Rawan Abusirdaneh
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
5
|
Clark GC, Lai A, Agarwal A, Liu Z, Wang XY. Biopterin metabolism and nitric oxide recoupling in cancer. Front Oncol 2024; 13:1321326. [PMID: 38469569 PMCID: PMC10925643 DOI: 10.3389/fonc.2023.1321326] [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: 10/13/2023] [Accepted: 12/26/2023] [Indexed: 03/13/2024] Open
Abstract
Tetrahydrobiopterin is a cofactor necessary for the activity of several enzymes, the most studied of which is nitric oxide synthase. The role of this cofactor-enzyme relationship in vascular biology is well established. Recently, tetrahydrobiopterin metabolism has received increasing attention in the field of cancer immunology and immunotherapy due to its involvement in the cytotoxic T cell response. Past research has demonstrated that when the availability of BH4 is low, as it is in chronic inflammatory conditions and tumors, electron transfer in the active site of nitric oxide synthase becomes uncoupled from the oxidation of arginine. This results in the production of radical species that are capable of a direct attack on tetrahydrobiopterin, further depleting its local availability. This feedforward loop may act like a molecular switch, reinforcing low tetrahydrobiopterin levels leading to altered NO signaling, restrained immune effector activity, and perpetual vascular inflammation within the tumor microenvironment. In this review, we discuss the evidence for this underappreciated mechanism in different aspects of tumor progression and therapeutic responses. Furthermore, we discuss the preclinical evidence supporting a clinical role for tetrahydrobiopterin supplementation to enhance immunotherapy and radiotherapy for solid tumors and the potential safety concerns.
Collapse
Affiliation(s)
- Gene Chatman Clark
- Department of Biochemistry, Virginia Commonwealth University, Richmond, VA, United States
- School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Alan Lai
- School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | | | - Zheng Liu
- Department of Human Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Xiang-Yang Wang
- Department of Human Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
- Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
6
|
Li B, Xuan H, Yin Y, Wu S, Du L. The N 6-methyladenosine modification in pathologic angiogenesis. Life Sci 2024; 339:122417. [PMID: 38244915 DOI: 10.1016/j.lfs.2024.122417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/03/2024] [Accepted: 01/07/2024] [Indexed: 01/22/2024]
Abstract
The vascular system is a vital circulatory network in the human body that plays a critical role in almost all physiological processes. The production of blood vessels in the body is a significant area of interest for researchers seeking to improve their understanding of vascular function and maintain normal vascular operation. However, an excessive or insufficient vascular regeneration process may lead to the development of various ailments such as cancer, eye diseases, and ischemic diseases. Recent preclinical and clinical studies have revealed new molecular targets and principles that may enhance the therapeutic effect of anti-angiogenic strategies. A thorough comprehension of the mechanism responsible for the abnormal vascular growth in disease processes can enable researchers to better target and effectively suppress or treat the disease. N6-methyladenosine (m6A), a common RNA methylation modification method, has emerged as a crucial regulator of various diseases by modulating vascular development. In this review, we will cover how m6A regulates various vascular-related diseases, such as cancer, ocular diseases, neurological diseases, ischemic diseases, emphasizing the mechanism of m6A methylation regulators on angiogenesis during pathological process.
Collapse
Affiliation(s)
- Bin Li
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Hanqin Xuan
- Department of Pathology, the First Affiliated Hospital of Soochow University, Jiangsu, China
| | - Yuye Yin
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Shusheng Wu
- Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu, China.
| | - Longfei Du
- Department of Laboratory Medicine, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
7
|
Thapa K, Khan H, Kaur G, Kumar P, Singh TG. Therapeutic targeting of angiopoietins in tumor angiogenesis and cancer development. Biochem Biophys Res Commun 2023; 687:149130. [PMID: 37944468 DOI: 10.1016/j.bbrc.2023.149130] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 11/12/2023]
Abstract
The formation and progression of tumors in humans are linked to the abnormal development of new blood vessels known as neo-angiogenesis. Angiogenesis is a broad word that encompasses endothelial cell migration, proliferation, tube formation, and intussusception, as well as peri-EC recruitment and extracellular matrix formation. Tumor angiogenesis is regulated by angiogenic factors, out of which some of the most potent angiogenic factors such as vascular endothelial growth factor and Angiopoietins (ANGs) in the body are produced by macrophages and other immune cells within the tumor microenvironment. ANGs have a distinct function in tumor angiogenesis and behavior. ANG1, ANG 2, ANG 3, and ANG 4 are the family members of ANG out of which ANG2 has been extensively investigated owing to its unique role in modifying angiogenesis and its tight association with tumor progression, growth, and invasion/metastasis, which makes it an excellent candidate for therapeutic intervention in human malignancies. ANG modulators have demonstrated encouraging outcomes in the treatment of tumor development, either alone or in conjunction with VEGF inhibitors. Future development of more ANG modulators targeting other ANGs is needed. The implication of ANG1, ANG3, and ANG4 as probable therapeutic targets for anti-angiogenesis treatment in tumor development should be also evaluated. The article has described the role of ANG in tumor angiogenesis as well as tumor growth and the treatment strategies modulating ANGs in tumor angiogenesis as demonstrated in clinical studies. The pharmacological modulation of ANGs and ANG-regulated pathways that are responsible for tumor angiogenesis and cancer development should be evaluated for the development of future molecular therapies.
Collapse
Affiliation(s)
- Komal Thapa
- Chitkara School of Pharmacy, Chitkara University, 174103, Himachal Pradesh, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | - Gagandeep Kaur
- Chitkara School of Pharmacy, Chitkara University, 174103, Himachal Pradesh, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda, 151401, Bathinda, India
| | | |
Collapse
|
8
|
Wang C, Xu J, Zhang Y, Nie G. Emerging nanotechnological approaches to regulating tumor vasculature for cancer therapy. J Control Release 2023; 362:647-666. [PMID: 37703928 DOI: 10.1016/j.jconrel.2023.09.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023]
Abstract
Abnormal angiogenesis stands for one of the most striking manifestations of malignant tumor. The pathologically and structurally abnormal tumor vasculature facilitates a hostile tumor microenvironment, providing an ideal refuge exclusively for cancer cells. The emergence of vascular regulation drugs has introduced a distinctive class of therapeutics capable of influencing nutrition supply and drug delivery efficacy without the need to penetrate a series of physical barriers to reach tumor cells. Nanomedicines have been further developed for more precise regulation of tumor vasculature with the capacity of co-delivering multiple active pharmaceutical ingredients, which overall reduces the systemic toxicity and boosts the therapeutic efficacy of free drugs. Additionally, precise structure design enables the integration of specific functional motifs, such as surface-targeting ligands, droppable shells, degradable framework, or stimuli-responsive components into nanomedicines, which can improve tissue-specific accumulation, enhance tissue penetration, and realize the controlled and stimulus-triggered release of the loaded cargo. This review describes the morphological and functional characteristics of tumor blood vessels and summarizes the pivotal molecular targets commonly used in nanomedicine design, and then highlights the recent cutting-edge advancements utilizing nanotechnologies for precise regulation of tumor vasculature. Finally, the challenges and future directions of this field are discussed.
Collapse
Affiliation(s)
- Chunling Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; Sino-Danish Center for Education and Research, Sino-Danish College of UCAS, Beijing 100190, China
| | - Junchao Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yinlong Zhang
- Sino-Danish Center for Education and Research, Sino-Danish College of UCAS, Beijing 100190, China; School of Nanoscience and Engineering, School of Chemical Science, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; Sino-Danish Center for Education and Research, Sino-Danish College of UCAS, Beijing 100190, China; GBA National Institute for Nanotechnology Innovation, Guangzhou 510530, China.
| |
Collapse
|
9
|
Yahya F, Mohd Bakri M, Hossain MZ, Syed Abdul Rahman SN, Mohammed Alabsi A, Ramanathan A. Combination Treatment of TRPV4 Agonist with Cisplatin Promotes Vessel Normalization in an Animal Model of Oral Squamous Cell Carcinoma. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58091229. [PMID: 36143906 PMCID: PMC9504292 DOI: 10.3390/medicina58091229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022]
Abstract
Background and Objectives: Oral squamous cell carcinoma (OSCC) is the sixth most common malignancy in the world. Transient receptor potential vanilloid 4 (TRPV4) channel has been shown to be involved in angiogenesis in multiple types of tumors. However, not much is known about TRPV4′s involvement in OSCC. Thus, in this study, we investigate the effect of administering a TRPV4 agonist on angiogenesis in OSCC. Materials and Methods: Thirty-six Sprague Dawley (SD) rats were used in this study. 4-nitroquinoline 1-oxide (4NQO) was used to induce OSCC. Cisplatin (an anticancer drug), and GSK1016790A (an agonist for TRPV4) was used in this study. Immunohistochemistry was employed to examine the TRPV4 expression. An RT2 Profiler PCR Array was performed for gene expression analysis of TRPV4, vascular growth factors that correspond directly with angiogenesis, such as angiopoietin (Ang-1 and Ang-2), and tyrosine kinase (Tie-1 and Tie-2) receptors. Tumor vessel maturity was assessed by microvessel density and microvessel-pericyte-coverage index. Results: RT2 profiler PCR array showed significant elevated levels of Ang-1 (2.1-fold change; p < 0.05) and Tie-2 (4.5-fold change; p < 0.05) in OSCC following the administration of a combination of GSK1016790A and cisplatin. Additionally, the combination treatment significantly reduced the microvessel density (p < 0.01) and significantly increased the percentage of microvessels covered with pericytes (p < 0.01) in OSCC. Furthermore, tumor size was significantly reduced (p < 0.05) in rats that received cisplatin alone. The combination treatment also greatly reduced the tumor size; however, the data were not statistically significant. Conclusions: The findings suggest that combining a TRPV4 agonist with cisplatin for treatment of OSCC promote vessels normalization via modulation of Ang-1/Tie-2 pathway.
Collapse
Affiliation(s)
- Farhana Yahya
- Department of Oral and Craniofacial Sciences, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (F.Y.); (S.N.S.A.R.)
| | - Marina Mohd Bakri
- Department of Oral and Craniofacial Sciences, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (F.Y.); (S.N.S.A.R.)
- Correspondence:
| | - Mohammad Zakir Hossain
- Department of Oral Physiology, School of Dentistry, Matsumoto Dental University, 1780 Gobara Hirooka, Shiojiri, Nagano 399-0781, Japan;
| | - Syarifah Nur Syed Abdul Rahman
- Department of Oral and Craniofacial Sciences, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (F.Y.); (S.N.S.A.R.)
| | - Aied Mohammed Alabsi
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Jenjarom 42610, Malaysia;
| | - Anand Ramanathan
- Department of Oral and Maxillofacial Clinical Sciences, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
- Oral Cancer Research and Coordinating Center, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| |
Collapse
|
10
|
Gioelli N, Neilson LJ, Wei N, Villari G, Chen W, Kuhle B, Ehling M, Maione F, Willox S, Brundu S, Avanzato D, Koulouras G, Mazzone M, Giraudo E, Yang XL, Valdembri D, Zanivan S, Serini G. Neuropilin 1 and its inhibitory ligand mini-tryptophanyl-tRNA synthetase inversely regulate VE-cadherin turnover and vascular permeability. Nat Commun 2022; 13:4188. [PMID: 35858913 PMCID: PMC9300702 DOI: 10.1038/s41467-022-31904-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 07/08/2022] [Indexed: 11/09/2022] Open
Abstract
The formation of a functional blood vessel network relies on the ability of endothelial cells (ECs) to dynamically rearrange their adhesive contacts in response to blood flow and guidance cues, such as vascular endothelial growth factor-A (VEGF-A) and class 3 semaphorins (SEMA3s). Neuropilin 1 (NRP1) is essential for blood vessel development, independently of its ligands VEGF-A and SEMA3, through poorly understood mechanisms. Grounding on unbiased proteomic analysis, we report here that NRP1 acts as an endocytic chaperone primarily for adhesion receptors on the surface of unstimulated ECs. NRP1 localizes at adherens junctions (AJs) where, interacting with VE-cadherin, promotes its basal internalization-dependent turnover and favors vascular permeability initiated by histamine in both cultured ECs and mice. We identify a splice variant of tryptophanyl-tRNA synthetase (mini-WARS) as an unconventionally secreted extracellular inhibitory ligand of NRP1 that, by stabilizing it at the AJs, slows down both VE-cadherin turnover and histamine-elicited endothelial leakage. Thus, our work shows a role for NRP1 as a major regulator of AJs plasticity and reveals how mini-WARS acts as a physiological NRP1 inhibitory ligand in the control of VE-cadherin endocytic turnover and vascular permeability.
Collapse
Affiliation(s)
- Noemi Gioelli
- Department of Oncology, University of Torino School of Medicine, Candiolo (TO), Italy
- Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Candiolo (TO), Italy
| | | | - Na Wei
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Giulia Villari
- Department of Oncology, University of Torino School of Medicine, Candiolo (TO), Italy
- Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Candiolo (TO), Italy
| | - Wenqian Chen
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Bernhard Kuhle
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Manuel Ehling
- Center for Cancer Biology, Department of Oncology, University of Leuven, Leuven, 3000, Belgium
- Center for Cancer Biology, VIB, Leuven, 3000, Belgium
| | - Federica Maione
- Department of Oncology, University of Torino School of Medicine, Candiolo (TO), Italy
- Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Candiolo (TO), Italy
| | - Sander Willox
- Center for Cancer Biology, Department of Oncology, University of Leuven, Leuven, 3000, Belgium
- Center for Cancer Biology, VIB, Leuven, 3000, Belgium
| | - Serena Brundu
- Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Candiolo (TO), Italy
- Department of Science and Drug Technology, University of Torino, Torino, Italy
| | - Daniele Avanzato
- Department of Oncology, University of Torino School of Medicine, Candiolo (TO), Italy
- Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Candiolo (TO), Italy
| | | | - Massimiliano Mazzone
- Center for Cancer Biology, Department of Oncology, University of Leuven, Leuven, 3000, Belgium
- Center for Cancer Biology, VIB, Leuven, 3000, Belgium
- Department of Science and Drug Technology, University of Torino, Torino, Italy
- Molecular Biotechnology Center (MBC), University of Torino, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Enrico Giraudo
- Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Candiolo (TO), Italy
- Department of Science and Drug Technology, University of Torino, Torino, Italy
| | - Xiang-Lei Yang
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Donatella Valdembri
- Department of Oncology, University of Torino School of Medicine, Candiolo (TO), Italy
- Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Candiolo (TO), Italy
| | - Sara Zanivan
- Cancer Research UK Beatson Institute, Glasgow, UK.
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| | - Guido Serini
- Department of Oncology, University of Torino School of Medicine, Candiolo (TO), Italy.
- Candiolo Cancer Institute - Fondazione del Piemonte per l'Oncologia (FPO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Candiolo (TO), Italy.
| |
Collapse
|
11
|
Schiffmann LM, Bruns CJ, Schmidt T. Resistance Mechanisms of the Metastatic Tumor Microenvironment to Anti-Angiogenic Therapy. Front Oncol 2022; 12:897927. [PMID: 35664794 PMCID: PMC9162757 DOI: 10.3389/fonc.2022.897927] [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: 03/16/2022] [Accepted: 04/21/2022] [Indexed: 11/23/2022] Open
Abstract
Angiogenesis describes the formation of blood vessels from an existing vascular network. Anti-angiogenic drugs that target tumor blood vessels have become standard of care in many cancer entities. Though very promising results in preclinical evaluation, anti-angiogenic treatments fell short of expectations in clinical trials. Patients develop resistance over time or are primarily refractory to anti-angiogenic therapies similar to conventional chemotherapy. To further improve efficacy and outcome to these therapies, a deeper understanding of mechanisms that mediate resistance to anti-angiogenic therapies is needed. The field has done tremendous efforts to gain knowledge about how tumors engage tumor cell and microenvironmental mechanisms to do so. This review highlights the current state of knowledge with special focus on the metastatic tumor site and potential therapeutic relevance of this understanding from a translational and clinical perspective.
Collapse
Affiliation(s)
- Lars M. Schiffmann
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | - Thomas Schmidt
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
12
|
Increasing Vascular Response to Injury Improves Tendon Early Healing Outcome in Aged Rats. Ann Biomed Eng 2022; 50:587-600. [PMID: 35303172 PMCID: PMC9107615 DOI: 10.1007/s10439-022-02948-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/09/2022] [Indexed: 11/01/2022]
Abstract
Tendon injuries positively correlate with patient age, as aging has significant effects on tendon homeostatic maintenance and healing potential after injury. Vascularity is also influenced by age, with both clinical and animal studies demonstrating reduced blood flow in aged tissues. However, it is unknown how aging effects vascularity following tendon injury, and if this vascular response can be modulated through the delivery of angiogenic factors. Therefore, the objective of this study is to evaluate the vascular response following Achilles tendon injury in adult and aged rats, and to define the alterations to tendon healing in an aged model following injection of angiogenic factors. It was determined that aged rat Achilles tendons have a reduced angiogenesis following injury. Further, the delivery of vascular endothelial growth factor, VEGF, caused an increase in vascular response to tendon injury and improved mechanical outcome in this aged population. This work suggests that reduced angiogenic potential with aging may be contributing to impaired tendon healing response and that the delivery of angiogenic factors can rescue this impaired response. This study was also the first to relate changes in vascular response in an aged model using in vivo measures of blood perfusion to alterations in healing properties.
Collapse
|
13
|
Wang Y, Zhang S, Song Z, Ouyang L, Li Y. Anti-Angiogenesis Maintenance Therapy in Newly Diagnosed and Relapsed Ovarian Cancer: A Meta-analysis of Phase III Randomized Controlled Trials. Front Pharmacol 2021; 12:726278. [PMID: 34867330 PMCID: PMC8636101 DOI: 10.3389/fphar.2021.726278] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/29/2021] [Indexed: 11/13/2022] Open
Abstract
Aim: Anti-angiogenesis agents have been added as maintenance therapy in ovarian cancer over the past decade. The aim of this meta-analysis was to analyze the efficacy of anti-angiogenesis therapy in newly diagnosed and relapsed ovarian cancer. Methods: PubMed, Embase, and Cochrane databases were searched for all phase III randomized controlled trials (RCTs) that assessed the efficacy and toxicity of anti-angiogenesis agents in ovarian cancer. Overall survival (OS) and progression-free survival (PFS) were used to evaluate the effectiveness of anti-angiogenesis therapy in ovarian cancer. Results: A total of 6097 patients with newly diagnosed ovarian cancer from 5 phase III RCTs and 2943 patients with relapsed ovarian cancer from 6 phase III RCTs were included in this meta-analysis. The pooled results showed that anti-angiogenesis maintenance therapy significantly improved PFS (hazard ratio [HR], 0.84; 95% confidence interval [CI], 0.76-0.93; p = 0.001), but not OS (HR, 0.98; 95% CI, 0.91-1.05; p = 0.49) compared with placebo in patients with newly diagnosed ovarian cancer. In patients with relapsed ovarian cancer, the pooled results showed a significant improvement on OS (HR, 0.89; 95% CI, 0.82-0.98; p = 0.02) and PFS (HR, 0.61; 95% CI, 0.52-0.72; p < 0.001). The pooled results also showed that the anti-angiogenesis agents were associated with an increase in the occurrence of severe hypertension, neutropenia, diarrhea, thrombocytopenia, headache, and bleeding in ovarian cancer. However, infrequent fatal adverse events occurred in the anti-angiogenesis groups. Conclusions: Study results suggest that anti-angiogenesis agents were an effective therapy for newly diagnosed and relapsed ovarian cancer, especially for relapsed ovarian cancer. Anti-angiogenesis agents may be associated with some severe but not fatal adverse events. Systematic Review Registration: https://www.crd.york.ac.uk/prospero/, identifier CRD42021283647.
Collapse
Affiliation(s)
- Yizi Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shitai Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zixuan Song
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ling Ouyang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yan Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
14
|
Multifaceted Pathomolecular Mechanism of a VWF Large Deletion Involved in the Pathogenesis of Severe VWD. Blood Adv 2021; 6:1038-1053. [PMID: 34861678 PMCID: PMC8945295 DOI: 10.1182/bloodadvances.2021005895] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/21/2021] [Indexed: 11/20/2022] Open
Abstract
The present study demonstrates the dominant-negative impact of an in-frame large deletion on VWF biosynthesis and biogenesis of the WPBs. The malformed WPBs/altered trafficking of its inflammatory cargos cause distresses in endothelial cell signaling pathways and phenotype.
An in-frame heterozygous large deletion of exons 4 through 34 of the von Willebrand factor (VWF) gene was identified in a type 3 von Willebrand disease (VWD) index patient (IP), as the only VWF variant. The IP exhibited severe bleeding episodes despite prophylaxis treatment, with a short VWF half-life after infusion of VWF/factor VIII concentrates. Transcript analysis confirmed transcription of normal VWF messenger RNA besides an aberrant deleted transcript. The IP endothelial colony-forming cells (ECFCs) exhibited a defect in the VWF multimers and Weibel-Palade bodies (WPBs) biogenesis, although demonstrating normal VWF secretion compared with healthy cells. Immunostaining of IP-ECFCs revealed subcellular mislocalization of WPBs pro-inflammatory cargos angiopoietin-2 (Ang2, nuclear accumulation) and P-selectin. Besides, the RNA-sequencing (RNA-seq) analysis showed upregulation of pro-inflammatory and proangiogenic genes, P-selectin, interleukin 8 (IL-8), IL-6, and GROα, copackaged with VWF into WPBs. Further, whole-transcriptome RNA-seq and subsequent gene ontology (GO) enrichment analysis indicated the most enriched GO-biological process terms among the differentially expressed genes in IP-ECFCs were regulation of cell differentiation, cell adhesion, leukocyte adhesion to vascular endothelial, blood vessel morphogenesis, and angiogenesis, which resemble downstream signaling pathways associated with inflammatory stimuli and Ang2 priming. Accordingly, our functional experiments exhibited an increased endothelial cell adhesiveness and interruption in endothelial cell–cell junctions of the IP-ECFCs. In conclusion, the deleted VWF has a dominant-negative impact on multimer assembly and the biogenesis of WPBs, leading to altered trafficking of their pro-inflammatory cargos uniquely, which, in turn, causes changes in cellular signaling pathways, phenotype, and function of the endothelial cells.
Collapse
|
15
|
Targeting the Ang2/Tie2 Axis with Tanshinone IIA Elicits Vascular Normalization in Ischemic Injury and Colon Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7037786. [PMID: 34804370 PMCID: PMC8598375 DOI: 10.1155/2021/7037786] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/04/2021] [Accepted: 10/15/2021] [Indexed: 02/06/2023]
Abstract
Pathological angiogenesis, as exhibited by aberrant vascular structure and function, has been well deemed to be a hallmark of cancer and various ischemic diseases. Therefore, strategies to normalize vasculature are of potential therapeutic interest in these diseases. Recently, identifying bioactive compounds from medicinal plant extracts to reverse abnormal vasculature has been gaining increasing attention. Tanshinone IIA (Tan IIA), an active component of Salvia miltiorrhiza, has been shown to play significant roles in improving blood circulation and delaying tumor progression. However, the underlying mechanisms responsible for the therapeutic effects of Tan IIA are not fully understood. Herein, we established animal models of HT-29 human colon cancer xenograft and hind limb ischemia to investigate the role of Tan IIA in regulating abnormal vasculature. Interestingly, our results demonstrated that Tan IIA could significantly promote the blood flow, alleviate the hypoxia, improve the muscle quality, and ameliorate the pathological damage after ischemic insult. Meanwhile, we also revealed that Tan IIA promoted the integrity of vascular structure, reduced vascular leakage, and attenuated the hypoxia in HT-29 tumors. Moreover, the circulating angiopoietin 2 (Ang2), which is extremely high in these two pathological states, was substantially depleted in the presence of Tan IIA. Also, the activation of Tie2 was potentiated by Tan IIA, resulting in decreased vascular permeability and elevated vascular integrity. Mechanistically, we uncovered that Tan IIA maintained vascular stability by targeting the Ang2-Tie2-AKT-MLCK cascade. Collectively, our data suggest that Tan IIA normalizes vessels in tumors and ischemic injury via regulating the Ang2/Tie2 signaling pathway.
Collapse
|
16
|
Gotts JE, Maishan M, Chun L, Fang X, Han C, Chiueh V, Khakoo AY, Lee T, Stolina M, Matthay MA. Delayed angiopoietin-2 blockade reduces influenza-induced lung injury and improves survival in mice. Physiol Rep 2021; 9:e15081. [PMID: 34755490 PMCID: PMC8578883 DOI: 10.14814/phy2.15081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/21/2021] [Accepted: 09/25/2021] [Indexed: 11/24/2022] Open
Abstract
Influenza remains a major cause of death and disability with limited treatment options. Studies of acute lung injury have identified angiopoietin-2 (Ang-2) as a key prognostic marker and a potential mediator of Acute respiratory distress syndrome. However, the role of Ang-2 in viral pneumonia remains poorly defined. This study characterized the time course of lung Ang-2 expression in severe influenza pneumonia and tested the therapeutic potential of Ang-2 inhibition. We inoculated adult mice with influenza A (PR8 strain) and measured angiopoietin-1 (Ang-1), Ang-2, and Tie2 expressions during the evolution of inflammatory lung injury over the first 7 days post-infection (dpi). We tested a peptide-antibody inhibitor of Ang-2, L1-7, administered at 2, 4, and 6 dpi and measured arterial oxygen saturation, survival, pulmonary edema, inflammatory cytokines, and viral load. Finally, we infected primary human alveolar type II epithelial (AT2) cells grown in air-liquid interface culture with influenza and measured Ang-2 RNA expression. Influenza caused severe lung injury between 5 and 7 dpi in association with increased Ang-2 lung RNA and a dramatic increase in Ang-2 protein in bronchoalveolar lavage. Inhibition of Ang-2 improved oxygenation and survival and reduced pulmonary edema and alveolar-capillary barrier permeability to protein without major effects on inflammation or viral load. Finally, influenza increased the expression of Ang-2 RNA in human AT2 cells. The increased Ang-2 levels in the airspaces during severe influenza pneumonia and the improvement in clinically relevant outcomes after Ang-2 antagonism suggest that the Ang-1/Ang-2 Tie-2 signaling axis is a promising therapeutic target in influenza and potentially other causes of viral pneumonia.
Collapse
Affiliation(s)
- Jeffrey E. Gotts
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Mazharul Maishan
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Lauren Chun
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Xiaohui Fang
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Chun‐Ya Han
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - Venice Chiueh
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - Aarif Y. Khakoo
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - TaeWeon Lee
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - Marina Stolina
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - Michael A. Matthay
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
17
|
Riggin CN, Rodriguez AB, Weiss SN, Raja HA, Chen M, Schultz SM, Sehgal CM, Soslowsky LJ. Modulation of vascular response after injury in the rat Achilles tendon alters healing capacity. J Orthop Res 2021; 39:2000-2016. [PMID: 32936495 PMCID: PMC7960560 DOI: 10.1002/jor.24861] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 09/02/2020] [Accepted: 09/14/2020] [Indexed: 02/04/2023]
Abstract
Tendons are relatively hypovascular but become hypervascular during both injury and degeneration. This is due to the angiogenic response, or the formation of new blood vessels, to tissue injury. The objective of this study was to evaluate the effect of vascular modulation in the rat Achilles tendons during healing. Fischer rats received a bilateral Achilles incisional injury followed by local injections of vascular endothelial growth factor (VEGF), anti-VEGF antibody (B20.4-1-1), or saline either early or late during the healing process. Vascular modulation and healing were evaluated using multiple in vivo ultrasound imaging modalities, in vivo functional assessment, and ex vivo measures of tendon compositional and mechanical properties. The late delivery of anti-VEGF antibody, B20, caused a temporary reduction in healing capacity during a time point where vascularity was also decreased, and then an improvement during a later time point where vascularity was increased relative to control. However, VEGF delivery had a minimal impact on healing and vascular changes in both early and late delivery times. This study was the first to evaluate vascular changes using both in vivo imaging methods and ex vivo histological methods, as well as functional and mechanical outcomes associated with these vascular changes. Clinical significance: this study demonstrates that the alteration of vascular response through the delivery of angiogenic growth factors has the ability to alter tendon healing properties.
Collapse
Affiliation(s)
- Corinne N Riggin
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, 307A Stemmler Hall, 36 Street & Hamilton Walk, Philadelphia, PA 19104
| | - Ashley B Rodriguez
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, 307A Stemmler Hall, 36 Street & Hamilton Walk, Philadelphia, PA 19104
| | - Stephanie N Weiss
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, 307A Stemmler Hall, 36 Street & Hamilton Walk, Philadelphia, PA 19104
| | - Harina A Raja
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, 307A Stemmler Hall, 36 Street & Hamilton Walk, Philadelphia, PA 19104
| | - Mengcun Chen
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, 307A Stemmler Hall, 36 Street & Hamilton Walk, Philadelphia, PA 19104
| | - Susan M Schultz
- Department of Radiology, University of Pennsylvania, 1 Silverstein, 3400 Spruce Street, Philadelphia, PA 19104
| | - Chandra M Sehgal
- Department of Radiology, University of Pennsylvania, 1 Silverstein, 3400 Spruce Street, Philadelphia, PA 19104
| | - Louis J Soslowsky
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, 307A Stemmler Hall, 36 Street & Hamilton Walk, Philadelphia, PA 19104,Corresponding Author: Louis J Soslowsky, McKay Orthopaedic Research Laboratory, University of Pennsylvania, 307A Stemmler Hall, 36 Street & Hamilton Walk, Philadelphia, PA 19104, , Phone: 215-898-8653, Fax: 215-573-2133
| |
Collapse
|
18
|
Magnussen AL, Mills IG. Vascular normalisation as the stepping stone into tumour microenvironment transformation. Br J Cancer 2021; 125:324-336. [PMID: 33828258 PMCID: PMC8329166 DOI: 10.1038/s41416-021-01330-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 01/17/2021] [Accepted: 02/17/2021] [Indexed: 02/01/2023] Open
Abstract
A functional vascular system is indispensable for drug delivery and fundamental for responsiveness of the tumour microenvironment to such medication. At the same time, the progression of a tumour is defined by the interactions of the cancer cells with their surrounding environment, including neovessels, and the vascular network continues to be the major route for the dissemination of tumour cells in cancer, facilitating metastasis. So how can this apparent conflict be reconciled? Vessel normalisation-in which redundant structures are pruned and the abnormal vasculature is stabilised and remodelled-is generally considered to be beneficial in the course of anti-cancer treatments. A causality between normalised vasculature and improved response to medication and treatment is observed. For this reason, it is important to discern the consequence of vessel normalisation on the tumour microenvironment and to modulate the vasculature advantageously. This article will highlight the challenges of controlled neovascular remodelling and outline how vascular normalisation can shape disease management.
Collapse
Affiliation(s)
- Anette L Magnussen
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Ian G Mills
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK.
- Patrick G Johnston Centre for Cancer Research, Queen's University of Belfast, Belfast, UK.
- Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway.
- Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
19
|
Khan KA, Wu FTH, Cruz‐Munoz W, Kerbel RS. Ang2 inhibitors and Tie2 activators: potential therapeutics in perioperative treatment of early stage cancer. EMBO Mol Med 2021; 13:e08253. [PMID: 34125494 PMCID: PMC8261516 DOI: 10.15252/emmm.201708253] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Anti-angiogenic drugs targeting the VEGF pathway are most effective in advanced metastatic disease settings of certain types of cancers, whereas they have been unsuccessful as adjuvant therapies of micrometastatic disease in numerous phase III trials involving early-stage (resectable) cancers. Newer investigational anti-angiogenic drugs have been designed to inhibit the Angiopoietin (Ang)-Tie pathway. Acting through Tie2 receptors, the Ang1 ligand is a gatekeeper of endothelial quiescence. Ang2 is a dynamically expressed pro-angiogenic destabilizer of endothelium, and its upregulation is associated with poor prognosis in cancer. Besides using Ang2 blockers as inhibitors of tumor angiogenesis, little attention has been paid to their use as stabilizers of blood vessels to suppress tumor cell extravasation and metastasis. In clinical trials, Ang2 blockers have shown limited efficacy in advanced metastatic disease settings. This review summarizes preclinical evidence suggesting the potential utility of Ang2 inhibitors or Tie2 activators as neoadjuvant or adjuvant therapies in the prevention or treatment of early-stage micrometastatic disease. We further discuss the rationale and potential of combining these strategies with immunotherapy, including immune checkpoint targeting antibodies.
Collapse
Affiliation(s)
- Kabir A Khan
- Department of Medical BiophysicsUniversity of TorontoTorontoONCanada
- Biological Sciences PlatformSunnybrook Research InstituteTorontoONCanada
| | - Florence TH Wu
- Department of Medical BiophysicsUniversity of TorontoTorontoONCanada
- Biological Sciences PlatformSunnybrook Research InstituteTorontoONCanada
| | - William Cruz‐Munoz
- Department of Medical BiophysicsUniversity of TorontoTorontoONCanada
- Biological Sciences PlatformSunnybrook Research InstituteTorontoONCanada
| | - Robert S Kerbel
- Department of Medical BiophysicsUniversity of TorontoTorontoONCanada
- Biological Sciences PlatformSunnybrook Research InstituteTorontoONCanada
| |
Collapse
|
20
|
Yuan K, Agarwal S, Chakraborty A, Condon DF, Patel H, Zhang S, Huang F, Mello SA, Kirk OI, Vasquez R, de Jesus Perez VA. Lung Pericytes in Pulmonary Vascular Physiology and Pathophysiology. Compr Physiol 2021; 11:2227-2247. [PMID: 34190345 PMCID: PMC10507675 DOI: 10.1002/cphy.c200027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pericytes are mesenchymal-derived mural cells localized within the basement membrane of pulmonary and systemic capillaries. Besides structural support, pericytes control vascular tone, produce extracellular matrix components, and cytokines responsible for promoting vascular homeostasis and angiogenesis. However, pericytes can also contribute to vascular pathology through the production of pro-inflammatory and pro-fibrotic cytokines, differentiation into myofibroblast-like cells, destruction of the extracellular matrix, and dissociation from the vessel wall. In the lung, pericytes are responsible for maintaining the integrity of the alveolar-capillary membrane and coordinating vascular repair in response to injury. Loss of pericyte communication with alveolar capillaries and a switch to a pro-inflammatory/pro-fibrotic phenotype are common features of lung disorders associated with vascular remodeling, inflammation, and fibrosis. In this article, we will address how to differentiate pericytes from other cells, discuss the molecular mechanisms that regulate the interactions of pericytes and endothelial cells in the pulmonary circulation, and the experimental tools currently used to study pericyte biology both in vivo and in vitro. We will also discuss evidence that links pericytes to the pathogenesis of clinically relevant lung disorders such as pulmonary hypertension, idiopathic lung fibrosis, sepsis, and SARS-COVID. Future studies dissecting the complex interactions of pericytes with other pulmonary cell populations will likely reveal critical insights into the origin of pulmonary diseases and offer opportunities to develop novel therapeutics to treat patients afflicted with these devastating disorders. © 2021 American Physiological Society. Compr Physiol 11:2227-2247, 2021.
Collapse
Affiliation(s)
- Ke Yuan
- Division of Respiratory Diseases Research, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Stuti Agarwal
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Ananya Chakraborty
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - David F. Condon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Hiral Patel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Serena Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Flora Huang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Salvador A. Mello
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | | | - Rocio Vasquez
- University of Central Florida, Orlando, Florida, USA
| | - Vinicio A. de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
21
|
Sun R, Kong X, Qiu X, Huang C, Wong PP. The Emerging Roles of Pericytes in Modulating Tumor Microenvironment. Front Cell Dev Biol 2021; 9:676342. [PMID: 34179005 PMCID: PMC8232225 DOI: 10.3389/fcell.2021.676342] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/06/2021] [Indexed: 12/17/2022] Open
Abstract
Pericytes (PCs), known as mural cells, play an important blood vessel (BV) supporting role in regulating vascular stabilization, permeability and blood flow in microcirculation as well as blood brain barrier. In carcinogenesis, defective interaction between PCs and endothelial cells (ECs) contributes to the formation of leaky, chaotic and dysfunctional vasculature in tumors. However, recent works from other laboratories and our own demonstrate that the direct interaction between PCs and other stromal cells/cancer cells can modulate tumor microenvironment (TME) to favor cancer growth and progression, independent of its BV supporting role. Furthermore, accumulating evidence suggests that PCs have an immunomodulatory role. In the current review, we focus on recent advancement in understanding PC's regulatory role in the TME by communicating with ECs, immune cells, and tumor cells, and discuss how we can target PC's functions to re-model TME for an improved cancer treatment strategy.
Collapse
Affiliation(s)
- Ruipu Sun
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiangzhan Kong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyi Qiu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Cheng Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ping-Pui Wong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
22
|
Vascular-immuno-phenotypic (VIP) model for locally advanced and oligo-metastatic cancer: A hypothesis. Med Hypotheses 2021; 152:110618. [PMID: 34102599 DOI: 10.1016/j.mehy.2021.110618] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/11/2021] [Accepted: 05/25/2021] [Indexed: 01/18/2023]
Abstract
Primary Hypothesis: In cancer therapy, normalization of the vasculature, and not disruption, to facilitate the reversal of the immuno-phenotypic changes, is the sine-qua-non for cancer elimination. The triad of normalization of the vasculature, leading to the improved immunological tumour microenvironment and increased susceptibility of resistant phenotypic cancer cells (VIP model), forms the basis of this hypothesis. This article hypothesizes the absolute need for vascular normalization for the eradication of cancer. Locally advanced and oligometastatic cancers have the potential to be cured with aggressive therapy. The focus on vascular normalization its clinical relevance in this situation is essential. Most traditional approaches have focused on the elimination of cancer by targeting and disrupting vasculature. Initially, antiangiogenic drugs showed significant promise in animal experiments. However, this vascular disruption approach has not paid the expected long-term dividends in the clinical setup. However, antiangiogenics are playing a significant role when used concurrently with chemotherapy/immunotherapy. Antiangiogenics have dual temporal actions - an initial normalization effect with improved oxygenation followed by pruning of blood vessels, resulting in exaggerated hypoxia along with a rebound progression. The literature is replete with phenomena of initial vascular normalization with a paradigm shift in the immuno-phenotypic milieu of cancer as part of vascular targeting approaches. The hypothesis in this article stresses the need to have strategies to extend this normalization window or to have pre-clinical trials to optimize the dose scheduling of antiangiogenics cyclically along with chemo/targeted/immune therapy and other combination therapies. We can implement this hypothesis by a combinatorial harmonization of present-day cancer therapies in the setting of tumor vasculature integrity. In addition, based on the proposed hypothesis, the current normalization effect of antiangiogenics and newer therapy development should focus primarily on normalization of the vasculature as well as targeting hypoxia-Inducible-factor-1 alpha (HIF-1 α) in the presence of differential genetic modulation of vascular endothelial cell resistance enhancement along with cancer cell sensitization. Also, the article enumerates six supporting hypotheses supplementing the primary hypothesis.
Collapse
|
23
|
Samson F, Fabunmi TE, Patrick AT, Jee D, Gutsaeva DR, Jahng WJ. Fatty Acid Composition and Stoichiometry Determine the Angiogenesis Microenvironment. ACS OMEGA 2021; 6:5953-5961. [PMID: 33681633 PMCID: PMC7931378 DOI: 10.1021/acsomega.1c00196] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/08/2021] [Indexed: 05/11/2023]
Abstract
The current study tested the hypothesis of whether specific lipids may control angiogenic reactions. Using the chorioallantoic membrane assay of the chick embryo, new vessel formation was analyzed quantitatively by gas chromatography and mass spectrometry as well as bioinformatics tools including an angiogenesis analyzer. Our biochemical experiments showed that a specific lipid composition and stoichiometry determine the angiogenesis microenvironment to accelerate or inhibit vessel formation. Specific lipids of angiogenesis determinants in the vessel area and the non-vessel area were identified as nitrooleic acid, docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), palmitic acid, oleic acid, linoleic acid, linolenic acid, epoxyoleic acid, lysophosphatidylcholine (LPC), cholesterol, 7-ketocholesterol, and docosahexaenoyl lysophosphatidylcholine (DHA-LPC). Vessel formation happens on the surface area of the hydrophilic membrane of the yolk. Our biochemical data demonstrated that angiogenesis was followed in the white lipid complex area to generate more branches, junctions, segments, and extremities. We analyzed lipid fragments in the vessel, non-vessel, and albumen area to show that each area contains a specific lipid composition and stoichiometry. Mass spectrometry data demonstrated that the vessel area has higher concentrations of nitrooleic acid, palmitic acid, stearic acid, LPC, lysophosphatidylethanolamine, cholesterol, oleic acid, linoleic acid, 7-ketocholesterol, and DHA-LPC; however, DHA and EPA were abundant in the hydrophobic non-vessel area. The purpose of vessel formation is to wrap up the yolk area to transport nutrients including specific fatty acids. Besides, angiogenesis requires aqueous albumen shown by distance-dependent vessel formation from albumen and oxygen. Higher concentrations of fatty acids are required for energy and carbon structure from the carbon-carbon bond, membrane building blocks, and amphiphilic detergent to solubilize a hydrophobic environment in the aqueous blood layer. The current study may guide that the uncovered hydrophobic or zwitterionic molecules such as DHA and DHA-LPC may control angiogenesis as antiangiogenic or proangiogenic molecules as potential drug targets for treating uncontrolled angiogenesis-related diseases, including diabetic retinopathy and age-related macular degeneration.
Collapse
Affiliation(s)
| | - Tosin Esther Fabunmi
- Department
of Petroleum Chemistry, American University
of Nigeria, Yola 640101, Nigeria
| | - Ambrose Teru Patrick
- Department
of Petroleum Chemistry, American University
of Nigeria, Yola 640101, Nigeria
| | - Donghyun Jee
- Department
of Ophthalmology and Visual Science, St. Vincent’s Hospital,
College of Medicine, The Catholic University
of Korea, Suwon 16247, Korea
| | - Diana R. Gutsaeva
- Department
of Ophthalmology, Augusta University, Augusta, Georgia 30912, United States
| | - Wan Jin Jahng
- Department
of Petroleum Chemistry, American University
of Nigeria, Yola 640101, Nigeria
- . Phone: +234-805-550-1032
| |
Collapse
|
24
|
Zhang Y, Cedervall J, Hamidi A, Herre M, Viitaniemi K, D'Amico G, Miao Z, Unnithan RVM, Vaccaro A, van Hooren L, Georganaki M, Thulin Å, Qiao Q, Andrae J, Siegbahn A, Heldin CH, Alitalo K, Betsholtz C, Dimberg A, Olsson AK. Platelet-Specific PDGFB Ablation Impairs Tumor Vessel Integrity and Promotes Metastasis. Cancer Res 2020; 80:3345-3358. [PMID: 32586981 DOI: 10.1158/0008-5472.can-19-3533] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 04/24/2020] [Accepted: 06/17/2020] [Indexed: 11/16/2022]
Abstract
Platelet-derived growth factor B (PDGFB) plays a crucial role in recruitment of PDGF receptor β-positive pericytes to blood vessels. The endothelium is an essential source of PDGFB in this process. Platelets constitute a major reservoir of PDGFB and are continuously activated in the tumor microenvironment, exposing tumors to the plethora of growth factors contained in platelet granules. Here, we show that tumor vascular function, as well as pericyte coverage is significantly impaired in mice with conditional knockout of PDGFB in platelets. A lack of PDGFB in platelets led to enhanced hypoxia and epithelial-to-mesenchymal transition in the primary tumors, elevated levels of circulating tumor cells, and increased spontaneous metastasis to the liver or lungs in two mouse models. These findings establish a previously unknown role for platelet-derived PDGFB, whereby it promotes and maintains vascular integrity in the tumor microenvironment by contributing to the recruitment of pericytes. SIGNIFICANCE: Conditional knockout of PDGFB in platelets demonstrates its previously unknown role in the maintenance of tumor vascular integrity and host protection against metastasis.
Collapse
Affiliation(s)
- Yanyu Zhang
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden
| | - Jessica Cedervall
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden
| | - Anahita Hamidi
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden
| | - Melanie Herre
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden
| | - Kati Viitaniemi
- Wihuri Research Institute and Translational Cancer Medicine Research Program, Biomedicum Helsinki, 00014 University of Helsinki, Yliopistonkatu, Helsinki, Finland
| | - Gabriela D'Amico
- Wihuri Research Institute and Translational Cancer Medicine Research Program, Biomedicum Helsinki, 00014 University of Helsinki, Yliopistonkatu, Helsinki, Finland
| | - Zuoxiu Miao
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden
| | - Ragaseema Valsala Madhavan Unnithan
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden.,Department of Biotechnology, Govt. Arts College, Thiruvananthapuram, India
| | - Alessandra Vaccaro
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - Luuk van Hooren
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - Maria Georganaki
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - Åsa Thulin
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Qi Qiao
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden
| | - Johanna Andrae
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - Agneta Siegbahn
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Carl-Henrik Heldin
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Medicine Research Program, Biomedicum Helsinki, 00014 University of Helsinki, Yliopistonkatu, Helsinki, Finland
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden.,ICMC (Integrated Cardio Metabolic Centre), Karolinska Institutet, Novum, Blickagången 6, Huddinge, Sweden
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden.
| |
Collapse
|
25
|
Andreuzzi E, Capuano A, Poletto E, Pivetta E, Fejza A, Favero A, Doliana R, Cannizzaro R, Spessotto P, Mongiat M. Role of Extracellular Matrix in Gastrointestinal Cancer-Associated Angiogenesis. Int J Mol Sci 2020; 21:E3686. [PMID: 32456248 PMCID: PMC7279269 DOI: 10.3390/ijms21103686] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal tumors are responsible for more cancer-related fatalities than any other type of tumors, and colorectal and gastric malignancies account for a large part of these diseases. Thus, there is an urgent need to develop new therapeutic approaches to improve the patients' outcome and the tumor microenvironment is a promising arena for the development of such treatments. In fact, the nature of the microenvironment in the different gastrointestinal tracts may significantly influence not only tumor development but also the therapy response. In particular, an important microenvironmental component and a potential therapeutic target is the vasculature. In this context, the extracellular matrix is a key component exerting an active effect in all the hallmarks of cancer, including angiogenesis. Here, we summarized the current knowledge on the role of extracellular matrix in affecting endothelial cell function and intratumoral vascularization in the context of colorectal and gastric cancer. The extracellular matrix acts both directly on endothelial cells and indirectly through its remodeling and the consequent release of growth factors. We envision that a deeper understanding of the role of extracellular matrix and of its remodeling during cancer progression is of chief importance for the development of new, more efficacious, targeted therapies.
Collapse
Affiliation(s)
- Eva Andreuzzi
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Alessandra Capuano
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Evelina Poletto
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Eliana Pivetta
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Albina Fejza
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Andrea Favero
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Roberto Doliana
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Renato Cannizzaro
- Department of Clinical Oncology, Experimental Gastrointestinal Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy;
| | - Paola Spessotto
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Maurizio Mongiat
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| |
Collapse
|
26
|
Tumor Endothelial Cell-A Biological Tool for Translational Cancer Research. Int J Mol Sci 2020; 21:ijms21093238. [PMID: 32375250 PMCID: PMC7247330 DOI: 10.3390/ijms21093238] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 12/14/2022] Open
Abstract
Going from bench to bedside is a simplified description of translational research, with the ultimate goal being to improve the health status of mankind. Tumor endothelial cells (TECs) perform angiogenesis to support the growth, establishment, and dissemination of tumors to distant organs. TECs have various features that distinguish them from normal endothelial cells, which include alterations in gene expression patterns, higher angiogenic and metabolic activities, and drug resistance tendencies. The special characteristics of TECs enhance the vulnerability of tumor blood vessels toward antiangiogenic therapeutic strategies. Therefore, apart from being a viable therapeutic target, TECs would act as a better mediator between the bench (i.e., angiogenesis research) and the bedside (i.e., clinical application of drugs discovered through research). Exploitation of TEC characteristics could reveal unidentified strategies of enhancing and monitoring antiangiogenic therapy in the treatment of cancer, which are discussed in this review.
Collapse
|
27
|
Okonkwo UA, Chen L, Ma D, Haywood VA, Barakat M, Urao N, DiPietro LA. Compromised angiogenesis and vascular Integrity in impaired diabetic wound healing. PLoS One 2020; 15:e0231962. [PMID: 32324828 PMCID: PMC7179900 DOI: 10.1371/journal.pone.0231962] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 04/03/2020] [Indexed: 12/19/2022] Open
Abstract
Vascular deficits are a fundamental contributing factor of diabetes-associated diseases. Although previous studies have demonstrated that the pro-angiogenic phase of wound healing is blunted in diabetes, a comprehensive understanding of the mechanisms that regulate skin revascularization and capillary stabilization in diabetic wounds is lacking. Using a mouse model of diabetic wound healing, we performed microCT analysis of the 3-dimensional architecture of the capillary bed. As compared to wild type, vessel surface area, branch junction number, total vessel length, and total branch number were significantly decreased in wounds of diabetic mice as compared to WT mice. Diabetic mouse wounds also had significantly increased capillary permeability and decreased pericyte coverage of capillaries. Diabetic wounds exhibited significant perturbations in the expression of factors that affect vascular regrowth, maturation and stability. Specifically, the expression of VEGF-A, Sprouty2, PEDF, LRP6, Thrombospondin 1, CXCL10, CXCR3, PDGFR-β, HB-EGF, EGFR, TGF-β1, Semaphorin3a, Neuropilin 1, angiopoietin 2, NG2, and RGS5 were down-regulated in diabetic wounds. Together, these studies provide novel information about the complexity of the perturbation of angiogenesis in diabetic wounds. Targeting factors responsible for wound resolution and vascular pruning, as well those that affect pericyte recruitment, maturation, and stability may have the potential to improve diabetic skin wound healing.
Collapse
Affiliation(s)
- Uzoagu A. Okonkwo
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Da Ma
- Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Guanghua School of Stomatology, SunYat-sen University, Guangzhou, Guangdong, China
| | - Veronica A. Haywood
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL, United States of America
| | - May Barakat
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Norifumi Urao
- Department of Pharmacology, Upstate Medical University, Syracuse, NY, United States of America
| | - Luisa A. DiPietro
- Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL, United States of America
- * E-mail:
| |
Collapse
|
28
|
Li S, Zhang Q, Hong Y. Tumor Vessel Normalization: A Window to Enhancing Cancer Immunotherapy. Technol Cancer Res Treat 2020; 19:1533033820980116. [PMID: 33287656 PMCID: PMC7727091 DOI: 10.1177/1533033820980116] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/13/2020] [Accepted: 10/30/2020] [Indexed: 01/05/2023] Open
Abstract
Hostile microenvironment produced by abnormal blood vessels, which is characterized by hypoxia, low pH value and increasing interstitial fluid pressure, would facilitate tumor progression, metastasis, immunosuppression and anticancer treatments resistance. These abnormalities are the result of the imbalance of pro-angiogenic and anti-angiogenic factors (such as VEGF and angiopoietin 2, ANG2). Prudent use of anti-angiogenesis drugs would normalize these aberrant tumor vessels, resulting in a transient window of vessel normalization. In addition, use of cancer immunotherapy including immune checkpoint blockers when vessel normalization is achieved brings better outcomes. In this review, we sum up the advances in the field of understanding and application of the concept of tumor vessels normalization window to treat cancer. Moreover, we also outline some challenges and opportunities ahead to optimize the combination of anti-angiogenic agents and immunotherapy, leading to improve patients' outcomes.
Collapse
Affiliation(s)
- Sai Li
- Department of gynecologic oncology, Women’s hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yupeng Hong
- Department of Oncology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
29
|
Zhang SS, Hu JQ, Liu XH, Chen LX, Chen H, Guo XH, Huang QB. Role of Moesin Phosphorylation in Retinal Pericyte Migration and Detachment Induced by Advanced Glycation Endproducts. Front Endocrinol (Lausanne) 2020; 11:603450. [PMID: 33312163 PMCID: PMC7708375 DOI: 10.3389/fendo.2020.603450] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 10/19/2020] [Indexed: 01/22/2023] Open
Abstract
Proliferative diabetic retinopathy (PDR) involves persistent, uncontrolled formation of premature blood vessels with reduced number of pericytes. Our previous work showed that advanced glycation endproducts (AGEs) induced angiogenesis in human umbilical vein endothelial cells, mouse retina, and aortic ring, which was associated with moesin phosphorylation. Here we investigated whether moesin phosphorylation may contribute to pericyte detachment and the development of PDR. Primary retinal microvascular pericytes (RMPs) were isolated, purified from weanling rats, and identified by cellular markers α-SMA, PDGFR-β, NG2, and desmin using immunofluorescence microscopy. Effects of AGE-BSA on proliferation and migration of RMPs were examined using CCK-8, wound healing, and transwell assays. Effects on moesin phosphorylation were examined using western blotting. The RMP response to AGE-BSA was also examined when cells expressed the non-phosphorylatable Thr558Ala mutant or phospho-mimicking Thr558Asp mutant of moesin or were treated with ROCK inhibitor Y27632. Colocalization and interaction between CD44, phospho-moesin, and F-actin were observed. Experiments with cultured primary RMPs showed that AGE-BSA inhibited the proliferation, enhanced the migration, and increased moesin phosphorylation in a dose- and time-dependent manner. AGE-BSA also triggered the rearrangement of F-actin and promoted the interaction of CD44 with phospho-moesin in RMPs. These effects were abrogated in cells expressing the non-phosphorylatable moesin mutant and the application of ROCK inhibitor Y27632 attenuated AGE-induced alteration in cultured RMPs by abolishing the phosphorylation of moesin. However, those AGE-induced pathological process occurred in RMPs expressed the phospho-mimicking moesin without AGE-BSA treatment. It is concluded that AGEs could activate ROCK to mediate moesin phosphorylation at Thr558, and resulting phospho-moesin interacts with CD44 to form CD44 cluster, which might stimulate the migration of RMPs and subsequent RMP detachment in microvessel. This pathway may provide new drug targets against immature neovessel formation in PDR.
Collapse
Affiliation(s)
- Shuang-Shuang Zhang
- Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jia-Qing Hu
- Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiao-Hui Liu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Li-Xian Chen
- Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hong Chen
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiao-Hua Guo
- Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qiao-Bing Huang
- Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Trauma Care Center, Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- *Correspondence: Qiao-Bing Huang,
| |
Collapse
|
30
|
Arai H, Battaglin F, Wang J, Lo JH, Soni S, Zhang W, Lenz HJ. Molecular insight of regorafenib treatment for colorectal cancer. Cancer Treat Rev 2019; 81:101912. [PMID: 31715423 PMCID: PMC7491975 DOI: 10.1016/j.ctrv.2019.101912] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 12/22/2022]
Abstract
Regorafenib is a multi-targeting kinase inhibitor approved for the treatment of metastatic colorectal cancer patients in refractory to standard chemotherapy. Similarly to sorafenib, this agent was originally developed as a RAF1 inhibitor. However, the kinase inhibitory profile is distinct from sorafenib. A broad-spectrum of kinase inhibition induces wide-range drug sensitivity, irrespective of mutation status of major oncogenes. This agent's main therapeutic effects are anti-angiogenesis and the remodeling of tumor microenvironment through several mechanisms of action. The dual blockade of VEGF receptors and TIE2 can lead to both additive anti-angiogenesis effects and the suggestive unique regulation of vessel stability. Additionally, it inhibits molecular escape pathways to VEGF inhibition (e.g., FGF, PIGF, and PDGF signaling), enabling its continuous antiangiogenic effect even in tumors resistant to VEGF inhibitors. Furthermore, regorafenib has the important effect of enhancing anti-tumor immunity via macrophage modulation. Based on this concept, clinical trials have been recently launched for the development of a combination strategy with immune checkpoint inhibitors. Contrary to regorafenib induced clinical benefits and advances in the novel strategy, currently no predictive biomarkers have been identified. In the present review, we revisit and summarize regorafenib's unique mechanisms of action. The review could highlight molecular insights and provide some perspective for the search of predictive biomarkers used in metastatic colorectal cancer patients treated with regorafenib.
Collapse
Affiliation(s)
- Hiroyuki Arai
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlate Avenue, Los Angeles, CA 90033, United States
| | - Francesca Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlate Avenue, Los Angeles, CA 90033, United States.
| | - Jingyuan Wang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlate Avenue, Los Angeles, CA 90033, United States.
| | - Jae Ho Lo
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlate Avenue, Los Angeles, CA 90033, United States.
| | - Shivani Soni
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlate Avenue, Los Angeles, CA 90033, United States
| | - Wu Zhang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlate Avenue, Los Angeles, CA 90033, United States.
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlate Avenue, Los Angeles, CA 90033, United States.
| |
Collapse
|
31
|
Payne LB, Zhao H, James CC, Darden J, McGuire D, Taylor S, Smyth JW, Chappell JC. The pericyte microenvironment during vascular development. Microcirculation 2019; 26:e12554. [PMID: 31066166 PMCID: PMC6834874 DOI: 10.1111/micc.12554] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 04/29/2019] [Accepted: 05/03/2019] [Indexed: 12/22/2022]
Abstract
Vascular pericytes provide critical contributions to the formation and integrity of the blood vessel wall within the microcirculation. Pericytes maintain vascular stability and homeostasis by promoting endothelial cell junctions and depositing extracellular matrix (ECM) components within the vascular basement membrane, among other vital functions. As their importance in sustaining microvessel health within various tissues and organs continues to emerge, so does their role in a number of pathological conditions including cancer, diabetic retinopathy, and neurological disorders. Here, we review vascular pericyte contributions to the development and remodeling of the microcirculation, with a focus on the local microenvironment during these processes. We discuss observations of their earliest involvement in vascular development and essential cues for their recruitment to the remodeling endothelium. Pericyte involvement in the angiogenic sprouting context is also considered with specific attention to crosstalk with endothelial cells such as through signaling regulation and ECM deposition. We also address specific aspects of the collective cell migration and dynamic interactions between pericytes and endothelial cells during angiogenic sprouting. Lastly, we discuss pericyte contributions to mechanisms underlying the transition from active vessel remodeling to the maturation and quiescence phase of vascular development.
Collapse
Affiliation(s)
- Laura Beth Payne
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, Roanoke, VA 24016, USA
| | - Huaning Zhao
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic State Institute and State University, Blacksburg, VA 24061, USA
| | - Carissa C. James
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, Roanoke, VA 24016, USA
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Jordan Darden
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, Roanoke, VA 24016, USA
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - David McGuire
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, Roanoke, VA 24016, USA
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Sarah Taylor
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, Roanoke, VA 24016, USA
| | - James W. Smyth
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, Roanoke, VA 24016, USA
- Department of Biological Sciences, College of Science, Virginia Polytechnic State Institute and State University, Blacksburg, VA 24061, USA
- Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - John C. Chappell
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic State Institute and State University, Blacksburg, VA 24061, USA
- Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| |
Collapse
|
32
|
Elbanna M, Orillion AR, Damayanti NP, Adelaiye-Ogala R, Shen L, Miles KM, Chintala S, Ciamporcero E, Ramakrishnan S, Ku SY, Rex K, Caenepeel S, Coxon A, Pili R. Dual Inhibition of Angiopoietin-TIE2 and MET Alters the Tumor Microenvironment and Prolongs Survival in a Metastatic Model of Renal Cell Carcinoma. Mol Cancer Ther 2019; 19:147-156. [PMID: 31582532 DOI: 10.1158/1535-7163.mct-18-1202] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 06/24/2019] [Accepted: 09/26/2019] [Indexed: 12/14/2022]
Abstract
Receptor tyrosine kinase inhibitors have shown clinical benefit in clear cell renal cell carcinoma (ccRCC), but novel therapeutic strategies are needed. The angiopoietin/Tie2 and MET pathways have been implicated in tumor angiogenesis, metastases, and macrophage infiltration. In our study, we used trebananib, an angiopoietin 1/2 inhibitor, and a novel small-molecule MET kinase inhibitor in patient-derived xenograft (PDX) models of ccRCC. Our goal was to assess the ability of these compounds to alter the status of tumor-infiltrating macrophages, inhibit tumor growth and metastases, and prolong survival. Seven-week-old SCID mice were implanted subcutaneously or orthotopically with human ccRCC models. One month postimplantation, mice were treated with angiopoietin 1/2 inhibitor trebananib (AMG 386), MET kinase inhibitor, or combination. In our metastatic ccRCC PDX model, RP-R-02LM, trebananib alone, and in combination with a MET kinase inhibitor, significantly reduced lung metastases and M2 macrophage infiltration (P = 0.0075 and P = 0.0205, respectively). Survival studies revealed that treatment of the orthotopically implanted RP-R-02LM tumors yielded a significant increase in survival in both trebananib and combination groups. In addition, resection of the subcutaneously implanted primary tumor allowed for a significant survival advantage to the combination group compared with vehicle and both single-agent groups. Our results show that the combination of trebananib with a MET kinase inhibitor significantly inhibits the spread of metastases, reduces infiltrating M2-type macrophages, and prolongs survival in our highly metastatic ccRCC PDX model, suggesting a potential use for this combination therapy in treating patients with ccRCC.
Collapse
Affiliation(s)
- May Elbanna
- Genitourinary Cancers Program, Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, Indiana
| | - Ashley R Orillion
- Genitourinary Cancers Program, Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, Indiana.,Department of Cellular and Molecular Biology, University at Buffalo, Roswell Park Cancer Institute, Buffalo, New York
| | - Nur P Damayanti
- Genitourinary Cancers Program, Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, Indiana
| | - Remi Adelaiye-Ogala
- Genitourinary Cancers Program, Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, Indiana.,Department of Cancer Pathology and Prevention, University at Buffalo, Roswell Park Cancer Institute, Buffalo, New York
| | - Li Shen
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York
| | - Kiersten Marie Miles
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, New York
| | - Sreenivasulu Chintala
- Genitourinary Cancers Program, Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, Indiana
| | - Eric Ciamporcero
- Department of Medicine and Experimental Oncology, University of Turin, Turin, Italy
| | - Swathi Ramakrishnan
- Department of Cancer Pathology and Prevention, University at Buffalo, Roswell Park Cancer Institute, Buffalo, New York
| | - Sheng-Yu Ku
- Department of Cancer Pathology and Prevention, University at Buffalo, Roswell Park Cancer Institute, Buffalo, New York
| | - Karen Rex
- Oncology Research, Amgen Inc., Thousand Oaks, California
| | - Sean Caenepeel
- Oncology Research, Amgen Inc., Thousand Oaks, California
| | - Angela Coxon
- Oncology Research, Amgen Inc., Thousand Oaks, California
| | - Roberto Pili
- Genitourinary Cancers Program, Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, Indiana.
| |
Collapse
|
33
|
Funcke JB, Scherer PE. Beyond adiponectin and leptin: adipose tissue-derived mediators of inter-organ communication. J Lipid Res 2019; 60:1648-1684. [PMID: 31209153 PMCID: PMC6795086 DOI: 10.1194/jlr.r094060] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/17/2019] [Indexed: 01/10/2023] Open
Abstract
The breakthrough discoveries of leptin and adiponectin more than two decades ago led to a widespread recognition of adipose tissue as an endocrine organ. Many more adipose tissue-secreted signaling mediators (adipokines) have been identified since then, and much has been learned about how adipose tissue communicates with other organs of the body to maintain systemic homeostasis. Beyond proteins, additional factors, such as lipids, metabolites, noncoding RNAs, and extracellular vesicles (EVs), released by adipose tissue participate in this process. Here, we review the diverse signaling mediators and mechanisms adipose tissue utilizes to relay information to other organs. We discuss recently identified adipokines (proteins, lipids, and metabolites) and briefly outline the contributions of noncoding RNAs and EVs to the ever-increasing complexities of adipose tissue inter-organ communication. We conclude by reflecting on central aspects of adipokine biology, namely, the contribution of distinct adipose tissue depots and cell types to adipokine secretion, the phenomenon of adipokine resistance, and the capacity of adipose tissue to act both as a source and sink of signaling mediators.
Collapse
Affiliation(s)
- Jan-Bernd Funcke
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
34
|
Biesiada L, Sakowicz A, Grzesiak M, Borowiec M, Lisowska M, Pietrucha T, von Kaisenberg C, Lewandowski K. Identification of placental genes linked to selective intrauterine growth restriction (IUGR) in dichorionic twin pregnancies: gene expression profiling study. Hum Genet 2019; 138:649-659. [PMID: 31041507 PMCID: PMC6554264 DOI: 10.1007/s00439-019-02016-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 04/16/2019] [Indexed: 11/28/2022]
Abstract
A linkage of dichorionic (DC) twin pregnancies with selective intrauterine growth restriction (IUGR) to alterations in placental gene expression is unclear. The aim of the study was to identify placental genes related to hypoxia, adipogenesis and human growth which may contribute to IUGR development. The study group (IUGR/AGA) comprised dichorionic (DC) twin pregnancies, where the weight of the twins differed by > 15%; in addition, one twin was small for gestational age (< 10th percentile-SGA) (IUGR) while the other was appropriate for gestational age (> 10th percentile-AGA). In the control group (AGA/AGA), both fetuses were AGA and their weights differed by < 15%. In the first step (selection), placental expression of 260 genes was analysed by commercial PCR profiler array or qPCR primer assay between six pairs of IUGR/AGA twins. In the second stage (verification), the expression of 20 genes with fold change (FC) > 1.5 selected from the first stage was investigated for 75 DC pregnancies: 23 IUGR/AGA vs. 52 AGA/AGA. The expression of Angiopoetin 2, Leptin and Kruppel-like factor 4 was significantly higher, and Glis Family Zinc Finger 3 was lower, in placentas of SGA fetuses (FC = 3.3; 4.4; 1.6; and - 1.8, respectively; p < 0.05). The dysregulation of gene expression related to angiogenesis and growth factors in placentas of twins born from IUGR/AGA pregnancies suggest that these alternations might represent biological fetal adaptation to the uteral condition. Moreover, DC twin pregnancies may be a good model to identify the differences in placental gene expression between SGA and AGA fetuses.
Collapse
Affiliation(s)
- Lidia Biesiada
- Department of Obstetrics, Perinatology and Gynecology, Polish Mother's Memorial Hospital-Research Institute in Lodz, Rzgowska 281/289, Lodz, Poland.
| | - Agata Sakowicz
- Department of Medical Biotechnology, Medical University of Lodz, Lodz, Poland
| | - Mariusz Grzesiak
- Department of Obstetrics, Perinatology and Gynecology, Polish Mother's Memorial Hospital-Research Institute in Lodz, Rzgowska 281/289, Lodz, Poland
| | - Maciej Borowiec
- Department of Clinical Genetic, Medical University of Lodz, Lodz, Poland
| | - Michalina Lisowska
- Department of Medical Biotechnology, Medical University of Lodz, Lodz, Poland
| | - Tadeusz Pietrucha
- Department of Medical Biotechnology, Medical University of Lodz, Lodz, Poland
| | | | - Krzysztof Lewandowski
- Department of Endocrinology and Metabolic Diseases, Polish Mother's Memorial Hospital-Research Institute in Lodz, Lodz, Poland
| |
Collapse
|
35
|
de Vega S, Kondo A, Suzuki M, Arai H, Jiapaer S, Sabit H, Nakada M, Ikeuchi T, Ishijima M, Arikawa-Hirasawa E, Yamada Y, Okada Y. Fibulin-7 is overexpressed in glioblastomas and modulates glioblastoma neovascularization through interaction with angiopoietin-1. Int J Cancer 2019; 145:2157-2169. [PMID: 30924128 DOI: 10.1002/ijc.32306] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 03/20/2019] [Indexed: 01/01/2023]
Abstract
Glioblastoma (GBM) is pathologically characterized by highly malignant neoplastic cells, focal necrosis and aberrant blood vessels composed of disorganized endothelial cells and pericytes. The recent cancer microarray database revealed upregulation of fibulin-7 (Fbln7), a member of the fibulin family, but provided no information on the tissue localization or biological function. In the present study, we demonstrated that Fbln7 is markedly overexpressed by the GBM tissue among astrocytic tumors, and immunolocalized mainly to endothelial cells and pericytes of the glomeruloid and hypertrophied microvessels. The production of Fbln7 by endothelial cells and pericytes was confirmed in cultured human umbilical vein endothelial cells (HUVEC) and human brain vascular pericytes (HBVP) and vascular endothelial growth factor (VEGF) stimulated the Fbln7 expression in HUVEC. Fbln7 bound to angiopoietin-1, but not angiopoietin-2 or Tie2 receptor, through interaction between the N-terminal portions of Fbln7 and angiopoietin-1, and it blocked phosphorylation of Tie2 receptor in HUVEC. In a coculture assay using HUVEC and HBVP, multilayered and irregular-shaped tube-like structures of HUVEC were induced by treatment with a high concentration of VEGF. This was accompanied by Fbln7 overproduction by HUVEC and angiopoietin-1 expression by HBVP. The production of aberrant VEGF-induced tube-like structures was attenuated by treatment with antibody or synthetic peptides specific to the Fbln7 N-terminal domain or knockdown of Fbln7. These data demonstrate that Fbln7 is overexpressed by endothelial cells and pericytes of the abnormal microvessels in GBM, and suggest that Fbln7 may contribute to the aberrant vessel formation by modulation of the angiopoietin-1/angiopoietin-2-Tie2 axis.
Collapse
Affiliation(s)
- Susana de Vega
- Department of Pathophysiology for Locomotive and Neoplastic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akihide Kondo
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mario Suzuki
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hajime Arai
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shabierjiang Jiapaer
- Department of Neurosurgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hemragul Sabit
- Department of Neurosurgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Mitsutoshi Nakada
- Department of Neurosurgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Tomoko Ikeuchi
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institute of Health, Bethesda, MD, USA
| | - Muneaki Ishijima
- Department of Pathophysiology for Locomotive and Neoplastic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Eri Arikawa-Hirasawa
- Research Institute for the Diseases of Old Age, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshihiko Yamada
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institute of Health, Bethesda, MD, USA
| | - Yasunori Okada
- Department of Pathophysiology for Locomotive and Neoplastic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
36
|
Jiang W, Ji M. Receptor tyrosine kinases in PI3K signaling: The therapeutic targets in cancer. Semin Cancer Biol 2019; 59:3-22. [PMID: 30943434 DOI: 10.1016/j.semcancer.2019.03.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 03/09/2019] [Accepted: 03/28/2019] [Indexed: 12/17/2022]
Abstract
The phosphoinositide 3-kinase (PI3K) pathway, one of the most commonly activated signaling pathways in human cancers, plays a crucial role in the regulation of cell proliferation, differentiation, and survival. This pathway is usually activated by receptor tyrosine kinases (RTKs), whose constitutive and aberrant activation is via gain-of-function mutations, chromosomal rearrangement, gene amplification and autocrine. Blockage of PI3K pathway by targeted therapy on RTKs with tyrosine kinases inhibitors (TKIs) and monoclonal antibodies (mAbs) has achieved great progress in past decades; however, there still remain big challenges during their clinical application. In this review, we provide an overview about the most frequently encountered alterations in RTKs and focus on current therapeutic agents developed to counteract their aberrant functions, accompanied with discussions of two major challenges to the RTKs-targeted therapy in cancer - resistance and toxicity.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Meiju Ji
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China; Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, PR China.
| |
Collapse
|
37
|
Zhao H, Chappell JC. Microvascular bioengineering: a focus on pericytes. J Biol Eng 2019; 13:26. [PMID: 30984287 PMCID: PMC6444752 DOI: 10.1186/s13036-019-0158-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/15/2019] [Indexed: 12/26/2022] Open
Abstract
Capillaries within the microcirculation are essential for oxygen delivery and nutrient/waste exchange, among other critical functions. Microvascular bioengineering approaches have sought to recapitulate many key features of these capillary networks, with an increasing appreciation for the necessity of incorporating vascular pericytes. Here, we briefly review established and more recent insights into important aspects of pericyte identification and function within the microvasculature. We then consider the importance of including vascular pericytes in various bioengineered microvessel platforms including 3D culturing and microfluidic systems. We also discuss how vascular pericytes are a vital component in the construction of computational models that simulate microcirculation phenomena including angiogenesis, microvascular biomechanics, and kinetics of exchange across the vessel wall. In reviewing these topics, we highlight the notion that incorporating pericytes into microvascular bioengineering applications will increase their utility and accelerate the translation of basic discoveries to clinical solutions for vascular-related pathologies.
Collapse
Affiliation(s)
- Huaning Zhao
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, 2 Riverside Circle, Roanoke, VA 24016 USA.,Department of Biomedical Engineering and Mechanics, Virginia Polytechnic State Institute and State University, Blacksburg, VA 24061 USA
| | - John C Chappell
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, 2 Riverside Circle, Roanoke, VA 24016 USA.,Department of Biomedical Engineering and Mechanics, Virginia Polytechnic State Institute and State University, Blacksburg, VA 24061 USA.,3Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016 USA
| |
Collapse
|
38
|
Deng P, Hu C, Xiong Z, Li Y, Jiang J, Yang H, Tang Y, Cao L, Lu R. Epigallocatechin-3-gallate-induced vascular normalization in A549-cell xenograft-bearing nude mice: therapeutic efficacy in combination with chemotherapy. Cancer Manag Res 2019; 11:2425-2439. [PMID: 30988640 PMCID: PMC6441461 DOI: 10.2147/cmar.s187750] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose Large-scale studies have revealed that appropriate antiangiogenic treatment enables the recovery of the normal structure and function of solid tumor vessels. Epigallocatechin-3-gallate (EGCG), a natural extract of green tea, has multiple effects on angiogenesis. However, normalization of blood vessels due to natural ingredients has not yet been reported. Therefore, we examined the microvasculature, microenvironment, and efficacy of EGCG combined with chemotherapy in a xenograft model. Methods We treated A549 cell (human lung adenocarcinoma cell line) xenograft-bearing nude mice with EGCG in vivo. CD31, αSMA, and collagen IV were labeled and detected using quantum-dot double-labeled immunofluorescence to measure microvessel density, microvessel pericyte-coverage index, and collagen IV expression. Vessel-perfusion function was determined by lectin injection, permeability by Evans blue extravasation, interstitial fluid pressure using the wick-in-needle technique, and hypoxia levels using a polarographic electrode and immunohistochemical pimonidazole labeling. Cisplatin concentration in tumor tissue was detected using graphite-furnace atomic absorption spectrophotometry. Xenograft mice were randomized into five groups: treated with saline, cisplatin, EGCG, EGCG + cisplatin on day 1, or EGCG + cisplatin during the vascular normalization window. Tumor-growth delay and tumor-suppression rate were measured to evaluate tumor growth. Results EGCG treatment in vivo caused temporary changes, including transient depression of microvessel density, microvessel pericyte-coverage index, and collagen IV expression, transient elevation of vessel perfusion and permeability, and decreased interstitial fluid pressure and hypoxia. During vascular normalization, pretreatment with EGCG increased cisplatin concentration in tumor tissue compared with treatment with cisplatin only. Tumor-growth delay after treatment in the five groups during the vascular normalization window was 6.3±1.51, 7.5±1.57, 8.3±1.79, 12.1±1.35, and 15.4±1.99 days, indicating synergistic EGCG–cisplatin effects, especially during the vascular normalization window (P<0.01). Conclusion EGCG-induced vascular normalization in human lung adenocarcinoma may be a novel modality for enhancing chemotherapy effects.
Collapse
Affiliation(s)
- Pengbo Deng
- Department of Respiratory Medicine, Key Site of National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China,
| | - Chengping Hu
- Department of Respiratory Medicine, Key Site of National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China,
| | - Zeng Xiong
- Department of Respiratory Medicine, Key Site of National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China,
| | - Yuanyuan Li
- Department of Respiratory Medicine, Key Site of National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China,
| | - Juan Jiang
- Department of Respiratory Medicine, Key Site of National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China,
| | - Huaping Yang
- Department of Respiratory Medicine, Key Site of National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China,
| | - Yongjun Tang
- Department of Respiratory Medicine, Key Site of National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China,
| | - Liming Cao
- Department of Respiratory Medicine, Key Site of National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China,
| | - Rongli Lu
- Department of Respiratory Medicine, Key Site of National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China,
| |
Collapse
|
39
|
Tanabe A, Kobayashi D, Maeda K, Taguchi M, Sahara H. Angiogenesis-related gene expression profile in clinical cases of canine cancer. Vet Med Sci 2019; 5:19-29. [PMID: 30265453 PMCID: PMC6376169 DOI: 10.1002/vms3.127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The balance between pro- and anti-angiogenic signalling is tightly regulated in normal tissues to maintain the functions of the vasculature. In contrast, the overproduction of angiogenic factors and enhanced angiogenesis are frequently observed in several types of tumours. Although there have been many reports on the correlation between tumour progression and angiogenesis in humans, little is known about tumour angiogenesis in canines. Hence, we attempted to clarify whether angiogenesis contributes to tumour progression in canines as well as humans. In this study, we investigated the expression of several angiogenesis-related genes, including CD34, VEGF-A, VEGFR-1, VEGFR-2, Ang-1, Ang-2, Tie1, and Tie2, in 66 canine tumour tissues and in the normal tissues surrounding the tumours by quantitative real-time PCR analysis. Our comparative analysis between canine tumour tissues and normal tissues revealed that several angiogenesis-related genes, such as vascular endothelial growth factor (VEGF) and VEGF-receptor genes, were significantly upregulated in canine tumour tissues when compared to the normal tissues. We also found that the angiopoietin (Ang)-1/Ang-2 gene expression ratio was lower in canine tumour tissues than in the normal tissues, suggesting less association between vascular endothelial cells and perivascular cells in the canine tumour tissues. Taken together, our results suggest that several angiogenesis-related genes may contribute to the malignant progression of canine tumours via tumour angiogenesis.
Collapse
Affiliation(s)
- Atsushi Tanabe
- Laboratory of BiologyAzabu University School of Veterinary MedicineSagamiharaKanagawaJapan
| | - Daisuke Kobayashi
- Laboratory of BiologyAzabu University School of Veterinary MedicineSagamiharaKanagawaJapan
| | - Koki Maeda
- Laboratory of BiologyAzabu University School of Veterinary MedicineSagamiharaKanagawaJapan
| | - Masayuki Taguchi
- Laboratory of BiologyAzabu University School of Veterinary MedicineSagamiharaKanagawaJapan
| | - Hiroeki Sahara
- Laboratory of BiologyAzabu University School of Veterinary MedicineSagamiharaKanagawaJapan
| |
Collapse
|
40
|
Du S, Xiong H, Xu C, Lu Y, Yao J. Attempts to strengthen and simplify the tumor vascular normalization strategy using tumor vessel normalization promoting nanomedicines. Biomater Sci 2019; 7:1147-1160. [DOI: 10.1039/c8bm01350k] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Tumor vascular normalization theory opened the door for the rational use of antiangiogenic agents and chemotherapeutics.
Collapse
Affiliation(s)
- Shi Du
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Stability of Biopharmaceuticals
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Hui Xiong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Stability of Biopharmaceuticals
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Cheng Xu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Stability of Biopharmaceuticals
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Yun Lu
- Pharmaceutical R&D Institute
- Jiangsu Hengrui Medicine Co
- Ltd
- 222047
- China
| | - Jing Yao
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Stability of Biopharmaceuticals
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| |
Collapse
|
41
|
Abstract
Tumor blood vessel formation (angiogenesis) is essential for tumor growth and metastasis. Two main endothelial ligand–receptor pathways regulating angiogenesis are vascular endothelial growth factor (VEGF) receptor and angiopoietin-TIE receptor pathways. The angiopoietin-TIE pathway is required for the remodeling and maturation of the blood and lymphatic vessels during embryonic development after VEGF and VEGF-C mediated development of the primary vascular plexus. Angiopoietin-1 (ANGPT1) stabilizes the vasculature after angiogenic processes, via tyrosine kinase with immunoglobulin-like and EGF-like domains 2 (TIE2) activation. In contrast, ANGPT2 is upregulated at sites of vascular remodeling. ANGPT2 is secreted by activated endothelial cells in inflammation, promoting vascular destabilization. ANGPT2 has been found to be expressed in many human cancers. Intriguingly, in preclinical models inhibition of ANGPT2 has provided promising results in preventing tumor angiogenesis, tumor growth, and metastasis, making it an attractive candidate to target in tumors. However, until now the first ANGPT2 targeting therapies have been less effective in clinical trials than in experimental models. Additionally, in preclinical models combined therapy against ANGPT2 and VEGF or immune checkpoint inhibitors has been superior to monotherapies, and these pathways are also targeted in early clinical trials. In order to improve current anti-angiogenic therapies and successfully exploit ANGPT2 as a target for cancer treatment, the biology of the angiopoietin-TIE pathway needs to be profoundly clarified.
Collapse
Affiliation(s)
- Dieter Marmé
- Tumor Biology Center, Freiburg, Baden-Württemberg Germany
| |
Collapse
|
42
|
Tsukita Y, Okazaki T, Ebihara S, Komatsu R, Nihei M, Kobayashi M, Hirano T, Sugiura H, Tamada T, Tanaka N, Sato Y, Yagita H, Ichinose M. Beneficial effects of sunitinib on tumor microenvironment and immunotherapy targeting death receptor5. Oncoimmunology 2018; 8:e1543526. [PMID: 30713805 DOI: 10.1080/2162402x.2018.1543526] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 10/18/2018] [Accepted: 10/25/2018] [Indexed: 01/08/2023] Open
Abstract
Tumor-associated blood vessels and lymphatics are abnormal and dysfunctional. These are hallmarks of the tumor microenvironment, which has an immunosuppressive nature, such as through hypoxia. Treatment with anti-death receptor5 (DR5) monoclonal antibody MD5-1, which induces tumor cell death, is a potent anti-tumor immunotherapy. Generally, MD5-1 induces cell death mainly via antigen presenting cells (APCs) and generates tumor-specific effector T cells. To date, the effects of a simultaneous functional improvement of abnormal blood vessels and lymphatics on the immune microenvironment are largely unknown. A combination therapy using sunitinib, vascular endothelial growth factor (VEGF) and platelet-derived growth factor receptor inhibitor, and MD5-1 substantially inhibited tumor growth. Sunitinib improved pericyte coverage on endothelial cells and the expression levels of regulator of G-protein signaling 5, suggesting blood vessel normalization. Sunitinib also increased lymph flow from tumors to central lymph nodes, suggesting improved lymphatic function. In concordance with improved vasculature functions, sunitinib alleviated the tumor hypoxia, suggesting an improved tumor microenvironment. Indeed, the combination therapy induced strong activation of CD8+ T cells and dendritic cells in draining lymph nodes. The combination therapy reduced the ratio of immune-suppressive T regulatory cells in the tumors and draining lymph nodes. The combination therapy enhanced the numbers and activation of tumor-infiltrating CD8+ T cells. CD4 and/or CD8 depletion, or APC inhibiting experiments showed the contribution of CD8+ T cells and APCs to the combination therapy. These findings suggest that targeting blood vessels and lymphatics may have potential benefits for immunotherapy mediated by CD8+ T cells and APCs.
Collapse
Affiliation(s)
- Yoko Tsukita
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tatsuma Okazaki
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.,Present address; Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Satoru Ebihara
- Department of Rehabilitation Medicine, Toho University Graduate School of Medicine, Tokyo, Japan
| | - Riyo Komatsu
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mayumi Nihei
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Makoto Kobayashi
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Taizou Hirano
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hisatoshi Sugiura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tsutomu Tamada
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Nobuyuki Tanaka
- Division of Tumor Immunobiology, Department of Cancer Medical Science, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasufumi Sato
- Department of Vascular Biology, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Masakazu Ichinose
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
43
|
Yin J, Wang B, Zhu C, Sun C, Liu X. [Local injection of angiopoietin 2 promotes angiogenesis in tissue engineered bone and repair of bone defect with autophagy induction in vivo]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2018; 32:1150-1156. [PMID: 30129346 PMCID: PMC8413973 DOI: 10.7507/1002-1892.201804105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 08/09/2018] [Indexed: 01/07/2023]
Abstract
Objective To investigate the mechanism of early vascularization of the tissue engineered bone in the treatment of rabbit radial bone defect by local injection of angiopoietin 2 (Ang-2). Methods A single 1.5 cm long radius defect model (left and right sides randomised) was constructed from 48 New Zealand white rabbits. After implantation of hydroxyapatite/collagen scaffolds in bone defects, the rabbits were randomly divided into 2 groups: control group (group A) and Ang-2 group (group B) were injected with 1 mL normal saline and 1 mL saline-soluble 400 ng/mL Ang-2 daily at the bone defect within 2 weeks after operation, respectively. Western blot was used to detect the expressions of autophagy related protein [microtubule associated protein 1 light chain 3 (LC3), Beclin-1], angiogenesis related protein [vascular endothelial growth factor (VEGF)], and autophagy degradable substrate protein (SQSTMl/p62) in callus. X-ray films examination and Lane-Sandhu X-ray scoring were performed to evaluate the bone defect repair at 4, 8, and 12 weeks after operation. The rabbits were sacrificed at 12 weeks after operation for gross observation, and the angiogenesis of bone defect was observed by HE staining. Results Western blot assay showed that the relative expression of LC3-II/LC3-I, Beclin-1, and VEGF in group B was significantly higher than that in group A, and the relative expression of SQSTMl/p62 was significantly lower than that in group A ( P<0.05). Radiographic and gross observation of specimens showed that only a small number of callus were formed in group A, the bone defect was not repaired; more callus were formed and complete repair of bone defect was observed in group B. The Lane-Sandhu scores in group B were significantly higher than those in group A at 4, 8, and 12 weeks after operation ( P<0.05). HE staining showed that the Harvard tubes in group B were well arranged and the number of new vessels was significantly higher than that in group A ( t=-11.879, P=0.000). Conclusion Local injection of appropriate concentration of Ang-2 may promote early vascularization and bone defect repair of rabbit tissue engineered bone by enhancing autophagy.
Collapse
Affiliation(s)
- Jian Yin
- Department of Orthopedics, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing Jiangsu, 211100, P.R.China
| | - Bin Wang
- Department of Orthopedics, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing Jiangsu, 211100, P.R.China
| | - Chao Zhu
- Department of Orthopedics, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing Jiangsu, 211100, P.R.China
| | - Chao Sun
- Department of Orthopedics, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing Jiangsu, 211100, P.R.China
| | - Xinhui Liu
- Department of Orthopedics, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing Jiangsu, 211100,
| |
Collapse
|
44
|
Tsuji-Tamura K, Ogawa M. Morphology regulation in vascular endothelial cells. Inflamm Regen 2018; 38:25. [PMID: 30214642 PMCID: PMC6130072 DOI: 10.1186/s41232-018-0083-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/07/2018] [Indexed: 12/22/2022] Open
Abstract
Morphological change in endothelial cells is an initial and crucial step in the process of establishing a functional vascular network. Following or associated with differentiation and proliferation, endothelial cells elongate and assemble into linear cord-like vessels, subsequently forming a perfusable vascular tube. In vivo and in vitro studies have begun to outline the underlying genetic and signaling mechanisms behind endothelial cell morphology regulation. This review focuses on the transcription factors and signaling pathways regulating endothelial cell behavior, involved in morphology, during vascular development.
Collapse
Affiliation(s)
- Kiyomi Tsuji-Tamura
- 1Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 860-0811 Japan.,2Present Address: Oral Biochemistry and Molecular Biology, Department of Oral Health Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586 Japan
| | - Minetaro Ogawa
- 1Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 860-0811 Japan
| |
Collapse
|
45
|
Hu J, Lin S, Huang JJ, Cheung PCK. Mechanistic Study of the In Vitro and In Vivo Inhibitory Effects of Protocatechuic Acid and Syringic Acid on VEGF-Induced Angiogenesis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:6742-6751. [PMID: 29886729 DOI: 10.1021/acs.jafc.8b01534] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The antiangiogenic activities of two structurally similar phenolics, protocatechuic acid (PA) and syringic acid (SA), were investigated. In vitro study using HUVECs demonstrated that both PA and SA (at 25 μM) significantly ( p < 0.05) inhibited VEGF-induced cell proliferation by 22.68 ± 5.6% and 21.93 ± 2.0%, respectively; cell migration by 50.04 ± 3.3% and 39.72 ± 4.7%, respectively; cell invasion by 44.16 ± 4.23% and 51.90 ± 2.73%, respectively; and cellular ROS generation by 11.48 ± 6.32% and 21.17 ± 9.10%, respectively. Our mechanistic study revealed that PA and SA blocked the VEGFR2-dependent Akt/ MMP2 and ERK pathways in HUVECs. These inhibitory effects were further confirmed by a decrease of endogenous alkaline phosphatase activity for PA and SA (21.47 ± 1.77% and 10.37 ± 1.27%, respectively) and the suppression of subintestinal vessel plexus formation in Tg (fli1a:EGFP) y1-type transgenic zebrafish embryos. PA and SA down-regulated the angiogenesis-related signal transduction pathway of VEGFα-VEGFR2 or Ang2- Tie2 in zebrafish. Moreover, it was also found that PA demonstrated a better inhibition on VEGF-induced migration of HUVEC and zebrafish vasculature. This might be due to the different number of hydroxyl and methoxy substituents possessed by PA and SA. Taken together, these findings indicate that phenolics PA and SA have potent antiangiogenic activities and are potential targets for the design and development of anticancer agents.
Collapse
Affiliation(s)
- Jiamiao Hu
- College of Food Science , Fujian Agriculture and Forestry University , Fuzhou 350002 , China
| | - Shaoling Lin
- College of Food Science , Fujian Agriculture and Forestry University , Fuzhou 350002 , China
- School of Life Sciences , The Chinese University of Hong Kong , Shatin , New Territories , Hong Kong, China
| | - Jim Junhui Huang
- Environmental Research Institute , National University of Singapore , 5 Science Drive 2 , Singapore 117597 , Republic of Singapore
| | - Peter C K Cheung
- School of Life Sciences , The Chinese University of Hong Kong , Shatin , New Territories , Hong Kong, China
| |
Collapse
|
46
|
Harrell CR, Simovic Markovic B, Fellabaum C, Arsenijevic A, Djonov V, Volarevic V. Molecular mechanisms underlying therapeutic potential of pericytes. J Biomed Sci 2018; 25:21. [PMID: 29519245 PMCID: PMC5844098 DOI: 10.1186/s12929-018-0423-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 02/21/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Pericytes are multipotent cells present in every vascularized tissue in the body. Despite the fact that they are well-known for more than a century, pericytes are still representing cells with intriguing properties. This is mainly because of their heterogeneity in terms of definition, tissue distribution, origin, phenotype and multi-functional properties. The body of knowledge illustrates importance of pericytes in the regulation of homeostatic and healing processes in the body. MAIN BODY In this review, we summarized current knowledge regarding identification, isolation, ontogeny and functional characteristics of pericytes and described molecular mechanisms involved in the crosstalk between pericytes and endothelial or immune cells. We highlighted the role of pericytes in the pathogenesis of fibrosis, diabetes-related complications (retinopathy, nephropathy, neuropathy and erectile dysfunction), ischemic organ failure, pulmonary hypertension, Alzheimer disease, tumor growth and metastasis with the focus on their therapeutic potential in the regenerative medicine. The functions and capabilities of pericytes are impressive and, as yet, incompletely understood. Molecular mechanisms responsible for pericyte-mediated regulation of vascular stability, angiogenesis and blood flow are well described while their regenerative and immunomodulatory characteristics are still not completely revealed. Strong evidence for pericytes' participation in physiological, as well as in pathological conditions reveals a broad potential for their therapeutic use. Recently published results obtained in animal studies showed that transplantation of pericytes could positively influence the healing of bone, muscle and skin and could support revascularization. However, the differences in their phenotype and function as well as the lack of standardized procedure for their isolation and characterization limit their use in clinical trials. CONCLUSION Critical to further progress in clinical application of pericytes will be identification of tissue specific pericyte phenotype and function, validation and standardization of the procedure for their isolation that will enable establishment of precise clinical settings in which pericyte-based therapy will be efficiently applied.
Collapse
Affiliation(s)
- C. Randall Harrell
- Regenerative Processing Plant, LLC, 34176 US Highway 19 N Palm Harbor, Palm Harbor, Florida USA
| | - Bojana Simovic Markovic
- Department of Microbiology and immunology, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Serbia, Faculty of Medical Sciences, 69 Svetozar Markovic Street, Kragujevac, 34000 Serbia
| | - Crissy Fellabaum
- Regenerative Processing Plant, LLC, 34176 US Highway 19 N Palm Harbor, Palm Harbor, Florida USA
| | - Aleksandar Arsenijevic
- Department of Microbiology and immunology, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Serbia, Faculty of Medical Sciences, 69 Svetozar Markovic Street, Kragujevac, 34000 Serbia
| | - Valentin Djonov
- University of Bern, Institute of Anatomy, Baltzerstrasse 2, Bern, Switzerland
| | - Vladislav Volarevic
- Department of Microbiology and immunology, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Serbia, Faculty of Medical Sciences, 69 Svetozar Markovic Street, Kragujevac, 34000 Serbia
| |
Collapse
|
47
|
Peng H, Su Q, Lin ZC, Zhu XH, Peng MS, Lv ZB. Potential suppressive effects of theophylline on human rectal cancer SW480 cells in vitro by inhibiting YKL-40 expression. Oncol Lett 2018; 15:7403-7408. [PMID: 29731892 DOI: 10.3892/ol.2018.8220] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 01/05/2018] [Indexed: 01/16/2023] Open
Abstract
Chitinase-3-like-1 protein (YKL-40), a member of the mammalian chitinase-like glycoproteins, serves a key role in the pathogenesis of rectal cancer. The present study examined the antitumor effect of theophylline, a pan-chitinase inhibitor, in rectal cancer in vitro and investigated the mechanism by which it acted. SW480 cell lines were treated with varying theophylline concentrations (10-2, 10-3, 10-4 and 10-5 mol/l). An MTT assay was used to observe cell proliferation and identify the optimal theophylline concentration. Western blotting was used to analyze YKL-40 expression. The cell cycle distribution of SW480 cell lines treated with theophylline was measured by flow cytometry. The angiopoietin-2 expression level was measured by ELISA. The expression levels of YKL-40 were evidently decreased in theophylline-treated SW480 cell lines. The proliferation of SW480 cells was inhibited following theophylline treatment, which was associated with G1 phase cell cycle arrest and a decrease in the expression of angiopoietin-2. The mechanism of theophylline action may involve the downregulation of YKL-40 expression, arrest of the cell cycle at G1 phase and inhibition of angiopoietin-2 expression. These results provide a rationale for the potential use of anti-YKL-40 and anti-angiogenic strategies in treating rectal cancer.
Collapse
Affiliation(s)
- Hong Peng
- Department of Anorectal Surgery, Nanchong Central Hospital, Nanchong, Sichuan 637000, P.R. China.,The Second Clinical College of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Qiang Su
- Department of Clinical Pharmacy, Nanchong Central Hospital, Nanchong, Sichuan 637000, P.R. China
| | - Zhong-Chao Lin
- Department of Anorectal Surgery, Nanchong Central Hospital, Nanchong, Sichuan 637000, P.R. China
| | - Xiu-Hua Zhu
- Department of Anorectal Surgery, Nanchong Central Hospital, Nanchong, Sichuan 637000, P.R. China
| | - Ming-Sha Peng
- Department of Anorectal Surgery, Nanchong Central Hospital, Nanchong, Sichuan 637000, P.R. China
| | - Zhen-Bing Lv
- Department of Gastrointestinal Surgery, Nanchong Central Hospital, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
48
|
Abstract
Immunotherapies have revolutionized medical oncology following the remarkable and, in some cases, unprecedented outcomes observed in certain groups of patients with cancer. Combination with other therapeutic modalities, including anti-angiogenic agents, is one of the many strategies currently under investigation to improve the response rates and duration of immunotherapies. Such a strategy might seem counterintuitive given that anti-angiogenic agents can increase tumour hypoxia and reduce the number of blood vessels within tumours. Herein, we review the additional effects mediated by drugs targeting VEGF-dependent signalling and other pathways, such as those mediated by angiopoietin 2 or HGF, which might increase the efficacy of immunotherapies. In addition, we discuss the seldom considered possibility that immunotherapies, and immune-checkpoint inhibitors in particular, might increase the efficacy of anti-angiogenic or other types of antivascular therapies and/or promote changes in the tumour vasculature. In short, we propose that interactions between both therapeutic modalities could be considered a 'two-way street'.
Collapse
|
49
|
Huang R, Andersen LMK, Rofstad EK. Metastatic pathway and the microvascular and physicochemical microenvironments of human melanoma xenografts. J Transl Med 2017; 15:203. [PMID: 29017512 PMCID: PMC5634823 DOI: 10.1186/s12967-017-1307-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 09/26/2017] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Malignant melanoma of the skin can metastasize through blood vessels and lymphatics. The primary tumor develops a vascular microenvironment characterized by abnormal blood vessels and lymphatics and a physicochemical microenvironment characterized by low oxygen tension, regions with hypoxic tissue, and high interstitial fluid pressure (IFP). This study aimed at identifying relationships between the metastatic route of melanomas and characteristic features of the microvascular and physicochemical microenvironments of the primary tumor. METHODS Two patient-derived xenograft (PDX) models (E-13, N-15) and four cell line-derived xenografts (CDX) models (C-10, D-12, R-18, T-22) of human melanoma were included in the study. Tumors were transplanted to an orthotopic site in BALB/c-nu/nu mice, and when the tumors had grown to a volume of 500-600 mm3, the IFP of the primary tumor was measured and the hypoxia marker pimonidazole was administered before the host mouse was euthanized. The primary tumor, lungs, and six pairs of lymph nodes were evaluated by examining hematoxylin/eosin-stained and immunostained histological preparations. The expression of angiogenesis-related genes was assessed by quantitative PCR. RESULTS C-10, D-12, and E-13 tumors disseminated primarily by the hematogenous route and developed pulmonary metastases. These tumors showed high angiogenic activity and high expression of the F3 gene as well as ANGPT2 and TIE1, genes encoding proteins of the angiopoietin-tie system. N-15, R-18, and T-22 tumors disseminated mainly by the lymphogenous route and developed metastases in draining lymph nodes. These tumors had highly elevated IFP and showed high expression of NRP2, a gene encoding neuropilin-2. CONCLUSION The primary metastatic route of orthotopic human melanoma xenografts and the development of lung and lymph node metastases are influenced significantly by the microvascular and physicochemical microenvironments of the primary tumor.
Collapse
Affiliation(s)
- Ruixia Huang
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Lise Mari K. Andersen
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Einar K. Rofstad
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Box 4953, Nydalen, 0424 Oslo, Norway
| |
Collapse
|
50
|
Zhou YH, Chen Y, Hu Y, Yu L, Tran K, Giedzinski E, Ru N, Gau A, Pan F, Qiao J, Atkin N, Ly KC, Lee N, Siegel ER, Linskey ME, Wang P, Limoli C. The role of EGFR double minutes in modulating the response of malignant gliomas to radiotherapy. Oncotarget 2017; 8:80853-80868. [PMID: 29113349 PMCID: PMC5655244 DOI: 10.18632/oncotarget.20714] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 08/04/2017] [Indexed: 01/05/2023] Open
Abstract
EGFR amplification in cells having double minute chromosomes (DM) is commonly found in glioblastoma multiforme (GBM); however, how much it contributes to the current failure to treat GBM successfully is unknown. We studied two syngeneic primary cultures derived from a GBM with and without cells carrying DM, for their differential molecular and metabolic profiles, in vivo growth patterns, and responses to irradiation (IR). Each cell line has a distinct molecular profile consistent with an invasive “go” (with DM) or angiogenic “grow” phenotype (without DM) demonstrated in vitro and in intracranial xenograft models. Cells with DM were relatively radio-resistant and used higher glycolytic respiration and lower oxidative phosphorylation in comparison to cells without them. The DM-containing cell was able to restore tumor heterogeneity by mis-segregation of the DM-chromosomes, giving rise to cell subpopulations without them. As a response to IR, DM-containing cells switched their respiration from glycolic metabolism to oxidative phosphorylation and shifted molecular profiles towards that of cells without DM. Irradiated cells with DM showed the capacity to alter their extracellular microenvironment to not only promote invasiveness of the surrounding cells, regardless of DM status, but also to create a pro-angiogenic tumor microenvironment. IR of cells without DM was found primarily to increase extracellular MMP2 activity. Overall, our data suggest that the DM-containing cells of GBM are responsible for tumor recurrence due to their high invasiveness and radio-resistance and the mis-segregation of their DM chromosomes, to give rise to fast-growing cells lacking DM chromosomes.
Collapse
Affiliation(s)
- Yi-Hong Zhou
- UC Irvine Brain Tumor Laboratory and Department of Surgery, University of California Irvine, Irvine, CA, USA
| | - Yumay Chen
- UC Irvine Diabetes Center and Department of Medicine, University of California Irvine, Irvine, CA, USA
| | - Yuanjie Hu
- UC Irvine Brain Tumor Laboratory and Department of Surgery, University of California Irvine, Irvine, CA, USA
| | - Liping Yu
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, USA
| | - Katherine Tran
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, USA
| | - Erich Giedzinski
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, USA
| | - Ning Ru
- UC Irvine Brain Tumor Laboratory and Department of Surgery, University of California Irvine, Irvine, CA, USA
| | - Alex Gau
- UC Irvine Brain Tumor Laboratory and Department of Surgery, University of California Irvine, Irvine, CA, USA
| | - Francine Pan
- UC Irvine Brain Tumor Laboratory and Department of Surgery, University of California Irvine, Irvine, CA, USA
| | - Jiao Qiao
- UC Irvine Brain Tumor Laboratory and Department of Surgery, University of California Irvine, Irvine, CA, USA
| | - Naomi Atkin
- UC Irvine Brain Tumor Laboratory and Department of Surgery, University of California Irvine, Irvine, CA, USA
| | - Khang Chi Ly
- UC Irvine Brain Tumor Laboratory and Department of Surgery, University of California Irvine, Irvine, CA, USA
| | - Nathan Lee
- UC Irvine Brain Tumor Laboratory and Department of Surgery, University of California Irvine, Irvine, CA, USA
| | - Eric R Siegel
- Departments of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Mark E Linskey
- UC Irvine Brain Tumor Laboratory and Department of Surgery, University of California Irvine, Irvine, CA, USA
| | - Ping Wang
- UC Irvine Diabetes Center and Department of Medicine, University of California Irvine, Irvine, CA, USA
| | - Charles Limoli
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, USA
| |
Collapse
|