1
|
Feng X, Cao F, Wu X, Xie W, Wang P, Jiang H. Targeting extracellular matrix stiffness for cancer therapy. Front Immunol 2024; 15:1467602. [PMID: 39697341 PMCID: PMC11653020 DOI: 10.3389/fimmu.2024.1467602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 11/06/2024] [Indexed: 12/20/2024] Open
Abstract
The physical characteristics of the tumor microenvironment (TME) include solid stress, interstitial fluid pressure, tissue stiffness and microarchitecture. Among them, abnormal changes in tissue stiffness hinder drug delivery, inhibit infiltration of immune killer cells to the tumor site, and contribute to tumor resistance to immunotherapy. Therefore, targeting tissue stiffness to increase the infiltration of drugs and immune cells can offer a powerful support and opportunities to improve the immunotherapy efficacy in solid tumors. In this review, we discuss the mechanical properties of tumors, the impact of a stiff TME on tumor cells and immune cells, and the strategies to modulate tumor mechanics.
Collapse
Affiliation(s)
- Xiuqin Feng
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fujun Cao
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiangji Wu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenyan Xie
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ping Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Jiang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Noack D, van Haperen A, van den Hout MCGN, Marshall EM, Koutstaal RW, van Duinen V, Bauer L, van Zonneveld AJ, van IJcken WFJ, Koopmans MPG, Rockx B. A three-dimensional vessel-on-chip model to study Puumala orthohantavirus pathogenesis. LAB ON A CHIP 2024. [PMID: 39292495 DOI: 10.1039/d4lc00543k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Puumala orthohantavirus (PUUV) infection in humans can result in hemorrhagic fever with renal syndrome. Endothelial cells (ECs) are primarily infected with increased vascular permeability as a central aspect of pathogenesis. Historically, most studies included ECs cultured under static two-dimensional (2D) conditions, thereby not recapitulating the physiological environment due to their lack of flow and inherent pro-inflammatory state. Here, we present a high-throughput model for culturing primary human umbilical vein ECs in 3D vessels-on-chip in which we compared host responses of these ECs to those of static 2D-cultured ECs on a transcriptional level. The phenotype of ECs in vessels-on-chip more closely resembled the in vivo situation due to higher similarity in expression of genes encoding described markers for disease severity and coagulopathy, including IDO1, LGALS3BP, IL6 and PLAT, and more diverse endothelial-leukocyte interactions in the context of PUUV infection. In these vessels-on-chip, PUUV infection did not directly increase vascular permeability, but increased monocyte adhesion. This platform can be used for studying pathogenesis and assessment of possible therapeutics for other endotheliotropic viruses even in high biocontainment facilities.
Collapse
Affiliation(s)
- Danny Noack
- Department of Viroscience, Erasmus University Medical Center, s-Gravendijkwal 230, 3015 CE, Rotterdam, the Netherlands.
| | - Anouk van Haperen
- Department of Viroscience, Erasmus University Medical Center, s-Gravendijkwal 230, 3015 CE, Rotterdam, the Netherlands.
| | - Mirjam C G N van den Hout
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Center for Biomics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Eleanor M Marshall
- Department of Viroscience, Erasmus University Medical Center, s-Gravendijkwal 230, 3015 CE, Rotterdam, the Netherlands.
| | - Rosanne W Koutstaal
- Department of Viroscience, Erasmus University Medical Center, s-Gravendijkwal 230, 3015 CE, Rotterdam, the Netherlands.
| | - Vincent van Duinen
- Department of Internal Medicine, Division of Nephrology and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Lisa Bauer
- Department of Viroscience, Erasmus University Medical Center, s-Gravendijkwal 230, 3015 CE, Rotterdam, the Netherlands.
| | - Anton Jan van Zonneveld
- Department of Internal Medicine, Division of Nephrology and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Wilfred F J van IJcken
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Center for Biomics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marion P G Koopmans
- Department of Viroscience, Erasmus University Medical Center, s-Gravendijkwal 230, 3015 CE, Rotterdam, the Netherlands.
| | - Barry Rockx
- Department of Viroscience, Erasmus University Medical Center, s-Gravendijkwal 230, 3015 CE, Rotterdam, the Netherlands.
| |
Collapse
|
3
|
Liu W, Li S, Yang M, Ma J, Liu L, Fei P, Xiang Q, Huang L, Zhao P, Yang Z, Zhu X. Dysfunction of Calcyphosine-Like gene impairs retinal angiogenesis through the MYC axis and is associated with familial exudative vitreoretinopathy. eLife 2024; 13:RP96907. [PMID: 39264149 PMCID: PMC11392532 DOI: 10.7554/elife.96907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024] Open
Abstract
Familial exudative vitreoretinopathy (FEVR) is a severe genetic disorder characterized by incomplete vascularization of the peripheral retina and associated symptoms that can lead to vision loss. However, the underlying genetic causes of approximately 50% of FEVR cases remain unknown. Here, we report two heterozygous variants in calcyphosine-like gene (CAPSL) that is associated with FEVR. Both variants exhibited compromised CAPSL protein expression. Vascular endothelial cell (EC)-specific inactivation of Capsl resulted in delayed radial/vertical vascular progression, compromised endothelial proliferation/migration, recapitulating the human FEVR phenotypes. CAPSL-depleted human retinal microvascular endothelial cells (HRECs) exhibited impaired tube formation, decreased cell proliferation, disrupted cell polarity establishment, and filopodia/lamellipodia formation, as well as disrupted collective cell migration. Transcriptomic and proteomic profiling revealed that CAPSL abolition inhibited the MYC signaling axis, in which the expression of core MYC targeted genes were profoundly decreased. Furthermore, a combined analysis of CAPSL-depleted HRECs and c-MYC-depleted human umbilical vein endothelial cells uncovered similar transcription patterns. Collectively, this study reports a novel FEVR-associated candidate gene, CAPSL, which provides valuable information for genetic counseling of FEVR. This study also reveals that compromised CAPSL function may cause FEVR through MYC axis, shedding light on the potential involvement of MYC signaling in the pathogenesis of FEVR.
Collapse
Affiliation(s)
- Wenjing Liu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
- Henan Branch of National Clinical Research Center for Ocular Diseases, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Shujin Li
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Mu Yang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Jie Ma
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lu Liu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ping Fei
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianchun Xiang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lulin Huang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Peiquan Zhao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenglin Yang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
- Jinfeng Laboratory, Chongqing, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | - Xianjun Zhu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
- Henan Branch of National Clinical Research Center for Ocular Diseases, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
- Jinfeng Laboratory, Chongqing, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| |
Collapse
|
4
|
Iwatake M, Nagamura-Inoue T, Doi R, Tanoue Y, Ishii M, Yukawa H, Matsumoto K, Tomoshige K, Nagayasu T, Tsuchiya T. Designer umbilical cord-stem cells induce alveolar wall regeneration in pulmonary disease models. Front Immunol 2024; 15:1384718. [PMID: 38745668 PMCID: PMC11091323 DOI: 10.3389/fimmu.2024.1384718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 04/02/2024] [Indexed: 05/16/2024] Open
Abstract
Background Researchers are focusing on cellular therapy for chronic obstructive pulmonary disease (COPD) using mesenchymal stem cells (MSCs), with human bone marrow-derived MSCs (hBM-MSCs) leading the way. However, BM-MSCs may not be as optimal as therapeutic cells owing to their low growth potential, invasive harvesting, and high expression of aging-related genes with poor differentiation potential. Consequently, umbilical cord-derived MSCs (hUC-MSCs), which have many excellent features as allogeneic heterologous stem cells, have received considerable attention. Allogeneic and heterologous hUC-MSCs appear to be promising owing to their excellent therapeutic properties. However, MSCs cannot remain in the lungs for long periods after intravenous infusion. Objective To develop designer hUC-MSCs (dUC-MSCs), which are novel therapeutic cells with modified cell-adhesion properties, to aid COPD treatment. Methods dUC-MSCs were cultured on type-I collagen gels and laminin 411, which are extracellular matrices. Mouse models of elastase-induced COPD were treated with hUC-MSCs. Biochemical analysis of the lungs of treated and control animals was performed. Results Increased efficiency of vascular induction was found with dUC-MSCs transplanted into COPD mouse models compared with that observed with transplanted hUC-MSCs cultured on plates. The transplanted dUC-MSCs inhibited apoptosis by downregulating pro-inflammatory cytokine production, enhancing adhesion of the extracellular matrix to alveolar tissue via integrin β1, promoting the polarity of M2 macrophages, and contributing to the repair of collapsed alveolar walls by forming smooth muscle fibers. dUC-MSCs inhibited osteoclastogenesis in COPD-induced osteoporosis. hUC-MSCs are a promising cell source and have many advantages over BM-MSCs and adipose tissue-derived MSCs. Conclusion We developed novel designer cells that may be involved in anti-inflammatory, homeostatic, injury repair, and disease resistance processes. dUC-MSCs repair and regenerate the alveolar wall by enhancing adhesion to the damaged site. Therefore, they can contribute to the treatment of COPD and systemic diseases such as osteoporosis.
Collapse
Affiliation(s)
- Mayumi Iwatake
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Nagoya, Japan
- Division of Surgical Oncology, Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ryoichiro Doi
- Division of Surgical Oncology, Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yukinori Tanoue
- Division of Surgical Oncology, Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Mitsutoshi Ishii
- Division of Surgical Oncology, Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hiroshi Yukawa
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Nagoya, Japan
| | - Keitaro Matsumoto
- Division of Surgical Oncology, Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Koichi Tomoshige
- Division of Surgical Oncology, Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takeshi Nagayasu
- Division of Surgical Oncology, Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tomoshi Tsuchiya
- Division of Surgical Oncology, Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Thoracic Surgery, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| |
Collapse
|
5
|
Bai X, Wang R, Hu X, Dai Q, Guo J, Cao T, Du W, Cheng Y, Xia S, Wang D, Yang L, Teng L, Chen D, Liu Y. Two-Dimensional Biodegradable Black Phosphorus Nanosheets Promote Large Full-Thickness Wound Healing through In Situ Regeneration Therapy. ACS NANO 2024; 18:3553-3574. [PMID: 38226901 PMCID: PMC10832999 DOI: 10.1021/acsnano.3c11177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/16/2023] [Accepted: 12/18/2023] [Indexed: 01/17/2024]
Abstract
Large full-thickness skin lesions have been one of the most challenging clinical problems in plastic surgery repair and reconstruction. To achieve in situ skin regeneration and perfect clinical outcomes, we must address two significant obstacles: angiogenesis deficiency and inflammatory dysfunction. Recently, black phosphorus has shown great promise in wound healing. However, few studies have explored the bio-effects of BP to promote in situ skin regeneration based on its nanoproperties. Here, to investigate whether black phosphorus nanosheets have positive bio-effects on in situ skin repair, we verified black phosphorus nanosheets' positive effects on angiogenic and anti-inflammatory abilities in vitro. Next, the in vivo evaluation performed on the rat large full-thickness excisional wound splinting model more comprehensively showed that the positive bio-effects of black phosphorus nanosheets are multilevel in wound healing, which can effectively enhance anti-inflammatory ability, angiogenesis, collagen deposition, and skin re-epithelialization. Then, multiomics analysis was performed to explore further the mechanism of black phosphorus nanosheets' regulation of endothelial cells in depth. Molecular mechanistically, black phosphorus nanosheets activated the JAK-STAT-OAS signaling pathway to promote cellular function and mitochondrial energy metabolism in endothelial cells. This study can provide a theoretical basis for applying two-dimensional black phosphorus nanosheets as nanomedicine to achieve in situ tissue regeneration in complex human pathological microenvironments, guiding the subsequent optimization of black phosphorus.
Collapse
Affiliation(s)
- Xueshan Bai
- Cranio-Maxillo-Facial
Surgery Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100144, China
| | - Renxian Wang
- Laboratory
of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials,
National Center for Orthopaedics, Beijing Research Institute of Traumatology
and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
- JST
sarcopenia Research Centre, National Center for Orthopaedics, Beijing
Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan
Hospital, Capital Medical University, Beijing 100035, China
| | - Xiaohua Hu
- Department
of Burns and Plastic Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Qiang Dai
- Department
of Burns and Plastic Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Jianxun Guo
- Laboratory
of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials,
National Center for Orthopaedics, Beijing Research Institute of Traumatology
and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Tongyu Cao
- Department
of Burns and Plastic Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Weili Du
- Department
of Burns and Plastic Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Yuning Cheng
- Laboratory
of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials,
National Center for Orthopaedics, Beijing Research Institute of Traumatology
and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Songxia Xia
- Cranio-Maxillo-Facial
Surgery Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100144, China
| | - Dingding Wang
- JST
sarcopenia Research Centre, National Center for Orthopaedics, Beijing
Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan
Hospital, Capital Medical University, Beijing 100035, China
| | - Liya Yang
- Cranio-Maxillo-Facial
Surgery Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100144, China
| | - Li Teng
- Cranio-Maxillo-Facial
Surgery Department, Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100144, China
| | - Dafu Chen
- Laboratory
of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials,
National Center for Orthopaedics, Beijing Research Institute of Traumatology
and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Yajun Liu
- JST
sarcopenia Research Centre, National Center for Orthopaedics, Beijing
Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan
Hospital, Capital Medical University, Beijing 100035, China
- Department
of Spine Surgery, Beijing Jishuitan Hospital, National Center for
Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| |
Collapse
|
6
|
Xu Z, Chen Y, Wang Y, Han W, Xu W, Liao X, Zhang T, Wang G. Matrix stiffness, endothelial dysfunction and atherosclerosis. Mol Biol Rep 2023; 50:7027-7041. [PMID: 37382775 DOI: 10.1007/s11033-023-08502-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 04/28/2023] [Indexed: 06/30/2023]
Abstract
Atherosclerosis (AS) is the leading cause of the human cardiovascular diseases (CVDs). Endothelial dysfunction promotes the monocytes infiltration and inflammation that participate fundamentally in atherogenesis. Endothelial cells (EC) have been recognized as mechanosensitive cells and have different responses to distinct mechanical stimuli. Emerging evidence shows matrix stiffness-mediated EC dysfunction plays a vital role in vascular disease, but the underlying mechanisms are not yet completely understood. This article aims to summarize the effect of matrix stiffness on the pro-atherosclerotic characteristics of EC including morphology, rigidity, biological behavior and function as well as the related mechanical signal. The review also discusses and compares the contribution of matrix stiffness-mediated phagocytosis of macrophages and EC to AS progression. These advances in our understanding of the relationship between matrix stiffness and EC dysfunction open the avenues to improve the prevention and treatment of now-ubiquitous atherosclerotic diseases.
Collapse
Affiliation(s)
- Zichen Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yi Chen
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection, Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Yi Wang
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Wenbo Han
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Wenfeng Xu
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection, Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Xiaoling Liao
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection, Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Tao Zhang
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection, Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
- Bioengineering College of Chongqing University, NO.174, Shazheng Street, Shapingba District, Chongqing, 400030, PR China.
| |
Collapse
|
7
|
Xiao P, Zhang Y, Zeng Y, Yang D, Mo J, Zheng Z, Wang J, Zhang Y, Zhou Z, Zhong X, Yan W. Impaired angiogenesis in ageing: the central role of the extracellular matrix. J Transl Med 2023; 21:457. [PMID: 37434156 PMCID: PMC10334673 DOI: 10.1186/s12967-023-04315-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/30/2023] [Indexed: 07/13/2023] Open
Abstract
Each step in angiogenesis is regulated by the extracellular matrix (ECM). Accumulating evidence indicates that ageing-related changes in the ECM driven by cellular senescence lead to a reduction in neovascularisation, reduced microvascular density, and an increased risk of tissue ischaemic injury. These changes can lead to health events that have major negative impacts on quality of life and place a significant financial burden on the healthcare system. Elucidating interactions between the ECM and cells during angiogenesis in the context of ageing is neceary to clarify the mechanisms underlying reduced angiogenesis in older adults. In this review, we summarize ageing-related changes in the composition, structure, and function of the ECM and their relevance for angiogenesis. Then, we explore in detail the mechanisms of interaction between the aged ECM and cells during impaired angiogenesis in the older population for the first time, discussing diseases caused by restricted angiogenesis. We also outline several novel pro-angiogenic therapeutic strategies targeting the ECM that can provide new insights into the choice of appropriate treatments for a variety of age-related diseases. Based on the knowledge gathered from recent reports and journal articles, we provide a better understanding of the mechanisms underlying impaired angiogenesis with age and contribute to the development of effective treatments that will enhance quality of life.
Collapse
Affiliation(s)
- Ping Xiao
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanli Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yuting Zeng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Dehong Yang
- Department of Orthopedics Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiayao Mo
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ziting Zheng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jilei Wang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuxin Zhang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhiyan Zhou
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xincen Zhong
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wenjuan Yan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
8
|
Davis GE, Kemp SS. Extracellular Matrix Regulation of Vascular Morphogenesis, Maturation, and Stabilization. Cold Spring Harb Perspect Med 2023; 13:a041156. [PMID: 35817544 PMCID: PMC10578078 DOI: 10.1101/cshperspect.a041156] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The extracellular matrix represents a critical regulator of tissue vascularization during embryonic development and postnatal life. In this perspective, we present key information and concepts that focus on how the extracellular matrix controls capillary assembly, maturation, and stabilization, and, in addition, contributes to tissue stability and health. In particular, we present and discuss mechanistic details underlying (1) the role of the extracellular matrix in controlling different steps of vascular morphogenesis, (2) the ability of endothelial cells (ECs) and pericytes to coassemble into elongated and narrow capillary EC-lined tubes with associated pericytes and basement membrane matrices, and (3) the identification of specific growth factor combinations ("factors") and peptides as well as coordinated "factor" and extracellular matrix receptor signaling pathways that are required to form stabilized capillary networks.
Collapse
Affiliation(s)
- George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida 33612, USA
| | - Scott S Kemp
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida 33612, USA
| |
Collapse
|
9
|
The Passenger Domain of Bartonella bacilliformis BafA Promotes Endothelial Cell Angiogenesis via the VEGF Receptor Signaling Pathway. mSphere 2022; 7:e0008122. [PMID: 35379004 PMCID: PMC9044958 DOI: 10.1128/msphere.00081-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Bartonella bacilliformis is a Gram-negative bacterial pathogen that provokes pathological angiogenesis and causes Carrion’s disease, a neglected tropical disease restricted to South America. Little is known about how B. bacilliformis facilitates vasoproliferation resulting in hemangioma in the skin in verruga peruana, the chronic phase of Carrion’s disease. Here, we demonstrate that B. bacilliformis extracellularly secrets a passenger domain of the autotransporter BafA exhibiting proangiogenic activity. The B. bacilliformis-derived BafA passenger domain (BafABba) increased the number of human umbilical endothelial cells (HUVECs) and promoted tube-like morphogenesis. Neutralizing antibody against BafABba detected the BafA derivatives from the culture supernatant of B. bacilliformis and inhibited the infection-mediated hyperproliferation of HUVECs. Moreover, stimulation with BafABba promoted phosphorylation of vascular endothelial growth factor receptor 2 (VEGFR2) and extracellular-signal-regulated kinase 1/2 in HUVECs. Suppression of VEGFR2 by anti-VEGFR2 antibody or RNA interference reduced the sensitivity of cells to BafABba. In addition, surface plasmon resonance analysis confirmed that BafABba directly interacts with VEGFR2 with lower affinity than VEGF or Bartonella henselae-derived BafA. These findings indicate that BafABba acts as a VEGFR2 agonist analogous to the previously identified B. henselae- and Bartonella quintana-derived BafA proteins despite the low sequence similarity. The identification of a proangiogenic factor produced by B. bacilliformis that directly stimulates endothelial cells provides an important insight into the pathophysiology of verruga peruana. IMPORTANCEBartonella bacilliformis causes life-threatening bacteremia or dermal eruption known as Carrion’s disease in South America. During infection, B. bacilliformis promotes endothelial cell proliferation and the angiogenic process, but the underlying molecular mechanism has not been well understood. We show that B. bacilliformis induces vasoproliferation and angiogenesis by producing the proangiogenic autotransporter BafA. As the cellular/molecular basis for angiogenesis, BafA stimulates the signaling pathway of vascular endothelial growth factor receptor 2 (VEGFR2). Identification of functional BafA protein from B. bacilliformis in addition to B. henselae and B. quintana, the causes of cat scratch disease and trench fever, raises the possibility that BafA is a common virulence factor for human-pathogenic Bartonella.
Collapse
|
10
|
Mechanical Aspects of Angiogenesis. Cancers (Basel) 2021; 13:cancers13194987. [PMID: 34638470 PMCID: PMC8508205 DOI: 10.3390/cancers13194987] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/01/2021] [Accepted: 10/01/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The formation of new blood vessels from already existing ones is a process of high clinical relevance, since it is of great importance for both physiological and pathological processes. In regard to tumors, the process is crucial, since it ensures the supply with nutrients and the growth of the tumor. The influence of mechanical factors on this biological process is an emerging field. Until now, the shear force of the blood flow has been considered the main mechanical parameter during angiogenesis. This review article provides an overview of further mechanical cues, with particular focus on the surrounding extracellular matrix impacting the cell behavior and, thus, regulating angiogenesis. This underlines the enormous importance of the mechanical properties of the extracellular matrix on cell biological processes and shows how changing the mechanics of the extracellular matrix could be used as a possible therapeutic approach in cancer therapy. Abstract Angiogenesis is of high clinical relevance as it plays a crucial role in physiological (e.g., tissue regeneration) and pathological processes (e.g., tumor growth). Besides chemical signals, such as VEGF, the relationship between cells and the extracellular matrix (ECM) can influence endothelial cell behavior during angiogenesis. Previously, in terms of the connection between angiogenesis and mechanical factors, researchers have focused on shear forces due to blood flow. However, it is becoming increasingly important to include the direct influence of the ECM on biological processes, such as angiogenesis. In this context, we focus on the stiffness of the surrounding ECM and the adhesion of cells to the ECM. Furthermore, we highlight the mechanical cues during the main stages of angiogenesis: cell migration, tip and stalk cells, and vessel stabilization. It becomes clear that the different stages of angiogenesis require various chemical and mechanical cues to be modulated by/modulate the stiffness of the ECM. Thus, changes of the ECM during tumor growth represent additional potential dysregulations of angiogenesis in addition to erroneous biochemical signals. This awareness could be the basis of therapeutic approaches to counteract specific processes in tumor angiogenesis.
Collapse
|
11
|
Wu M, Chen Y, Feng L, Dai H, Fang S, Xu J. MiR-206 promotes extracellular matrix accumulation and relieves infantile hemangioma through targeted inhibition of DNMT3A. Cell Cycle 2021; 20:978-992. [PMID: 33945391 PMCID: PMC8172163 DOI: 10.1080/15384101.2021.1919820] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 10/21/2022] Open
Abstract
MiR-206 is abnormally expressed in infant hemangioma endothelial cells (HemECs), but the mechanism is not clear. We explored the intervention of miR-206 in HemECs in relation to extracellular matrix (ECM) metabolism. We selected 48 cases of infantile hemangioma (IH) from volunteer organizations. After the isolated and extracted HemECs were interfered with overexpressed or silenced miR-206, the effects of miR-206 on the proliferation, migration and invasion of HemECs were examined through basic cell function experiments. The expression differences of miR-206, DNA Methyltransferase 3A (DNMT3A) and ECM-related genes were analyzed as needed by qRT-PCR or Western blot. TargetScan and dual-luciferase experiments were applied to predict and confirm the binding relationship between miR-206 and DNMT3A. The correlation between miR-206 and DNMT3A was analyzed in IH tissues by Pearson correlation coefficient, and further confirmed in HemECs by conducting rescue experiments. A nude mouse model of xenograft tumor was constructed to verify the results of in vitro experiments. MiR-206, which was downregulated in proliferative hemangioma, suppressed the malignant development of HemECs by regulating ECM-related genes. As the target gene of miR-206, DNMT3A was high-expressed in IH tissues and was negatively correlated with miR-206. Overexpressed DNMT3A counteracted the inhibitory effect of miR-206 mimic on HemECs and its regulatory effect on ECM. The results of in vivo experiments were consistent with those from cell experiments. Thus, miR-206 could promote ECM accumulation through targeted inhibition of DNMT3A, further inhibiting the malignant development of HemECs and relieving IH.
Collapse
Affiliation(s)
- Minliang Wu
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yong Chen
- Department of Plastic Surgery, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, Jiangsu, China
| | - Ling Feng
- Department of Pharmacy, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Haiying Dai
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Shuo Fang
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jianguo Xu
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
12
|
Pouw AE, Greiner MA, Coussa RG, Jiao C, Han IC, Skeie JM, Fingert JH, Mullins RF, Sohn EH. Cell-Matrix Interactions in the Eye: From Cornea to Choroid. Cells 2021; 10:687. [PMID: 33804633 PMCID: PMC8003714 DOI: 10.3390/cells10030687] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/03/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
Abstract
The extracellular matrix (ECM) plays a crucial role in all parts of the eye, from maintaining clarity and hydration of the cornea and vitreous to regulating angiogenesis, intraocular pressure maintenance, and vascular signaling. This review focuses on the interactions of the ECM for homeostasis of normal physiologic functions of the cornea, vitreous, retina, retinal pigment epithelium, Bruch's membrane, and choroid as well as trabecular meshwork, optic nerve, conjunctiva and tenon's layer as it relates to glaucoma. A variety of pathways and key factors related to ECM in the eye are discussed, including but not limited to those related to transforming growth factor-β, vascular endothelial growth factor, basic-fibroblastic growth factor, connective tissue growth factor, matrix metalloproteinases (including MMP-2 and MMP-9, and MMP-14), collagen IV, fibronectin, elastin, canonical signaling, integrins, and endothelial morphogenesis consistent of cellular activation-tubulogenesis and cellular differentiation-stabilization. Alterations contributing to disease states such as wound healing, diabetes-related complications, Fuchs endothelial corneal dystrophy, angiogenesis, fibrosis, age-related macular degeneration, retinal detachment, and posteriorly inserted vitreous base are also reviewed.
Collapse
Affiliation(s)
- Andrew E. Pouw
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA; (A.E.P.); (M.A.G.); (R.G.C.); (C.J.); (I.C.H.); (J.M.S.); (J.H.F.); (R.F.M.)
- Institute for Vision Research, University of Iowa, Iowa City, IA 52242, USA
| | - Mark A. Greiner
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA; (A.E.P.); (M.A.G.); (R.G.C.); (C.J.); (I.C.H.); (J.M.S.); (J.H.F.); (R.F.M.)
- Institute for Vision Research, University of Iowa, Iowa City, IA 52242, USA
| | - Razek G. Coussa
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA; (A.E.P.); (M.A.G.); (R.G.C.); (C.J.); (I.C.H.); (J.M.S.); (J.H.F.); (R.F.M.)
- Institute for Vision Research, University of Iowa, Iowa City, IA 52242, USA
| | - Chunhua Jiao
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA; (A.E.P.); (M.A.G.); (R.G.C.); (C.J.); (I.C.H.); (J.M.S.); (J.H.F.); (R.F.M.)
- Institute for Vision Research, University of Iowa, Iowa City, IA 52242, USA
| | - Ian C. Han
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA; (A.E.P.); (M.A.G.); (R.G.C.); (C.J.); (I.C.H.); (J.M.S.); (J.H.F.); (R.F.M.)
- Institute for Vision Research, University of Iowa, Iowa City, IA 52242, USA
| | - Jessica M. Skeie
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA; (A.E.P.); (M.A.G.); (R.G.C.); (C.J.); (I.C.H.); (J.M.S.); (J.H.F.); (R.F.M.)
| | - John H. Fingert
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA; (A.E.P.); (M.A.G.); (R.G.C.); (C.J.); (I.C.H.); (J.M.S.); (J.H.F.); (R.F.M.)
- Institute for Vision Research, University of Iowa, Iowa City, IA 52242, USA
| | - Robert F. Mullins
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA; (A.E.P.); (M.A.G.); (R.G.C.); (C.J.); (I.C.H.); (J.M.S.); (J.H.F.); (R.F.M.)
- Institute for Vision Research, University of Iowa, Iowa City, IA 52242, USA
| | - Elliott H. Sohn
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA; (A.E.P.); (M.A.G.); (R.G.C.); (C.J.); (I.C.H.); (J.M.S.); (J.H.F.); (R.F.M.)
- Institute for Vision Research, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
13
|
Dhavalikar P, Robinson A, Lan Z, Jenkins D, Chwatko M, Salhadar K, Jose A, Kar R, Shoga E, Kannapiran A, Cosgriff-Hernandez E. Review of Integrin-Targeting Biomaterials in Tissue Engineering. Adv Healthc Mater 2020; 9:e2000795. [PMID: 32940020 PMCID: PMC7960574 DOI: 10.1002/adhm.202000795] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/27/2020] [Indexed: 12/12/2022]
Abstract
The ability to direct cell behavior has been central to the success of numerous therapeutics to regenerate tissue or facilitate device integration. Biomaterial scientists are challenged to understand and modulate the interactions of biomaterials with biological systems in order to achieve effective tissue repair. One key area of research investigates the use of extracellular matrix-derived ligands to target specific integrin interactions and induce cellular responses, such as increased cell migration, proliferation, and differentiation of mesenchymal stem cells. These integrin-targeting proteins and peptides have been implemented in a variety of different polymeric scaffolds and devices to enhance tissue regeneration and integration. This review first presents an overview of integrin-mediated cellular processes that have been identified in angiogenesis, wound healing, and bone regeneration. Then, research utilizing biomaterials are highlighted with integrin-targeting motifs as a means to direct these cellular processes to enhance tissue regeneration. In addition to providing improved materials for tissue repair and device integration, these innovative biomaterials provide new tools to probe the complex processes of tissue remodeling in order to enhance the rational design of biomaterial scaffolds and guide tissue regeneration strategies.
Collapse
Affiliation(s)
- Prachi Dhavalikar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrew Robinson
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ziyang Lan
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Dana Jenkins
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Malgorzata Chwatko
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Karim Salhadar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Anupriya Jose
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ronit Kar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Erik Shoga
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Aparajith Kannapiran
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | | |
Collapse
|
14
|
Abstract
Microvasculature functions at the tissue and cell level, regulating local mass exchange of oxygen and nutrient-rich blood. While there has been considerable success in the biofabrication of large- and small-vessel replacements, functional microvasculature has been particularly challenging to engineer due to its size and complexity. Recently, three-dimensional bioprinting has expanded the possibilities of fabricating sophisticated microvascular systems by enabling precise spatiotemporal placement of cells and biomaterials based on computer-aided design. However, there are still significant challenges facing the development of printable biomaterials that promote robust formation and controlled 3D organization of microvascular networks. This review provides a thorough examination and critical evaluation of contemporary biomaterials and their specific roles in bioprinting microvasculature. We first provide an overview of bioprinting methods and techniques that enable the fabrication of microvessels. We then offer an in-depth critical analysis on the use of hydrogel bioinks for printing microvascularized constructs within the framework of current bioprinting modalities. We end with a review of recent applications of bioprinted microvasculature for disease modeling, drug testing, and tissue engineering, and conclude with an outlook on the challenges facing the evolution of biomaterials design for bioprinting microvasculature with physiological complexity.
Collapse
Affiliation(s)
- Ryan W. Barrs
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jia Jia
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Sophia E. Silver
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael Yost
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
15
|
Schwefel K, Spiegler S, Kirchmaier BC, Dellweg PKE, Much CD, Pané-Farré J, Strom TM, Riedel K, Felbor U, Rath M. Fibronectin rescues aberrant phenotype of endothelial cells lacking either CCM1, CCM2 or CCM3. FASEB J 2020; 34:9018-9033. [PMID: 32515053 DOI: 10.1096/fj.201902888r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/17/2020] [Accepted: 04/08/2020] [Indexed: 12/16/2022]
Abstract
Loss-of-function variants in CCM1/KRIT1, CCM2, and CCM3/PDCD10 are associated with autosomal dominant cerebral cavernous malformations (CCMs). CRISPR/Cas9-mediated CCM3 inactivation in human endothelial cells (ECs) has been shown to induce profound defects in cell-cell interaction as well as actin cytoskeleton organization. We here show that CCM3 inactivation impairs fibronectin expression and consequently leads to reduced fibers in the extracellular matrix. Despite the complexity and high molecular weight of fibronectin fibrils, our in vitro model allowed us to reveal that fibronectin supplementation restored aberrant spheroid formation as well as altered EC morphology, and suppressed actin stress fiber formation. Yet, fibronectin replacement neither enhanced the stability of tube-like structures nor inhibited the survival advantage of CCM3-/- ECs. Importantly, CRISPR/Cas9-mediated introduction of biallelic loss-of-function variants into either CCM1 or CCM2 demonstrated that the impaired production of a functional fibronectin matrix is a common feature of CCM1-, CCM2-, and CCM3-deficient ECs.
Collapse
Affiliation(s)
- Konrad Schwefel
- Department of Human Genetics, University Medicine Greifswald, Greifswald, Germany.,Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Stefanie Spiegler
- Department of Human Genetics, University Medicine Greifswald, Greifswald, Germany.,Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Bettina C Kirchmaier
- Institute of Cell Biology and Neuroscience, University of Frankfurt, Frankfurt am Main, Germany.,Buchmann Institute for Molecular Life Sciences, University of Frankfurt, Frankfurt am Main, Germany
| | - Patricia K E Dellweg
- Department of Human Genetics, University Medicine Greifswald, Greifswald, Germany.,Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Christiane D Much
- Department of Human Genetics, University Medicine Greifswald, Greifswald, Germany.,Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Jan Pané-Farré
- Department of Microbial Physiology and Molecular Biology, Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Tim M Strom
- Institute of Human Genetics, Technische Universität München, Munich, Germany.,Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Katharina Riedel
- Department of Microbial Physiology and Molecular Biology, Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Ute Felbor
- Department of Human Genetics, University Medicine Greifswald, Greifswald, Germany.,Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Matthias Rath
- Department of Human Genetics, University Medicine Greifswald, Greifswald, Germany.,Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| |
Collapse
|
16
|
van Dijk CG, Louzao-Martinez L, van Mulligen E, Boermans B, Demmers JA, van den Bosch TP, Goumans MJ, Duncker DJ, Verhaar MC, Cheng C. Extracellular Matrix Analysis of Human Renal Arteries in Both Quiescent and Active Vascular State. Int J Mol Sci 2020; 21:E3905. [PMID: 32486169 PMCID: PMC7313045 DOI: 10.3390/ijms21113905] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [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: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 01/30/2023] Open
Abstract
In vascular tissue engineering strategies, the addition of vascular-specific extracellular matrix (ECM) components may better mimic the in vivo microenvironment and potentially enhance cell-matrix interactions and subsequent tissue growth. For this purpose, the exact composition of the human vascular ECM first needs to be fully characterized. Most research has focused on characterizing ECM components in mature vascular tissue; however, the developing fetal ECM matches the active environment required in vascular tissue engineering more closely. Consequently, we characterized the ECM protein composition of active (fetal) and quiescent (mature) renal arteries using a proteome analysis of decellularized tissue. The obtained human fetal renal artery ECM proteome dataset contains higher levels of 15 ECM proteins versus the mature renal artery ECM proteome, whereas 16 ECM proteins showed higher levels in the mature tissue compared to fetal. Elastic ECM proteins EMILIN1 and FBN1 are significantly enriched in fetal renal arteries and are mainly produced by cells of mesenchymal origin. We functionally tested the role of EMILIN1 and FBN1 by anchoring the ECM secreted by vascular smooth muscle cells (SMCs) to glass coverslips. This ECM layer was depleted from either EMILIN1 or FBN1 by using siRNA targeting of the SMCs. Cultured endothelial cells (ECs) on this modified ECM layer showed alterations on the transcriptome level of multiple pathways, especially the Rho GTPase controlled pathways. However, no significant alterations in adhesion, migration or proliferation were observed when ECs were cultured on EMILIN1- or FNB1-deficient ECM. To conclude, the proteome analysis identified unique ECM proteins involved in the embryonic development of renal arteries. Alterations in transcriptome levels of ECs cultured on EMILIN1- or FBN1-deficient ECM showed that these candidate proteins could affect the endothelial (regenerative) response.
Collapse
Affiliation(s)
- Christian G.M. van Dijk
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands; (C.G.M.v.D.); (E.v.M.); (B.B.); (M.C.V.)
| | - Laura Louzao-Martinez
- Center for Proteomics, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands; (L.L.-M.); (J.A.A.D.)
| | - Elise van Mulligen
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands; (C.G.M.v.D.); (E.v.M.); (B.B.); (M.C.V.)
| | - Bart Boermans
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands; (C.G.M.v.D.); (E.v.M.); (B.B.); (M.C.V.)
| | - Jeroen A.A. Demmers
- Center for Proteomics, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands; (L.L.-M.); (J.A.A.D.)
| | | | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands;
| | - Dirk J. Duncker
- Experimental Cardiology, Department of Cardiology, Thorax center, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands;
| | - Marianne C. Verhaar
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands; (C.G.M.v.D.); (E.v.M.); (B.B.); (M.C.V.)
| | - Caroline Cheng
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, PO Box 85500, 3508 GA Utrecht, The Netherlands; (C.G.M.v.D.); (E.v.M.); (B.B.); (M.C.V.)
- Experimental Cardiology, Department of Cardiology, Thorax center, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands;
| |
Collapse
|
17
|
Medfai H, Khalil A, Rousseau A, Nuyens V, Paumann-Page M, Sevcnikar B, Furtmüller PG, Obinger C, Moguilevsky N, Peulen O, Herfs M, Castronovo V, Amri M, Van Antwerpen P, Vanhamme L, Zouaoui Boudjeltia K. Human peroxidasin 1 promotes angiogenesis through ERK1/2, Akt, and FAK pathways. Cardiovasc Res 2020; 115:463-475. [PMID: 29982533 DOI: 10.1093/cvr/cvy179] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 07/03/2018] [Indexed: 12/17/2022] Open
Abstract
Aims The term angiogenesis refers to sprouting of new blood vessels from pre-existing ones. The angiogenic process involves cell migration and tubulogenesis requiring interaction between endothelial cells and the extracellular matrix. Human peroxidasin 1 (hsPxd01) is a multidomain heme peroxidase found embedded in the basement membranes. As it promotes the stabilization of extracellular matrix, we investigated its possible role in angiogenesis both in vitro and in vivo. Methods and results We analysed the effects of peroxidasin 1 gene silencing and supplementation by recombinant hsPxd01 in TeloHAEC endothelial cells on cell migration, tubulogenesis in matrigel, and intracellular signal transduction as assessed by kinase phosphorylation and expression of pro-angiogenic genes as measured by qRT-PCR. We further evaluated the angiogenic potential of recombinant peroxidasin in a chicken chorioallantoic membrane model. RNA silencing of endogenous hsPxd01 significantly reduced tube formation and cell migration, whereas supplementation by the recombinant peroxidase promoted tube formation in vitro and stimulated vascularization in vivo through its catalytic activity. Moreover, recombinant hsPxd01 promoted phosphorylation of Extracellular signal-Regulated Kinases (ERK1/2), Protein kinase B (Akt), and Focal Adhesion Kinase (FAK), and induced the expression of pro-angiogenic downstream genes: Platelet Derived Growth Factor Subunit B (PDGFB), endothelial-derived Heparin Binding EGF-like growth factor (HB-EGF), CXCL-1, Hairy-Related Transcription Factor 1 (HEY-1), DNA-binding protein inhibitor (ID-2), Snail Family Zinc Finger 1 (SNAI-1), as well as endogenous hsPxd01. However, peroxidasin silencing significantly reduced Akt and FAK phosphorylation but induced ERK1/2 activation after supplementation by recombinant hsPxd01. While hsPxd01 silencing significantly reduced expression of HEY-1, ID-2, and PDGFB, it did not affect expression of SNAI-1, HB-EGF, and CXCL-1 after supplementation by recombinant hsPxd01. Conclusion Our findings suggest a role of enzymatically active peroxidasin 1 as a pro-angiogenic peroxidase and a modulator of ERK1/2, Akt and FAK signalling.
Collapse
Affiliation(s)
- Hayfa Medfai
- Laboratory of Experimental Medicine (ULB 222 Unit), Faculté de Médecine, CHU de Charleroi, A. Vésale Hospital, Université Libre de Bruxelles, Hôpital André Vésale, 706, Rue de Gozée, 6110 Montigny-le-Tilleul, Charleroi, Belgium.,Department of Biological Sciences, Laboratory of Functional Neurophysiology and Pathology, UR/11ES09, Université de Tunis El Manar, Faculté des Sciences de Tunis, 20 Rue de Tolède, 2092 Manar II, Tunis,Tunisia
| | - Alia Khalil
- Laboratory of Experimental Medicine (ULB 222 Unit), Faculté de Médecine, CHU de Charleroi, A. Vésale Hospital, Université Libre de Bruxelles, Hôpital André Vésale, 706, Rue de Gozée, 6110 Montigny-le-Tilleul, Charleroi, Belgium
| | - Alexandre Rousseau
- Laboratory of Experimental Medicine (ULB 222 Unit), Faculté de Médecine, CHU de Charleroi, A. Vésale Hospital, Université Libre de Bruxelles, Hôpital André Vésale, 706, Rue de Gozée, 6110 Montigny-le-Tilleul, Charleroi, Belgium
| | - Vincent Nuyens
- Laboratory of Experimental Medicine (ULB 222 Unit), Faculté de Médecine, CHU de Charleroi, A. Vésale Hospital, Université Libre de Bruxelles, Hôpital André Vésale, 706, Rue de Gozée, 6110 Montigny-le-Tilleul, Charleroi, Belgium
| | - Martina Paumann-Page
- Division of Biochemistry, Department of Chemistry, BOKU, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Benjamin Sevcnikar
- Division of Biochemistry, Department of Chemistry, BOKU, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Paul G Furtmüller
- Division of Biochemistry, Department of Chemistry, BOKU, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Christian Obinger
- Division of Biochemistry, Department of Chemistry, BOKU, University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Nicole Moguilevsky
- Laboratory of Experimental Medicine (ULB 222 Unit), Faculté de Médecine, CHU de Charleroi, A. Vésale Hospital, Université Libre de Bruxelles, Hôpital André Vésale, 706, Rue de Gozée, 6110 Montigny-le-Tilleul, Charleroi, Belgium
| | - Olivier Peulen
- Metastasis Research Laboratory, Giga-Cancer, University of Liege, Quartier Hopital, Avenue de l'Hopital, 11, 4000 Liège, Belgium
| | - Michael Herfs
- Department of Pathology, Laboratory of Experimental Pathology, Giga-Cancer, University of Liege, Quartier Hopital, Avenue de l'Hopital, 11, 4000 Liège, Belgium
| | - Vincent Castronovo
- Metastasis Research Laboratory, Giga-Cancer, University of Liege, Quartier Hopital, Avenue de l'Hopital, 11, 4000 Liège, Belgium
| | - Mohamed Amri
- Department of Biological Sciences, Laboratory of Functional Neurophysiology and Pathology, UR/11ES09, Université de Tunis El Manar, Faculté des Sciences de Tunis, 20 Rue de Tolède, 2092 Manar II, Tunis,Tunisia
| | - Pierre Van Antwerpen
- Pharmacognosy, Bioanalysis and Drug Discovery and Analytical Platform of the Faculty of Pharmacy, Faculty of Pharmacy, Université Libre de Bruxelles, Campus de la plaine CP205/09, Boulevard du Triomphe, 1050 Bruxelles, Belgium; and
| | - Luc Vanhamme
- Laboratory of Molecular Parasitology, IBMM, Faculty of Sciences, Université Libre de Bruxelles, Rue Adrienne Bolland 8, 6041 Gosselies, Belgium
| | - Karim Zouaoui Boudjeltia
- Laboratory of Experimental Medicine (ULB 222 Unit), Faculté de Médecine, CHU de Charleroi, A. Vésale Hospital, Université Libre de Bruxelles, Hôpital André Vésale, 706, Rue de Gozée, 6110 Montigny-le-Tilleul, Charleroi, Belgium
| |
Collapse
|
18
|
Sharma D, Ross D, Wang G, Jia W, Kirkpatrick SJ, Zhao F. Upgrading prevascularization in tissue engineering: A review of strategies for promoting highly organized microvascular network formation. Acta Biomater 2019; 95:112-130. [PMID: 30878450 DOI: 10.1016/j.actbio.2019.03.016] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/20/2019] [Accepted: 03/06/2019] [Indexed: 01/05/2023]
Abstract
Functional and perfusable vascular network formation is critical to ensure the long-term survival and functionality of engineered tissues after their transplantation. Although several vascularization strategies have been reviewed in past, the significance of microvessel organization in three-dimensional (3D) scaffolds has been largely ignored. Advances in high-resolution microscopy and image processing have revealed that the majority of tissues including cardiac, skeletal muscle, bone, and skin contain highly organized microvessels that orient themselves to align with tissue architecture for optimum molecular exchange and functional performance. Here, we review strategies to develop highly organized and mature vascular networks in engineered tissues, with a focus on electromechanical stimulation, surface topography, micro scaffolding, surface-patterning, microfluidics and 3D printing. This review will provide researchers with state of the art approaches to engineer vascularized functional tissues for diverse applications. STATEMENT OF SIGNIFICANCE: Vascularization is one of the critical challenges facing tissue engineering. Recent technological advances have enabled researchers to develop microvascular networks in engineered tissues. Although far from translational applications, current vascularization strategies have shown promising outcomes. This review emphasizes the most recent technological advances and future challenges for developing organized microvascular networks in vitro. The next critical step is to achieve highly perfusable, dense, mature and organized microvascular networks representative of native tissues.
Collapse
|
19
|
Klomp JE, Shaaya M, Matsche J, Rebiai R, Aaron JS, Collins KB, Huyot V, Gonzalez AM, Muller WA, Chew TL, Malik AB, Karginov AV. Time-Variant SRC Kinase Activation Determines Endothelial Permeability Response. Cell Chem Biol 2019; 26:1081-1094.e6. [PMID: 31130521 DOI: 10.1016/j.chembiol.2019.04.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 03/25/2019] [Accepted: 04/05/2019] [Indexed: 12/31/2022]
Abstract
In the current model of endothelial barrier regulation, the tyrosine kinase SRC is purported to induce disassembly of endothelial adherens junctions (AJs) via phosphorylation of VE cadherin, and thereby increase junctional permeability. Here, using a chemical biology approach to temporally control SRC activation, we show that SRC exerts distinct time-variant effects on the endothelial barrier. We discovered that the immediate effect of SRC activation was to transiently enhance endothelial barrier function as the result of accumulation of VE cadherin at AJs and formation of morphologically distinct reticular AJs. Endothelial barrier enhancement via SRC required phosphorylation of VE cadherin at Y731. In contrast, prolonged SRC activation induced VE cadherin phosphorylation at Y685, resulting in increased endothelial permeability. Thus, time-variant SRC activation differentially phosphorylates VE cadherin and shapes AJs to fine-tune endothelial barrier function. Our work demonstrates important advantages of synthetic biology tools in dissecting complex signaling systems.
Collapse
Affiliation(s)
- Jennifer E Klomp
- Department of Pharmacology, The University of Illinois College of Medicine, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| | - Mark Shaaya
- Department of Pharmacology, The University of Illinois College of Medicine, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| | - Jacob Matsche
- Department of Pharmacology, The University of Illinois College of Medicine, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| | - Rima Rebiai
- Department of Pharmacology, The University of Illinois College of Medicine, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| | - Jesse S Aaron
- Advanced Imaging Center at Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Kerrie B Collins
- Department of Pharmacology, The University of Illinois College of Medicine, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| | - Vincent Huyot
- Department of Pharmacology, The University of Illinois College of Medicine, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| | - Annette M Gonzalez
- Department of Pathology, The Feinberg School of Medicine at Northwestern University, Chicago, IL 60611, USA
| | - William A Muller
- Department of Pathology, The Feinberg School of Medicine at Northwestern University, Chicago, IL 60611, USA
| | - Teng-Leong Chew
- Advanced Imaging Center at Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Asrar B Malik
- Department of Pharmacology, The University of Illinois College of Medicine, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| | - Andrei V Karginov
- Department of Pharmacology, The University of Illinois College of Medicine, 835 S. Wolcott Avenue, Chicago, IL 60612, USA.
| |
Collapse
|
20
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
21
|
Abstract
During vascular development, endothelial cells (ECs) and neighboring stromal cells interact and communicate through autocrine and paracrine signaling mechanisms involving extracellular matrix (ECM) proteins and their cell surface integrin adhesion receptors. Integrin-mediated adhesion and signaling pathways are crucial for normal vascular development and physiology, and alterations in integrin expression and/or function drive several vascular-related pathologies including thrombosis, autoimmune disorders, and cancer. The purpose of this chapter is to discuss integrin adhesion and signaling pathways important for EC growth, survival, and migration. Integrin-mediated paracrine links between ECs and surrounding stromal cells in the organ microenvironment will also be discussed. Lastly, we will review roles for integrins in vascular pathologies and discuss possible targets for therapeutic intervention.
Collapse
Affiliation(s)
- Paola A Guerrero
- University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Joseph H McCarty
- University of Texas MD Anderson Cancer Center, Houston, TX, United States.
| |
Collapse
|
22
|
Rocca A, Tafuri D, Paccone M, Giuliani A, Zamboli AGI, Surfaro G, Paccone A, Compagna R, Amato M, Serra R, Amato B. Cell Based Therapeutic Approach in Vascular Surgery: Application and Review. Open Med (Wars) 2017; 12:308-322. [PMID: 29071303 PMCID: PMC5651406 DOI: 10.1515/med-2017-0045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 08/16/2017] [Indexed: 01/14/2023] Open
Abstract
Multipotent stem cells - such as mesenchymal stem/stromal cells and stem cells derived from different sources like vascular wall are intensely studied to try to rapidly translate their discovered features from bench to bedside. Vascular wall resident stem cells recruitment, differentiation, survival, proliferation, growth factor production, and signaling pathways transduced were analyzed. We studied biological properties of vascular resident stem cells and explored the relationship from several factors as Matrix Metalloproteinases (MMPs) and regulations of biological, translational and clinical features of these cells. In this review we described a translational and clinical approach to Adult Vascular Wall Resident Multipotent Vascular Stem Cells (VW-SCs) and reported their involvement in alternative clinical approach as cells based therapy in vascular disease like arterial aneurysms or peripheral arterial obstructive disease.
Collapse
Affiliation(s)
- Aldo Rocca
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, ItalyVia Sergio Pansini, 80131Naples, Italy
| | - Domenico Tafuri
- Department of Sport Sciences and Wellness, University of Naples “Parthenope”, Naples, Italy
| | - Marianna Paccone
- Department of Medicine and Health Sciences Vincenzo Tiberio, University of Molise, Campobasso, Italy
| | - Antonio Giuliani
- A.O.R.N. A. Cardarelli Hepatobiliary and Liver Transplatation Center, Naples, Italy
| | | | - Giuseppe Surfaro
- Antonio Cardarelli Hospital, General Surgery Unit, Campobasso, Italy
| | - Andrea Paccone
- Department of Medicine and Health Sciences Vincenzo Tiberio, University of Molise, Campobasso, Italy
| | - Rita Compagna
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Maurizo Amato
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Raffaele Serra
- Department of Medical and Surgical Sciences, University of Catanzaro, Catanzaro, Italy
| | - Bruno Amato
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
23
|
Yuan K, Orcholski ME, Huang NF, de Jesus Perez VA. In Vivo Study of Human Endothelial-Pericyte Interaction Using the Matrix Gel Plug Assay in Mouse. J Vis Exp 2016. [PMID: 28060266 DOI: 10.3791/54617] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Angiogenesis is the process by which new blood vessels are formed from existing vessels. New vessel growth requires coordinated endothelial cell proliferation, migration, and alignment to form tubular structures followed by recruitment of pericytes to provide mural support and facilitate vessel maturation. Current in vitro cell culture approaches cannot fully reproduce the complex biological environment where endothelial cells and pericytes interact to produce functional vessels. We present a novel application of the in vivo matrix gel plug assay to study endothelial-pericyte interactions and formation of functional blood vessels using severe combined immune deficiency mutation (SCID) mice. Briefly, matrix gel is mixed with a solution containing endothelial cells with or without pericytes followed by injection into the back of anesthetized SCID mice. After 14 days, the matrix gel plugs are removed, fixed and sectioned for histological analysis. The length, number, size and extent of pericyte coverage of mature vessels (defined by the presence of red blood cells in the lumen) can be quantified and compared between experimental groups using commercial statistical platforms. Beyond its use as an angiogenesis assay, this matrix gel plug assay can be used to conduct genetic studies and as a platform for drug discovery. In conclusion, this protocol will allow researchers to complement available in vitro assays for the study of endothelial-pericyte interactions and their relevance to either systemic or pulmonary angiogenesis.
Collapse
Affiliation(s)
- Ke Yuan
- Division of Pulmonary and Critical Care Medicine, School of Medicine, Stanford University; Stanford Cardiovascular Institute, School of Medicine, Stanford University
| | - Mark E Orcholski
- Division of Pulmonary and Critical Care Medicine, School of Medicine, Stanford University; Stanford Cardiovascular Institute, School of Medicine, Stanford University
| | - Ngan F Huang
- Stanford Cardiovascular Institute, School of Medicine, Stanford University; VA Palo Alto Health Care System, Department of Cardiothoracic Surgery, School of Medicine, Stanford University
| | - Vinicio A de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, School of Medicine, Stanford University; Stanford Cardiovascular Institute, School of Medicine, Stanford University;
| |
Collapse
|
24
|
Nakamichi M, Akishima-Fukasawa Y, Fujisawa C, Mikami T, Onishi K, Akasaka Y. Basic Fibroblast Growth Factor Induces Angiogenic Properties of Fibrocytes to Stimulate Vascular Formation during Wound Healing. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:3203-3216. [PMID: 27773739 DOI: 10.1016/j.ajpath.2016.08.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 07/27/2016] [Accepted: 08/19/2016] [Indexed: 11/17/2022]
Abstract
The role of fibrocytes in wound angiogenesis remains unclear. We therefore demonstrated the specific changes in fibrocyte accumulation for angiogesis in basic fibroblast growth factor (bFGF)-treated wounds. bFGF-treated wounds exhibited marked formation of arterioles and inhibition of podoplanin+ lymph vessels that were lacking in vascular endothelial growth factor-A-treated wounds. Real-time PCR in bFGF-treated wounds manifested enhanced expression of CD34, CD31, and bFGF mRNA and reduced expression of podoplanin and collagen type I, III, and IV mRNA. Double immunofluorescence staining focusing on fibrocyte detection in bFGF-treated wounds showed increased formation of capillary-like structures composed of CD34+/procollagen I+ fibrocytes, with a lack of capillary-like structures formed by CD45+/procollagen I+ or CD11b+/procollagen I+ fibrocytes. However, vascular endothelial growth factor-A-treated wounds lacked capillary-like structures composed of CD34+/procollagen I+ fibrocytes, with increased numbers of CD34+/fetal liver kinase-1+ endothelial progenitor cells. Furthermore, fibroblast growth factor receptor 1 siRNA injection into wounds, followed by bFGF, inhibited the formation of capillary-like structures composed of CD34+/procollagen I+ fibrocytes, together with inhibited mRNA expression of CD34 and CD31 and enhanced mRNA expression of collagen type I, indicating the requirements of bFGF/fibroblast growth factor receptor 1 system for capillary structure formation. This study highlights the angiogenic properties of CD34+/procollagen I+ fibrocytes specifically induced by bFGF, providing new insight into the active contribution of fibrocytes for vascular formation during wound healing.
Collapse
Affiliation(s)
- Miho Nakamichi
- Department of Plastic and Reconstructive Surgery, Toho University Omori Medical Center, Tokyo, Japan
| | | | - Chie Fujisawa
- Division of Research Promotion and Development, Advanced Research Center, Toho University, Tokyo, Japan
| | - Tetuo Mikami
- Department of Pathology, School of Medicine, Toho University, Tokyo, Japan
| | - Kiyoshi Onishi
- Department of Plastic and Reconstructive Surgery, Toho University Omori Medical Center, Tokyo, Japan
| | - Yoshikiyo Akasaka
- Department of Pathology, School of Medicine, Toho University, Tokyo, Japan; Regenerative Disease Research Unit, Advanced Research Center, Toho University, Tokyo, Japan.
| |
Collapse
|
25
|
Matrix viscoplasticity and its shielding by active mechanics in microtissue models: experiments and mathematical modeling. Sci Rep 2016; 6:33919. [PMID: 27671239 PMCID: PMC5037370 DOI: 10.1038/srep33919] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 09/06/2016] [Indexed: 12/30/2022] Open
Abstract
The biomechanical behavior of tissues under mechanical stimulation is critically important to physiological function. We report a combined experimental and modeling study of bioengineered 3D smooth muscle microtissues that reveals a previously unappreciated interaction between active cell mechanics and the viscoplastic properties of the extracellular matrix. The microtissues’ response to stretch/unstretch actuations, as probed by microcantilever force sensors, was dominated by cellular actomyosin dynamics. However, cell lysis revealed a viscoplastic response of the underlying model collagen/fibrin matrix. A model coupling Hill-type actomyosin dynamics with a plastic perfectly viscoplastic description of the matrix quantitatively accounts for the microtissue dynamics, including notably the cells’ shielding of the matrix plasticity. Stretch measurements of single cells confirmed the active cell dynamics, and were well described by a single-cell version of our model. These results reveal the need for new focus on matrix plasticity and its interactions with active cell mechanics in describing tissue dynamics.
Collapse
|
26
|
Steward AJ, Cole JH, Ligler FS, Loboa EG. Mechanical and Vascular Cues Synergistically Enhance Osteogenesis in Human Mesenchymal Stem Cells. Tissue Eng Part A 2016; 22:997-1005. [PMID: 27392567 DOI: 10.1089/ten.tea.2015.0533] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Development and maintenance of a vascular network are critical for bone growth and homeostasis; strategies that promote vascular function are critical for clinical success of tissue-engineered bone constructs. Co-culture of endothelial cells (ECs) with mesenchymal stem cells (MSCs) and exposure to 10% cyclic tensile strain have both been shown to regulate osteogenesis in isolation, but potential synergistic effects have yet to be explored. The objective of this study was to expose an MSC-EC co-culture to 10% cyclic tensile strain to examine the role of this mechanical stimulus on MSC-EC behavior. We hypothesized that paracrine signaling from ECs would stimulate osteogenesis of MSCs, and exposure to 10% cyclic tensile strain would enhance this anabolic signal. Human umbilical vein ECs and human bone marrow-derived MSCs were either monocultured or co-cultured at a 1:1 ratio in a mixed osteo/angiogenic medium, exposed to 10% cyclic tensile strain at 1 Hz for 4 h/day for 2 weeks, and biochemically and histologically analyzed for endothelial and osteogenic markers. While neither 10% cyclic tensile strain nor co-culture alone had a significant effect on osteogenesis, the concurrent application of strain to an MSC-EC co-culture resulted in a significant increase in calcium accretion and mineral deposition, suggesting that co-culture and strain synergistically enhance osteogenesis. Neither co-culture, 10% cyclic tensile strain, nor a combination of these stimuli affected endothelial markers, indicating that the endothelial phenotype remained stable, but unresponsive to the stimuli evaluated in this study. This study is the first to investigate the role of cyclic tensile strain on the complex interplay between ECs and MSCs in co-culture. The results of this study provide key insights into the synergistic effects of 10% cyclic tensile strain and co-culture on osteogenesis. Understanding mechanobiological factors affecting MSC-EC crosstalk will help enhance strategies for creating vascularized tissues in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Andrew J Steward
- 1 Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and North Carolina State University , Raleigh, North Carolina
| | - Jacqueline H Cole
- 1 Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and North Carolina State University , Raleigh, North Carolina
| | - Frances S Ligler
- 1 Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and North Carolina State University , Raleigh, North Carolina
| | - Elizabeth G Loboa
- 1 Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill and North Carolina State University , Raleigh, North Carolina.,2 College of Engineering, University of Missouri , Columbia, Missouri
| |
Collapse
|
27
|
Wollina U, Schmidt WD, Krönert C, Nelskamp C, Scheibe A, Fassler D. Some Effects of a Topical Collagen-Based Matrix on the Microcirculation and Wound Healing in Patients With Chronic Venous Leg Ulcers: Preliminary Observations. INT J LOW EXTR WOUND 2016; 4:214-24. [PMID: 16286373 DOI: 10.1177/1534734605283001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Themicrocirculation of the wound bed is a key parameter for improving granulation tissue formation and, hence, wound healing. The aim of this study was to determine whether a wound dressing comprising collagen/oxidized regenerated cellulose has effects over a short term on wound healing. Wounds were evaluated using a clinical wound score; pain associated with wounds was measured using a visual analogue scale. Wound microcirculation was evaluated using a technique based on noncontact remission spectroscopy. A prospective trial was performed in 40 patients with chronic venous leg ulcers (mean age 74 years; range, 43-93 years; 25 females and 15 males). Patients in group A were treated with PROMOGRAN® Matrix (Johnson& Johnson, New Brunswick, NJ) combined with “good” ulcer care for 2 weeks. Control group B consisted of 10 patientswho received only good ulcer care. The authors measured a favorable clinical response in 76.9% (group A) versus 66.7% (group B). Themean reduction of ulcer area was statistically significant in group A (P < .05). The wound score improved in group A from 2.28 ± 1.24 (before treatment) increasing to 3.72 ± 1.57 (after 1 week; P < .00023) and 4.92 ± 1.68 (after 2 weeks; P < .000027). In group B, the score improved from 1.44 ± 1.33 (before treatment) to 3.22 ± 1.30 (after 1 week; P < .0077). The mean visual analogue pain score before treatment was 8.72 (group A) and 7.88 (group B) (ns,P > .05). After 1 week of treatment, the score dropped to 5.76 (group A) and 6.66 (group B). In the second week, group A patients had a mean pain score of 3.84 compared with the pain score before treatment (P < .05). After 1 week of treatment, in group A there was a decrease in remission spectroscopy, which is considered to reflect an improvement in microcirculation.
Collapse
Affiliation(s)
- Uwe Wollina
- Department of Dermatology, Hospital Dresden-Friedrichstadt, Academic Teaching Hospital of the Technical University of Dresden, Germany.
| | | | | | | | | | | |
Collapse
|
28
|
Helal-Neto E, Brandão-Costa RM, Saldanha-Gama R, Ribeiro-Pereira C, Midlej V, Benchimol M, Morandi V, Barja-Fidalgo C. Priming Endothelial Cells With a Melanoma-Derived Extracellular Matrix Triggers the Activation of αvβ3/VEGFR2 Axis. J Cell Physiol 2016; 231:2464-73. [DOI: 10.1002/jcp.25358] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/22/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Edward Helal-Neto
- Laboratório de Farmacologia Celular e Molecular, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
- Laboratório de Biologia da Célula Endotelial e da Angiogênese, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
| | - Renata M. Brandão-Costa
- Laboratório de Farmacologia Celular e Molecular, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
| | - Roberta Saldanha-Gama
- Laboratório de Farmacologia Celular e Molecular, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
| | - Cristiane Ribeiro-Pereira
- Laboratório de Farmacologia Celular e Molecular, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
| | - Victor Midlej
- Instituto de Biofísica Carlos Chagas Filho; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
| | - Marlene Benchimol
- Instituto de Biofísica Carlos Chagas Filho; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
- Unigranrio; Universidade do Grande Rio; Rio de Janeiro Brazil
| | - Verônica Morandi
- Laboratório de Biologia da Célula Endotelial e da Angiogênese, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
| | - Christina Barja-Fidalgo
- Laboratório de Farmacologia Celular e Molecular, IBRAG; Universidade do Estado do Rio de Janeiro; Rio de Janeiro Brazil
| |
Collapse
|
29
|
Ribeiro AJS, Denisin AK, Wilson RE, Pruitt BL. For whom the cells pull: Hydrogel and micropost devices for measuring traction forces. Methods 2016; 94:51-64. [PMID: 26265073 PMCID: PMC4746112 DOI: 10.1016/j.ymeth.2015.08.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Revised: 07/10/2015] [Accepted: 08/06/2015] [Indexed: 01/16/2023] Open
Abstract
While performing several functions, adherent cells deform their surrounding substrate via stable adhesions that connect the intracellular cytoskeleton to the extracellular matrix. The traction forces that deform the substrate are studied in mechanotrasduction because they are affected by the mechanics of the extracellular milieu. We review the development and application of two methods widely used to measure traction forces generated by cells on 2D substrates: (i) traction force microscopy with polyacrylamide hydrogels and (ii) calculation of traction forces with arrays of deformable microposts. Measuring forces with these methods relies on measuring substrate displacements and converting them into forces. We describe approaches to determine force from displacements and elaborate on the necessary experimental conditions for this type of analysis. We emphasize device fabrication, mechanical calibration of substrates and covalent attachment of extracellular matrix proteins to substrates as key features in the design of experiments to measure cell traction forces with polyacrylamide hydrogels or microposts. We also report the challenges and achievements in integrating these methods with platforms for the mechanical stimulation of adherent cells. The approaches described here will enable new studies to understand cell mechanical outputs as a function of mechanical inputs and advance the understanding of mechanotransduction mechanisms.
Collapse
Affiliation(s)
- Alexandre J S Ribeiro
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, United States; Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, United States
| | - Aleksandra K Denisin
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, United States; Stanford Bioengineering, Stanford University, Stanford, CA 94305, United States
| | - Robin E Wilson
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, United States
| | - Beth L Pruitt
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, United States; Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, United States; Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA 94305, United States.
| |
Collapse
|
30
|
Liu CK, Dou Q, Liow SS, Kumar JN, Loh XJ. Cationic Micelles Based on Polyhedral Oligomeric Silsesquioxanes for Enhanced Gene Transfection. Aust J Chem 2016. [DOI: 10.1071/ch15636] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The synthesis and gene transfection efficiency of a series of amphiphilic copolymers, poly(2-(dimethylamino)ethyl methacrylate)-poly (methacrylate isobutyl polyhedral oligomeric silsesquioxane) (PDMAEMA-POSS) copolymers are reported. The hydrophobic POSS interior allows a cell-sensitizing drug such as paclitaxel to be incorporated, whereas the cationic and hydrophilic PDMAEMA corona allows the complexation of anionic DNA to form a nano-sized polyplex. These drug-encapsulated copolymers display excellent gene transfection efficiency compared with polyethylenimine or PDMAEMA homopolymers.
Collapse
|
31
|
Chin VT, Nagrial AM, Chou A, Biankin AV, Gill AJ, Timpson P, Pajic M. Rho-associated kinase signalling and the cancer microenvironment: novel biological implications and therapeutic opportunities. Expert Rev Mol Med 2015; 17:e17. [PMID: 26507949 PMCID: PMC4836205 DOI: 10.1017/erm.2015.17] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The Rho/ROCK pathway is involved in numerous pivotal cellular processes that have made it an area of intense study in cancer medicine, however, Rho-associated coiled-coil containing protein kinase (ROCK) inhibitors are yet to make an appearance in the clinical cancer setting. Their performance as an anti-cancer therapy has been varied in pre-clinical studies, however, they have been shown to be effective vasodilators in the treatment of hypertension and post-ischaemic stroke vasospasm. This review addresses the various roles the Rho/ROCK pathway plays in angiogenesis, tumour vascular tone and reciprocal feedback from the tumour microenvironment and explores the potential utility of ROCK inhibitors as effective vascular normalising agents. ROCK inhibitors may potentially enhance the delivery and efficacy of chemotherapy agents and improve the effectiveness of radiotherapy. As such, repurposing of these agents as adjuncts to standard treatments may significantly improve outcomes for patients with cancer. A deeper understanding of the controlled and dynamic regulation of the key components of the Rho pathway may lead to effective use of the Rho/ROCK inhibitors in the clinical management of cancer.
Collapse
Affiliation(s)
- Venessa T. Chin
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia
| | - Adnan M. Nagrial
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia
- The Department of Medical Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, NSW, Australia
| | - Angela Chou
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia
- Anatomical Pathology, Sydpath, St Vincent's Hospital, Sydney, Australia
| | - Andrew V. Biankin
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia
- Department of Surgery, Bankstown Hospital, Eldridge Road, Bankstown, Sydney, NSW 2200, Australia
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow, Scotland G61 1BD, UK
| | - Anthony J. Gill
- Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia
- University of Sydney, Sydney, NSW 2006, Australia
| | - Paul Timpson
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia
- Faculty of Medicine, St Vincent's Clinical School, University of NSW, Australia
| | - Marina Pajic
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia
- Faculty of Medicine, St Vincent's Clinical School, University of NSW, Australia
| |
Collapse
|
32
|
|
33
|
Park JA, Kim DY, Kim YM, Lee IK, Kwon YG. Endothelial Snail Regulates Capillary Branching Morphogenesis via Vascular Endothelial Growth Factor Receptor 3 Expression. PLoS Genet 2015; 11:e1005324. [PMID: 26147525 PMCID: PMC4493050 DOI: 10.1371/journal.pgen.1005324] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 05/29/2015] [Indexed: 11/19/2022] Open
Abstract
Vascular branching morphogenesis is activated and maintained by several signaling pathways. Among them, vascular endothelial growth factor receptor 2 (VEGFR2) signaling is largely presented in arteries, and VEGFR3 signaling is in veins and capillaries. Recent reports have documented that Snail, a well-known epithelial-to-mesenchymal transition protein, is expressed in endothelial cells, where it regulates sprouting angiogenesis and embryonic vascular development. Here, we identified Snail as a regulator of VEGFR3 expression during capillary branching morphogenesis. Snail was dramatically upregulated in sprouting vessels in the developing retinal vasculature, including the leading-edged vessels and vertical sprouting vessels for capillary extension toward the deep retina. Results from in vitro functional studies demonstrate that Snail expression colocalized with VEGFR3 and upregulated VEGFR3 mRNA by directly binding to the VEGFR3 promoter via cooperating with early growth response protein-1. Snail knockdown in postnatal mice attenuated the formation of the deep capillary plexus, not only by impairing vertical sprouting vessels but also by downregulating VEGFR3 expression. Collectively, these data suggest that the Snail-VEGFR3 axis controls capillary extension, especially in vessels expressing VEGFR2 at low levels.
Collapse
Affiliation(s)
- Jeong Ae Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Dong Young Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Young-Myeong Kim
- Vascular System Research Center, Kangwon National University, Kangwon-Do, Korea
| | - In-Kyu Lee
- Department of Internal Medicine, Kyungpook National University School of Medicine and Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Medical Center, Daegu, Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
- * E-mail:
| |
Collapse
|
34
|
Dietary L-arginine supplementation protects weanling pigs from deoxynivalenol-induced toxicity. Toxins (Basel) 2015; 7:1341-54. [PMID: 25884909 PMCID: PMC4417970 DOI: 10.3390/toxins7041341] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/01/2015] [Accepted: 04/07/2015] [Indexed: 12/20/2022] Open
Abstract
This study was conducted to determine the positive effects of dietary supplementation with l-arginine (Arg) on piglets fed a deoxynivalenol (DON)-contaminated diet. A total of eighteen, 28-day-old healthy weanling pigs were randomly assigned into one of three groups: uncontaminated basal diet (control group), 6 mg/kg DON-contaminated diet (DON group) and 6 mg/kg DON + 1% l-arginine (DON + ARG group). After 21 days of Arg supplementation, piglets in the DON and DON + ARG groups were challenged by feeding 6 mg/kg DON-contaminated diet for seven days. The results showed that DON resulted in damage to piglets. However, clinical parameters, including jejunal morphology, amino acid concentrations in the serum, jejunum and ileum, were improved by Arg (p < 0.05). Furthermore, the mRNA levels for sodium-glucose transporter-1 (SGLT-1), glucose transporter type-2 (GLUT-2) and y+l-type amino acid transporter-1 (y+LAT-1) were downregulated in the DON group, but the values were increased in the DON + ARG group (p < 0.05). Collectively, these results indicate that dietary supplementation with Arg exerts a protective role in pigs fed DON-contaminated diets.
Collapse
|
35
|
Amato B, Compagna R, Amato M, Grande R, Butrico L, Rossi A, Naso A, Ruggiero M, de Franciscis S, Serra R. Adult vascular wall resident multipotent vascular stem cells, matrix metalloproteinases, and arterial aneurysms. Stem Cells Int 2015; 2015:434962. [PMID: 25866513 PMCID: PMC4381852 DOI: 10.1155/2015/434962] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 02/23/2015] [Accepted: 03/06/2015] [Indexed: 12/20/2022] Open
Abstract
Evidences have shown the presence of multipotent stem cells (SCs) at sites of arterial aneurysms: they can differentiate into smooth muscle cells (SMCs) and are activated after residing in a quiescent state in the vascular wall. Recent studies have implicated the role of matrix metalloproteinases in the pathogenesis of arterial aneurysms: in fact the increased synthesis of MMPs by arterial SMCs is thought to be a pivotal mechanism in aneurysm formation. The factors and signaling pathways involved in regulating wall resident SC recruitment, survival, proliferation, growth factor production, and differentiation may be also related to selective expression of different MMPs. This review explores the relationship between adult vascular wall resident multipotent vascular SCs, MMPs, and arterial aneurysms.
Collapse
Affiliation(s)
- Bruno Amato
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology, Magna Graecia University of Catanzaro, Viale Europa, 88100 Catanzaro, Italy ; Department of Clinical Medicine and Surgery, University of Naples "Federico II", 80100 Naples, Italy
| | - Rita Compagna
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology, Magna Graecia University of Catanzaro, Viale Europa, 88100 Catanzaro, Italy ; Department of Clinical Medicine and Surgery, University of Naples "Federico II", 80100 Naples, Italy
| | - Maurizio Amato
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", 80100 Naples, Italy
| | - Raffaele Grande
- Department of Medical and Surgical Sciences, University of Catanzaro, 88100 Catanzaro, Italy
| | - Lucia Butrico
- Department of Medical and Surgical Sciences, University of Catanzaro, 88100 Catanzaro, Italy
| | - Alessio Rossi
- Department of Medicine and Health Sciences, University of Molise, 88100 Campobasso, Italy
| | - Agostino Naso
- Department of Medical and Surgical Sciences, University of Catanzaro, 88100 Catanzaro, Italy
| | - Michele Ruggiero
- Department of Medical and Surgical Sciences, University of Catanzaro, 88100 Catanzaro, Italy
| | - Stefano de Franciscis
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology, Magna Graecia University of Catanzaro, Viale Europa, 88100 Catanzaro, Italy ; Department of Medical and Surgical Sciences, University of Catanzaro, 88100 Catanzaro, Italy
| | - Raffaele Serra
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology, Magna Graecia University of Catanzaro, Viale Europa, 88100 Catanzaro, Italy ; Department of Medical and Surgical Sciences, University of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
36
|
Sankaran KK, Subramanian A, Krishnan UM, Sethuraman S. Nanoarchitecture of scaffolds and endothelial cells in engineering small diameter vascular grafts. Biotechnol J 2015; 10:96-108. [DOI: 10.1002/biot.201400415] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 10/27/2014] [Accepted: 12/03/2014] [Indexed: 12/14/2022]
|
37
|
Targeted delivery of extracellular matrix protected against neurologic defects after focal ischemia reperfusion in rats. J Stroke Cerebrovasc Dis 2014; 24:154-62. [PMID: 25440330 DOI: 10.1016/j.jstrokecerebrovasdis.2014.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 05/28/2014] [Accepted: 08/09/2014] [Indexed: 12/22/2022] Open
Abstract
Ischemic stroke is one of the leading causes of morbidity and mortality worldwide and characterized by defective angiogenesis. The functional sequences (RGDs, GRGDSPASSPISC) derived from fibronectin have been confirmed to augment angiogenesis in vivo and in vitro. However, delivery of peptides into the brain parenchyma has been hampered by the presence of the blood-brain barrier (BBB). We fused RGDs with penetratin (Antp) derived from Drosophila antennapedia homeodomain protein to improve the penetration of peptides through BBB into ischemic hemisphere. We found Antp-RGDs successfully not only penetrate the SH-SY5Y cells but also penetrated through BBB into ischemic hemisphere by intraperitoneal injection. In addition, application of Antp-RGDs to the focal cerebral ischemic reperfusion injury in rats resulted in the reduction of cerebral ischemic volume and the improvement of neurologic score according to the 21-point score. We further demonstrated that activation of phosphorylation-extracellular-signal related kinase 1/2 (p-ERK 1/2) and upregulation of gene VEGF resulted from post-treatment with Antp-RGDs 2 hours after reperfusion onset might at least partly contribute to the benefic changes after focal cerebral ischemic reperfusion injury in rats. Our data suggested that Antp-RGDs may serve as an attractive therapeutic intervention for treating ischemic stroke.
Collapse
|
38
|
Kodama Y, Ohkubo C, Kurosaki T, Egashira K, Sato K, Fumoto S, Nishida K, Higuchi N, Kitahara T, Nakamura T, Sasaki H. Secure and effective gene delivery system of plasmid DNA coated by polynucleotide. J Drug Target 2014; 23:43-51. [PMID: 25148610 DOI: 10.3109/1061186x.2014.950665] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Polynucleotides are anionic macromolecules which are expected to transfer into the targeted cells through specific uptake mechanisms. So, we developed polynucleotides coating complexes of plasmid DNA (pDNA) and polyethylenimine (PEI) for a secure and efficient gene delivery system and evaluated their usefulness. Polyadenylic acid (polyA), polyuridylic acid (polyU), polycytidylic acid (polyC), and polyguanylic acid (polyG) were examined as the coating materials. pDNA/PEI/polyA, pDNA/PEI/polyU, and pDNA/PEI/polyC complexes formed nanoparticles with a negative surface charge although pDNA/PEI/polyG was aggregated. The pDNA/PEI/polyC complex showed high transgene efficiency in B16-F10 cells although there was little efficiency in pDNA/PEI/polyA and pDNA/PEI/polyU complexes. An inhibition study strongly indicated the specific uptake mechanism of pDNA/PEI/polyC complex. Polynucleotide coating complexes had lower cytotoxicity than pDNA/PEI complex. The pDNA/PEI/polyC complex showed high gene expression selectively in the spleen after intravenous injection into mice. The pDNA/PEI/polyC complex showed no agglutination with erythrocytes and no acute toxicity although these were observed in pDNA/PEI complex. Thus, we developed polynucleotide coating complexes as novel vectors for clinical gene therapy, and the pDNA/PEI/polyC complex as a useful candidate for a gene delivery system.
Collapse
Affiliation(s)
- Yukinobu Kodama
- Department of Hospital Pharmacy, Nagasaki University Hospital , Nagasaki , Japan and
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Choi YS, Lee MY, David AE, Park YS. Nanoparticles for gene delivery: therapeutic and toxic effects. Mol Cell Toxicol 2014. [DOI: 10.1007/s13273-014-0001-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
40
|
Li R, Luo M, Ren M, Chen N, Xia J, Deng X, Zeng M, Yan K, Luo T, Wu J. Vitronectin regulation of vascular endothelial growth factor-mediated angiogenesis. J Vasc Res 2014; 51:110-7. [PMID: 24603119 DOI: 10.1159/000360085] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 01/22/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) plays a key role in regulating angiogenesis, and this process is largely dependent on the newly formed extracellular matrix (ECM). The levels of vitronectin (VN) are increased in patients with various cardiovascular diseases. A role for VN in regulating VEGF-induced angiogenesis has not been previously reported. We tested the hypothesis that VN regulates VEGFR-2 activation via effects on αvβ3, thus contributing to angiogenesis. METHODS We used a 3-dimensional angiogenesis assay, and examined the effects of VN on VEGF-mediated angiogenesis in aortic endothelial cells (ECs) isolated from wild-type and VN-deficient mice. RESULTS The addition of multimeric VN significantly enhanced VEGF-induced increases in EC migration and capillary formation. In vitro, Vn(-/-) ECs migrated significantly slower than wild-type ECs. The addition of VN to Vn(-/-) ECs increased EC migration and augmented the promigratory effect of VEGF in a manner that involved VEGFR-2 and Src signaling. Analysis of the mechanisms involved revealed that multimeric VN, but not monomeric VN, binds VEGF and enhances VEGF-induced VEGFR-2/Src activation in ECs. CONCLUSION These results underscore the importance of VN in the regulation of angiogenesis induced by VEGF.
Collapse
Affiliation(s)
- Rong Li
- Drug Discovery Research Center, Luzhou Medical College, Luzhou, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Clinical significance of L-type amino acid transporter 1 expression as a prognostic marker and potential of new targeting therapy in biliary tract cancer. BMC Cancer 2013; 13:482. [PMID: 24131658 PMCID: PMC4016614 DOI: 10.1186/1471-2407-13-482] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Accepted: 09/25/2013] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The expression of L-type amino acid transporter 1 (LAT1) has been described to play essential roles in tumor cell growth and survival. However, it remains unclear about the clinicopathological significance of LAT1 expression in biliary tract cancer. This study was conducted to determine biological significance of LAT1 expression and investigate whether LAT1 could be a prognostic biomarker for biliary tract cancer. METHODS A total of 139 consecutive patients with resected pathologic stage I-IV biliary tract adenocarcinoma were retrospectively reviewed. Tumor specimens were stained by immunohistochemistry for LAT1, Ki-67, microvessel density determined by CD34, and p53; and prognosis of patients was correlated. Biological significance of LAT1 expression was investigated by in vitro and in vivo experiments with LAT inhibitor, 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid (BCH) using cholangiocarcinoma cell line. RESULTS In total patients, high LAT1 expressions were recognized in 64.0%. The expression of LAT1 was closely correlated with lymphatic metastases, cell proliferation and angiogenesis, and was a significant indicator for predicting poor outcome after surgery. LAT1 expression was a significant independent predictor by multivariate analysis. Both in vitro and in vivo preliminary experiments indicated that BCH significantly suppressed growth of the tumor and yielded an additive therapeutic efficacy to gemcitabine and 5-FU. CONCLUSIONS High expression of LAT1 is a promising pathological marker to predict the outcome in patients with biliary tract adenocarcinoma. Inhibition of LAT1 may be an effective targeted therapy for this distressing disease.
Collapse
|
42
|
Nagao RJ, Ouyang Y, Keller R, Lee C, Suggs LJ, Schmidt CE. Preservation of Capillary-beds in Rat Lung Tissue Using Optimized Chemical Decellularization. J Mater Chem B 2013; 1:4801-4808. [PMID: 25558373 DOI: 10.1039/c3tb20640h] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Promoting regeneration using scaffolds created by decellularizing native tissue is becoming a popular technique applied to a variety of tissues. We demonstrate a method to decellularize highly vascular tissue keeping the vascular structure intact down to the capillary scale. Using vascular corrosion casting (VCC), we created a method for quantitatively assessing the functionality of vascular extracellular matrix (ECM) following decellularization. Murine lung tissue was decellularized using a number of techniques, then characterized using standard histological methods, as well as our quantitative VCC (qVCC) technique. Using an optimized acellular method, we successfully decellularized lung tissue while leaving behind a patent vascular network based on qualitative and quantitative histological methods.
Collapse
Affiliation(s)
- Ryan J Nagao
- Department of Biomedical Engineering, The University of Texas at Austin, 107 West Dean Keeton Street, Austin, Texas, TX 78712, United States of America
| | - Yafei Ouyang
- Department of Biomedical Engineering, The University of Texas at Austin, 107 West Dean Keeton Street, Austin, Texas, TX 78712, United States of America
| | - Renee Keller
- Department of Biomedical Engineering, The University of Texas at Austin, 107 West Dean Keeton Street, Austin, Texas, TX 78712, United States of America
| | - Curtis Lee
- Department of Biomedical Engineering, The University of Texas at Austin, 107 West Dean Keeton Street, Austin, Texas, TX 78712, United States of America
| | - Laura J Suggs
- Department of Biomedical Engineering, The University of Texas at Austin, 107 West Dean Keeton Street, Austin, Texas, TX 78712, United States of America
| | - Christine E Schmidt
- Department of Biomedical Engineering, The University of Texas at Austin, 107 West Dean Keeton Street, Austin, Texas, TX 78712, United States of America ; J. Crayton Pruitt Family Department of Biomedical Engineering, The University of Florida, Gainesville, FL 32611, United States of America
| |
Collapse
|
43
|
McConnell KI, Gomez EJ, Suh J. The identity of the cell adhesive protein substrate affects the efficiency of adeno-associated virus reverse transduction. Acta Biomater 2012; 8:4073-9. [PMID: 22771459 DOI: 10.1016/j.actbio.2012.06.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 06/25/2012] [Accepted: 06/27/2012] [Indexed: 11/25/2022]
Abstract
Delivering genes from surfaces, called substrate-mediated gene delivery or reverse transduction, is a useful method to achieve spatial localization of gene delivery. We tested the compatibility of adeno-associated virus (AAV) vectors with various cell adhesive proteins to mediate gene delivery from surfaces. Our studies demonstrate that AAV vectors can be successfully adsorbed on collagen I, elastin, and laminin substrates leading to robust gene delivery to overlying cells. Notably, AAV immobilization on laminin yields the highest efficiency of gene expression. This increased gene expression cannot be explained by increases in the levels of virus deposition, transcriptional activity of cells, or virus vector uptake into cells. Further refinement of our knowledge of AAV interactions with extracellular matrix proteins may have important implications in a variety of applications ranging from tissue engineering to in vivo gene therapy.
Collapse
|
44
|
Malik M, Segars J, Catherino WH. Integrin β1 regulates leiomyoma cytoskeletal integrity and growth. Matrix Biol 2012; 31:389-97. [PMID: 23023061 DOI: 10.1016/j.matbio.2012.09.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 09/14/2012] [Accepted: 09/18/2012] [Indexed: 11/17/2022]
Abstract
Uterine leiomyomas are characterized by an excessive extracellular matrix, increased mechanical stress, and increased active RhoA. Previously, we observed that mechanical signaling was attenuated in leiomyoma, but the mechanisms responsible remain unclear. Integrins, especially integrin β1, are transmembrane adhesion receptors that couple extracellular matrix stresses to the intracellular cytoskeleton to influence cell proliferation and differentiation. Here we characterized integrin and laminin to signaling in leiomyoma cells. We observed a 2.25±0.32 fold increased expression of integrin β1 in leiomyoma cells, compared to myometrial cells. Antibody-mediated inhibition of integrin β1 led to significant growth inhibition in leiomyoma cells and a loss of cytoskeletal integrity. Specifically, polymerization of actin filaments and formation of focal adhesions were reduced by inhibition of integrin β1. Inhibition of integrin β1 in leiomyoma cells led to 0.81±0.02 fold decrease in active RhoA, and resembled levels found in serum-starved cells. Likewise, inhibition of integrin β1 was accompanied by a decrease in phospho-ERK. Compared to myometrial cells, leiomyoma cells demonstrated increased expression of integrin α6 subunit to laminin receptor (1.91±0.11 fold), and increased expression of laminin 5α (1.52±0.02), laminin 5β (3.06±0.92), and laminin 5γ (1.66±0.06). Of note, leiomyoma cells grown on laminin matrix appear to realign themselves. Taken together, the findings reveal that the attenuated mechanical signaling in leiomyoma cells is accompanied by an increased expression and a dependence on integrin β1 signaling in leiomyoma cells, compared to myometrial cells.
Collapse
Affiliation(s)
- Minnie Malik
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States
| | | | | |
Collapse
|
45
|
|
46
|
Khan M, Ong ZY, Wiradharma N, Attia ABE, Yang YY. Advanced materials for co-delivery of drugs and genes in cancer therapy. Adv Healthc Mater 2012. [PMID: 23184770 DOI: 10.1002/adhm.201200109] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
With cancer being the major cause of mortality worldwide, the continued development of safe and efficacious treatments is warranted. A better understanding of the molecular mechanism and genetic basis of tumor initiation and progression, coupled with advances in chemistry, molecular biology and engineering have led to discovery of a wide range of therapeutic agents for cancer therapy. However, multidrug-resistance, which is mainly caused by malfunction of genes, has become a major problem in chemotherapy. To overcome this problem, the simultaneous delivery of genes to cancer cells has been proposed to correct the malfunctioned genes to sensitize the cells to chemotherapeutics. This progress report summarizes key advances in drug and gene delivery with focus on the development of polymers, peptides, liposomes and inorganic materials as nanocarriers for co-delivery of small molecular drugs and macromolecular genes or proteins. In addition, challenges and future perspectives in the design of nanocarriers for the co-delivery of therapeutic drugs and genes are discussed.
Collapse
Affiliation(s)
- Majad Khan
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, Singapore 138669
| | | | | | | | | |
Collapse
|
47
|
Li R, Ren M, Chen N, Luo M, Zhang Z, Wu J. Vitronectin increases vascular permeability by promoting VE-cadherin internalization at cell junctions. PLoS One 2012; 7:e37195. [PMID: 22606350 PMCID: PMC3350505 DOI: 10.1371/journal.pone.0037195] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 04/17/2012] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Cross-talk between integrins and cadherins regulates cell function. We tested the hypothesis that vitronectin (VN), a multi-functional adhesion molecule present in the extracellular matrix and plasma, regulates vascular permeability via effects on VE-cadherin, a critical regulator of endothelial cell (EC) adhesion. METHODOLOGY/PRINCIPAL FINDINGS Addition of multimeric VN (mult VN) significantly increased VE-cadherin internalization in human umbilical vein EC (HUVEC) monolayers. This effect was blocked by the anti-α(V)β(3) antibody, pharmacological inhibition and knockdown of Src kinase. In contrast to mult VN, monomeric VN did not trigger VE-cadherin internalization. In a modified Miles assay, VN deficiency impaired vascular endothelial growth factor-induced permeability. Furthermore, ischemia-induced enhancement of vascular permeability, expressed as the ratio of FITC-dextran leakage from the circulation into the ischemic and non-ischemic hindlimb muscle, was significantly greater in the WT mice than in the Vn(-/-) mice. Similarly, ischemia-mediated macrophage infiltration was significantly reduced in the Vn(-/-) mice vs. the WT controls. We evaluated changes in the multimerization of VN in ischemic tissue in a mouse hindlimb ischemia model. VN plays a previously unrecognized role in regulating endothelial permeability via conformational- and integrin-dependent effects on VE-cadherin trafficking. CONCLUSION/SIGNIFICANCE These results have important implications for the regulation of endothelial function and angiogenesis by VN under normal and pathological conditions.
Collapse
Affiliation(s)
- Rong Li
- Drug Discovery Research Center of Luzhou Medical College, Luzhou Medical College, Luzhou, Sichuan, People's Republic of China
| | - Meiping Ren
- Drug Discovery Research Center of Luzhou Medical College, Luzhou Medical College, Luzhou, Sichuan, People's Republic of China
| | - Ni Chen
- Drug Discovery Research Center of Luzhou Medical College, Luzhou Medical College, Luzhou, Sichuan, People's Republic of China
| | - Mao Luo
- Drug Discovery Research Center of Luzhou Medical College, Luzhou Medical College, Luzhou, Sichuan, People's Republic of China
| | - Zhuo Zhang
- Drug Discovery Research Center of Luzhou Medical College, Luzhou Medical College, Luzhou, Sichuan, People's Republic of China
| | - Jianbo Wu
- Drug Discovery Research Center of Luzhou Medical College, Luzhou Medical College, Luzhou, Sichuan, People's Republic of China
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States of America
| |
Collapse
|
48
|
Wendel C, Hemping-Bovenkerk A, Krasnyanska J, Mees ST, Kochetkova M, Stoeppeler S, Haier J. CXCR4/CXCL12 participate in extravasation of metastasizing breast cancer cells within the liver in a rat model. PLoS One 2012; 7:e30046. [PMID: 22253872 PMCID: PMC3258260 DOI: 10.1371/journal.pone.0030046] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 12/08/2011] [Indexed: 01/31/2023] Open
Abstract
INTRODUCTION Organ-specific composition of extracellular matrix proteins (ECM) is a determinant of metastatic host organ involvement. The chemokine CXCL12 and its receptor CXCR4 play important roles in the colonization of human breast cancer cells to their metastatic target organs. In this study, we investigated the effects of chemokine stimulation on adhesion and migration of different human breast cancer cell lines in vivo and in vitro with particular focus on the liver as a major metastatic site in breast cancer. METHODS Time lapse microscopy, in vitro adhesion and migration assays were performed under CXCL12 stimulation. Activation of small GTPases showed chemokine receptor signalling dependence from ECM components. The initial events of hepatic colonisation of MDA-MB-231 and MDA-MB-468 cells were investigated by intravital microscopy of the liver in a rat model and under shRNA inhibition of CXCR4. RESULTS In vitro, stimulation with CXCL12 induced increased chemotactic cell motility (p<0.05). This effect was dependent on adhesive substrates (type I collagen, fibronectin and laminin) and induced different responses in small GTPases, such as RhoA and Rac-1 activation, and changes in cell morphology. In addition, binding to various ECM components caused redistribution of chemokine receptors at tumour cell surfaces. In vivo, blocking CXCR4 decreased extravasation of highly metastatic MDA-MB-231 cells (p<0.05), but initial cell adhesion within the liver sinusoids was not affected. In contrast, the less metastatic MDA-MB-468 cells showed reduced cell adhesion but similar migration within the hepatic microcirculation. CONCLUSION Chemokine-induced extravasation of breast cancer cells along specific ECM components appears to be an important regulator but not a rate-limiting factor of their metastatic organ colonization.
Collapse
Affiliation(s)
- Claudia Wendel
- Department of General and Visceral Surgery, University Hospital Muenster, Muenster, Germany
| | | | - Julia Krasnyanska
- Department of General and Visceral Surgery, University Hospital Muenster, Muenster, Germany
| | - Sören Torge Mees
- Department of General and Visceral Surgery, University Hospital Muenster, Muenster, Germany
| | - Marina Kochetkova
- Chemokine Biology Division, School of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - Sandra Stoeppeler
- Department of General and Visceral Surgery, University Hospital Muenster, Muenster, Germany
| | - Jörg Haier
- Comprehensive Cancer Center Muenster, University Hospital Muenster, Muenster, Germany
- * E-mail:
| |
Collapse
|
49
|
Su SC, Bayless KJ. Utilizing sphingosine-1-phosphate to stimulate sprouting angiogenesis. Methods Mol Biol 2012; 874:201-13. [PMID: 22528450 DOI: 10.1007/978-1-61779-800-9_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In vitro models are useful for dissecting cell behavior under controlled conditions. Angiogenesis is a multistep process where endothelial cells (ECs) are activated by pro-angiogenic factors to degrade the basement membrane, migrate into the surrounding matrix, and form sprouting structures connecting neighboring vessels. Sphingosine-1-phosphate (S1P), a biologically active sphingolipid, promotes vessel morphogenesis and angiogenesis during embryonic development and in adults under normal and pathological conditions via its actions on ECs. Here, we describe an in vitro endothelial morphogenic assay that is significantly enhanced by S1P. This method allows for testing whether molecules and their related signaling pathways regulate the initiation of angiogenic sprouts stimulated by S1P, as well as whether individual compounds have pro- or anti-angiogenic properties.
Collapse
Affiliation(s)
- Shih-Chi Su
- Department of Molecular & Cellular Medicine, Texas A&M Health Science Center, College Station, TX, USA
| | | |
Collapse
|
50
|
Wu Y, He L, Zhang L, Chen J, Yi Z, Zhang J, Liu M, Pang X. Anacardic acid (6-pentadecylsalicylic acid) inhibits tumor angiogenesis by targeting Src/FAK/Rho GTPases signaling pathway. J Pharmacol Exp Ther 2011; 339:403-11. [PMID: 21828260 DOI: 10.1124/jpet.111.181891] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
Anacardic acid (6-pentadecylsalicylic acid), a natural inhibitor of histone acetyltransferase from Amphipterygium adstringens, has been shown to have anti-inflammatory, anticancer, antioxidative, and antimicrobial functions. However, whether this salicylic acid could block angiogenesis has not been elucidated to date. Here, we postulate that anacardic acid affects multiple steps of tumor angiogenesis to contribute to tumor inhibition. In this study, we found that vascular endothelial growth factor (VEGF)-induced cell proliferation, migration, and adhesion and capillary-like structure formation of primary cultured human umbilical vascular endothelial cells (HUVECs) could all be significantly suppressed by anacardic acid in vitro, without detectable cellular toxicity. Furthermore, anacardic acid effectively inhibited vascular development in chick embryo chorioallantoic membrane ex vivo (n = 10) and VEGF-triggered corneal neovascularization in vivo (n = 10). A mechanistic study revealed that anacardic acid blocked activities of Src and FAK kinases in concentration- and time-dependent manners in HUVECs, resulting in activation of RhoA-GTPase and inactivation of Rac1- and Cdc42-GTPases. Of note, when anacardic acid (2 mg/kg per day) was subcutaneously administrated to mice bearing human prostate tumor xenografts (n = 6-7), the volume and weight of solid tumors were significantly retarded. Src, Ki-67, and CD31 immunohistochemical staining further revealed that Src protein expression, tumor cell proliferation, and microvessel density could be remarkably suppressed by anacardic acid. Taken together, our findings demonstrate for the first time that anacardic acid functions as a potent tumor angiogenesis inhibitor by targeting the Src/FAK/Rho GTPase signaling pathway, leading to significant suppression of prostate tumor growth.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Rd., Shanghai 200241, China
| | | | | | | | | | | | | | | |
Collapse
|