1
|
Du YX, Li X, Ji SW, Niu N. Hypertension toxicity of VEGFR-TKIs in cancer treatment: incidence, mechanisms, and management strategies. Arch Toxicol 2024:10.1007/s00204-024-03874-4. [PMID: 39347999 DOI: 10.1007/s00204-024-03874-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024]
Abstract
Vascular endothelial growth factor receptor tyrosine kinase inhibitors (VEGFR-TKIs) are a class of targeted anticancer agents that include pazopanib, sunitinib, axitinib, and others. Currently, VEGFR-TKIs are widely used in the clinical treatment of various tumors, which can prolong patients' survival and even cure tumors. However, the use of VEGFR-TKIs is frequently associated with the occurrence of cardiovascular adverse events, with hypertension being the most prevalent. Hypertension and its complications can significantly impact the prognosis of patients, potentially jeopardizing their lives and resulting in the reduction or even cessation of treatment in severe cases. This review addresses the incidence of hypertension due to VEGFR-TKIs, mechanisms of toxicity, management strategies, and future research directions. In addition, hypertension due to VEGFR-TKIs may be associated with salt sensitivity, and possible mechanisms of hypertensive side effects are vasodilator imbalance, decreased capillary density, renal injury, impaired endothelial function due to oxidative stress, decreased lymphatic vascular density, and "off-target effect". A comprehensive understanding of hypertension toxicity due to cancer treatment with VEGFR-TKIs, can enhance clinical practice, thereby improving the prognostic outcomes of VEGFR-TKIs in oncology patients.
Collapse
Affiliation(s)
- Yan-Xi Du
- School of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, China
| | - Xu Li
- School of Pharmacy, North Sichuan Medical College, Nanchong, 637000, China
| | - Si-Wen Ji
- Office of Academic Affairs, North Sichuan Medical College, Nanchong, 637000, China
| | - Na Niu
- School of Pharmacy, North Sichuan Medical College, Nanchong, 637000, China.
| |
Collapse
|
2
|
Dumitru CS, Raica M. A Splice Form of VEGF, a Potential Anti-Angiogenetic Form of Head and Neck Squamous Cell Cancer Inhibition. Int J Mol Sci 2024; 25:8855. [PMID: 39201541 PMCID: PMC11354464 DOI: 10.3390/ijms25168855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/10/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Angiogenesis, primarily mediated by vascular endothelial growth factor (VEGF), is a fundamental step in the progression and metastasis of head and neck squamous cell carcinoma (HNSCC). Traditional anti-angiogenic therapies that target the VEGF pathway have shown promise but are often associated with significant side effects and variable efficacy due to the complexity of the angiogenic signaling pathway. This review highlights the potential of a specific VEGF splice form, VEGF165b, as an innovative therapeutic target for HNSCC. VEGF165b, unlike standard VEGF, is a natural inhibitor that binds to VEGF receptors without triggering pro-angiogenic signaling. Its distinct molecular structure and behavior suggest ways to modulate angiogenesis. This concept is particularly relevant when studying HNSCC, as introducing VEGF165b's anti-angiogenic properties offers a novel approach to understanding and potentially influencing the disease's dynamics. The review synthesizes experimental evidence suggesting the efficacy of VEGF165b in inhibiting tumor-induced angiogenesis and provides insight into a novel therapeutic strategy that could better manage HNSCC by selectively targeting aberrant vascular growth. This approach not only provides a potential pathway for more targeted and effective treatment options but also opens the door to a new paradigm in anti-angiogenic therapy with the possibility of reduced systemic toxicity. Our investigation is reshaping the future of HNSCC treatment by setting the stage for future research on VEGF splice variants as a tool for personalized medicine.
Collapse
Affiliation(s)
- Cristina Stefania Dumitru
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | | |
Collapse
|
3
|
Zhao C, Rong K, Liu P, Kong K, Li H, Zhang P, Chen X, Fu Q, Wang X. Preventing periprosthetic osteolysis in aging populations through lymphatic activation and stem cell-associated secretory phenotype inhibition. Commun Biol 2024; 7:962. [PMID: 39122919 PMCID: PMC11315686 DOI: 10.1038/s42003-024-06664-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
With increases in life expectancy, the number of patients requiring joint replacement therapy and experiencing periprosthetic osteolysis, the most common complication leading to implant failure, is growing or underestimated. In this study, we found that osteolysis progression and osteoclast differentiation in the surface of the skull bone of adult mice were accompanied by significant expansion of lymphatic vessels within bones. Using recombinant VEGF-C protein to activate VEGFR3 and promote proliferation of lymphatic vessels in bone, we counteracted excessive differentiation of osteoclasts and osteolysis caused by titanium alloy particles or inflammatory cytokines LPS/TNF-α. However, this effect was not observed in aged mice because adipogenically differentiated mesenchymal stem cells (MSCs) inhibited the response of lymphatic endothelial cells to agonist proteins. The addition of the JAK inhibitor ruxolitinib restored the response of lymphatic vessels to external stimuli in aged mice to protect against osteolysis progression. These findings suggest that inhibiting SASP secretion by adipogenically differentiated MSCs while activating lymphatic vessels in bone offers a new method to prevent periprosthetic osteolysis during joint replacement follow-up.
Collapse
Affiliation(s)
- Chen Zhao
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedics Implant, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kewei Rong
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedics Implant, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pengcheng Liu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Keyu Kong
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedics Implant, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haikuo Li
- Division of Biology & Biomedical Sciences, Washington University in St. Louis School of Medicine, St. Louis, USA
| | - Pu Zhang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedics Implant, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuzhuo Chen
- Department of Oral Surgery, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiang Fu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiaoqing Wang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedics Implant, the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
4
|
Virtej A, Marti L, Wagner M, Wiig H, Xue Y, Bletsa A, Berggreen E. Contribution of initial lymphatics to oral wound healing after tooth extraction. Eur J Oral Sci 2024; 132:e13006. [PMID: 38989803 DOI: 10.1111/eos.13006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 07/12/2024]
Abstract
Lymphatics are involved in the resolution of inflammation and wound healing, but their role in the oral wound healing process after tooth extraction has never been investigated. We therefore sought to evaluate the healing process following the extraction of maxillary molars in two transgenic mouse models: K14-VEGFR3-Ig mice, which lack initial mucosal lymphatic vessels, and K14-VEGFC mice, which have hyperplastic mucosal lymphatics. Maxillary molars were extracted from both transgenic mouse types and their corresponding wild-type (WT) controls. Mucosal and alveolar bone healing were evaluated. A delayed epithelialization and bone regeneration were observed in K14-VEGFR3-Ig mice compared with their WT littermates. The hampered wound closure was accompanied by decreased levels of epidermal growth factor (EGF) and persistent inflammation, characterized by infiltrates of immune cells and elevated levels of pro-inflammatory markers in the wounds. Hyperplastic mucosal lymphatics did not enhance the healing process after tooth extraction in K14-VEGFC mice. The findings indicate that initial mucosal lymphatics play a major role in the initial phase of the oral wound healing process.
Collapse
Affiliation(s)
- Anca Virtej
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Department of Clinical Dentistry, University of Bergen, Bergen, Norway
- Department of Oral Surgery, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Dentistry, The Arctic University of Tromsø, Faculty of Health Sciences, Tromsø, Norway
| | - Larissa Marti
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Marek Wagner
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Łukasiewicz Research Network - PORT Polish Center for Technology Development, Cancer Biomarkers Research Group, Wroclaw, Poland
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Ying Xue
- Department of Clinical Dentistry, University of Bergen, Bergen, Norway
- Department of Clinical Dentistry, The Arctic University of Tromsø, Faculty of Health Sciences, Tromsø, Norway
| | - Athanasia Bletsa
- Department of Clinical Dentistry, University of Bergen, Bergen, Norway
- Oral Centre of Expertise in Western Norway, Bergen, Norway
| | - Ellen Berggreen
- Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|
5
|
Dietrich-Ntoukas T, Bock F, Onderka J, Hos D, Bachmann BO, Zahn G, Cursiefen C. Selective, Temporary Postoperative Inhibition of Lymphangiogenesis by Integrin α5β1 Blockade Improves Allograft Survival in a Murine Model of High-Risk Corneal Transplantation. J Clin Med 2024; 13:4418. [PMID: 39124685 PMCID: PMC11313630 DOI: 10.3390/jcm13154418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/05/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Background: Corneal inflammatory hem- and lymphangiogenesis significantly increase the risk for immune rejection after subsequent allogeneic corneal transplantation. The purpose of this study was to analyze the impact of temporary selective inhibition of lymphangiogenesis after transplantation on graft survival. Methods: Allogeneic transplantation from C57BL/6 mice to BalbC mice was performed as "high-risk" keratoplasty in a prevascularized corneal host bed (suture-induced inflammatory corneal neovascularization). The treatment group received integrin α5β1-blocking small molecules (JSM6427) at the time of transplantation and for two weeks afterwards. Control mice received a vehicle solution. Grafts were evaluated weekly for graft rejection using an opacity score. At the end of the follow-up, immunohistochemical staining of corneal wholemounts for lymphatic vessels as well as CD11b+ immune cells was performed. Results: Temporary postoperative inhibition of lymphangiogenesis by JSM6427 improved the corneal graft survival significantly. At the end of the follow-up, no significant reduction in CD11b+ immunoreactive cells within the graft compared to controls was found. Conclusions: The significant improvement of corneal graft survival by the selective, temporary postoperative inhibition of lymphangiogenesis after keratoplasty using integrin antagonists shows the impact of lymphatic vessels in the early postoperative phase. Retarding lymphatic vessel ingrowth into the graft might be sufficient for the shift to immunological tolerance in the postoperative period, even after high-risk keratoplasty.
Collapse
Affiliation(s)
- Tina Dietrich-Ntoukas
- Department of Ophthalmology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, 13353 Berlin, Germany
- Department of Ophthalmology, University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Felix Bock
- Department of Ophthalmology, University Hospital of Cologne, 50937 Cologne, Germany; (F.B.); (D.H.); (B.O.B.); (C.C.)
| | - Jasmine Onderka
- Department of Ophthalmology, University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Deniz Hos
- Department of Ophthalmology, University Hospital of Cologne, 50937 Cologne, Germany; (F.B.); (D.H.); (B.O.B.); (C.C.)
| | - Bjoern O. Bachmann
- Department of Ophthalmology, University Hospital of Cologne, 50937 Cologne, Germany; (F.B.); (D.H.); (B.O.B.); (C.C.)
| | - Grit Zahn
- Eternygen GmbH, 10178 Berlin, Germany
| | - Claus Cursiefen
- Department of Ophthalmology, University Hospital of Cologne, 50937 Cologne, Germany; (F.B.); (D.H.); (B.O.B.); (C.C.)
| |
Collapse
|
6
|
Johannessen AL, Alstrup M, Hjortdal VE, Palmfeldt J, Offersen BV, Mohanakumar S. Increased Microvascular Filtration and Vascular Endothelial Growth Factor-D associated with Changed Lymphatic Vessel Morphology in Breast Cancer Treated Patients. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2024; 12:e5968. [PMID: 39036591 PMCID: PMC11259384 DOI: 10.1097/gox.0000000000005968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/01/2024] [Indexed: 07/23/2024]
Abstract
Background Vascular endothelial growth factors (VEGF) and inflammatory cytokines are indicated to be implicated in lymphedema development. We aimed to describe changes in microvascular filtration and VEGFs in a patient cohort vulnerable to breast cancer-related lymphedema development correlated with data on lymphatic morphology and function. Methods Consecutive node-positive breast cancer patients operated in the axilla and evaluated approximately 12 months after adjuvant locoregional radiotherapy were studied. Capillary filtration rate (CFR) and isovolumetric pressure of the arms were measured by strain gauge plethysmography, and 13 blood proteins were quantified by Luminex and Elisa technology in 28 patients and 18 healthy controls. Results The CFR was reduced in both arms from baseline to 1-year follow-up (ipsilateral: P = 0.016 and contralateral: P = 0.001). When stratifying lymphatic complications (morphologic abnormalities and/or breast cancer-related lymphedema), CFR reached a lower steady-state in the arms with normal morphology (I:P = 0.013 and C:P = 0.013) whereas the ipsilateral arm with lymphatic complications remained unchanged (P = 0.457). In patients with lymphatic abnormal vessels, the levels of VEGF-D were 86% higher than in patients with normal lymphatic vessels (P = 0.042), whereas levels of VEGFR-3 were 64% higher (P = 0.016). Conclusions Through one year of follow-up, CFR did not decrease in the lymphatic complicated treated arms as observed in noncomplicated treated arms. The patients had increased levels of VEGF-D and VEGFR-3. This correlation suggests that VEGF plays a role in the appearance of subcutaneous abnormal lymphatic vessels in the treated arms, which also maintain a fluid filtration/drainage mismatch up to one year after breast cancer treatment.
Collapse
Affiliation(s)
- Andreas L. Johannessen
- From the Department of Cardiothoracic and Vascular Surgery, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Vascular Surgery, Hospitalsenheden Midt, Viborg, Denmark
| | - Mathias Alstrup
- From the Department of Cardiothoracic and Vascular Surgery, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Vibeke E. Hjortdal
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Cardiothoracic Surgery, Rigshospitalet, Copenhagen, Denmark
| | - Johan Palmfeldt
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Research Unit for Molecular Medicine Research, Aarhus University Hospital, Aarhus, Denmark
| | - Birgitte V. Offersen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Sheyanth Mohanakumar
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Radiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Radiology, Regionshospitalet Horsens, Horsens, Denmark
| |
Collapse
|
7
|
Lu Z, Ding L, Jiang X, Zhang S, Yan M, Yang G, Tian X, Wang Q. Single-nucleus RNA transcriptome profiling reveals murine adipose tissue endothelial cell proliferation gene networks involved in obesity development. Arch Biochem Biophys 2024; 757:110029. [PMID: 38729594 DOI: 10.1016/j.abb.2024.110029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/18/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
Endothelial cells play an important role in the metabolism of adipose tissue (AT). This study aimed to analyze the changes that adipose tissue in AT endothelial cells undergo during the development of obesity, using single-nucleus RNA sequence (snRNA-seq). Mouse paraepididymal AT cells were subjected to snRNA-seq with the 10X Genomics platform. The cell types were then clustered using t-distributed stochastic neighbor embedding and unbiased computational informatics analyses. Protein-protein interactions network was established using the STRING database and visualized using Cytoscape. The dataset was subjected to differential gene enrichment analysis. In total, 21,333 cells acquired from 24 mouse paraepididymal AT samples were analyzed using snRNA-seq. This study identified 18 distinct clusters and annotated macrophages, fibroblasts, epithelial cells, T cells, endothelial cells, stem cells, neutrophil cells, and neutrophil cell types based on representative markers. Cluster 12 was defined as endothelial cells. The proportion of endothelial cells decreased with the development of obesity. Inflammatory factors, such as Vegfa and Prdm16 were upregulated in the medium obesity group but downregulated in the obesity group. Genes, such as Prox1, Erg, Flt4, Kdr, Flt1, and Pecam1 promoted the proliferation of AT endothelial cells and maintained the internal environment of AT. This study established a reference model and general framework for studying the mechanisms, biomarkers, and therapeutic targets of endothelial cell dysfunction-related diseases at the single-cell level.
Collapse
Affiliation(s)
- Zhimin Lu
- College of Sport and Health, Shandong Sport University, 250102, Jinan, China
| | - Ling Ding
- College of Sport and Health, Shandong Sport University, 250102, Jinan, China
| | - Xing Jiang
- College of Sport and Health, Shandong Sport University, 250102, Jinan, China
| | - Sen Zhang
- College of Sport and Health, Shandong Sport University, 250102, Jinan, China
| | - Min Yan
- College of Sport and Health, Shandong Sport University, 250102, Jinan, China
| | - Guangxin Yang
- College of Sport and Health, Shandong Sport University, 250102, Jinan, China
| | - Xuewen Tian
- College of Sport and Health, Shandong Sport University, 250102, Jinan, China.
| | - Qinglu Wang
- College of Sport and Health, Shandong Sport University, 250102, Jinan, China.
| |
Collapse
|
8
|
Hashem M, Mohandesi Khosroshahi E, Aliahmady M, Ghanei M, Soofi Rezaie Y, alsadat Jafari Y, rezaei F, Khodaparast eskadehi R, Kia Kojoori K, jamshidian F, Nabavi N, Rashidi M, Hasani Sadi F, Taheriazam A, Entezari M. Non-coding RNA transcripts, incredible modulators of cisplatin chemo-resistance in bladder cancer through operating a broad spectrum of cellular processes and signaling mechanism. Noncoding RNA Res 2024; 9:560-582. [PMID: 38515791 PMCID: PMC10955558 DOI: 10.1016/j.ncrna.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/12/2024] [Accepted: 01/14/2024] [Indexed: 03/23/2024] Open
Abstract
Bladder cancer (BC) is a highly frequent neoplasm in correlation with significant rate of morbidity, mortality, and cost. The onset of BC is predominantly triggered by environmental and/or occupational exposures to carcinogens, such as tobacco. There are two distinct pathways by which BC can be developed, including non-muscle-invasive papillary tumors (NMIBC) and non-papillary (or solid) muscle-invasive tumors (MIBC). The Cancer Genome Atlas project has further recognized key genetic drivers of MIBC along with its subtypes with particular properties and therapeutic responses; nonetheless, NMIBC is the predominant BC presentation among the suffering individuals. Radical cystoprostatectomy, radiotherapy, and chemotherapy have been verified to be the common therapeutic interventions in metastatic tumors, among which chemotherapeutics are more conventionally utilized. Although multiple chemo drugs have been broadly administered for BC treatment, cisplatin is reportedly the most effective chemo drug against the corresponding malignancy. Notwithstanding, tumor recurrence is usually occurred following the consumption of cisplatin regimens, particularly due to the progression of chemo-resistant trait. In this framework, non-coding RNAs (ncRNAs), as abundant RNA transcripts arise from the human genome, are introduced to serve as crucial contributors to tumor expansion and cisplatin chemo-resistance in bladder neoplasm. In the current review, we first investigated the best-known ncRNAs, i.e. microRNAs (miRNAs), long ncRNAs (lncRNAs), and circular RNAs (circRNAs), correlated with cisplatin chemo-resistance in BC cells and tissues. We noticed that these ncRNAs could mediate the BC-related cisplatin-resistant phenotype through diverse cellular processes and signaling mechanisms, reviewed here. Eventually, diagnostic and prognostic potential of ncRNAs, as well as their therapeutic capabilities were highlighted in regard to BC management.
Collapse
Affiliation(s)
- Mehrdad Hashem
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elaheh Mohandesi Khosroshahi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Melika Aliahmady
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Morvarid Ghanei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Yasamin Soofi Rezaie
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Yasamin alsadat Jafari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Fatemeh rezaei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Ramtin Khodaparast eskadehi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Kimia Kia Kojoori
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - faranak jamshidian
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Mohsen Rashidi
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Farzaneh Hasani Sadi
- General Practitioner, Kerman University of Medical Sciences, Kerman, 7616913555, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
9
|
Karakousi T, Mudianto T, Lund AW. Lymphatic vessels in the age of cancer immunotherapy. Nat Rev Cancer 2024; 24:363-381. [PMID: 38605228 DOI: 10.1038/s41568-024-00681-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/27/2024] [Indexed: 04/13/2024]
Abstract
Lymphatic transport maintains homeostatic health and is necessary for immune surveillance, and yet lymphatic growth is often associated with solid tumour development and dissemination. Although tumour-associated lymphatic remodelling and growth were initially presumed to simply expand a passive route for regional metastasis, emerging research puts lymphatic vessels and their active transport at the interface of metastasis, tumour-associated inflammation and systemic immune surveillance. Here, we discuss active mechanisms through which lymphatic vessels shape their transport function to influence peripheral tissue immunity and the current understanding of how tumour-associated lymphatic vessels may both augment and disrupt antitumour immune surveillance. We end by looking forward to emerging areas of interest in the field of cancer immunotherapy in which lymphatic vessels and their transport function are likely key players: the formation of tertiary lymphoid structures, immune surveillance in the central nervous system, the microbiome, obesity and ageing. The lessons learnt support a working framework that defines the lymphatic system as a key determinant of both local and systemic inflammatory networks and thereby a crucial player in the response to cancer immunotherapy.
Collapse
Affiliation(s)
- Triantafyllia Karakousi
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Tenny Mudianto
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA
| | - Amanda W Lund
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA.
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA.
| |
Collapse
|
10
|
Yin X, Zhang S, Lee JH, Dong H, Mourgkos G, Terwilliger G, Kraus A, Geraldo LH, Poulet M, Fischer S, Zhou T, Mohammed FS, Zhou J, Wang Y, Malloy S, Rohner N, Sharma L, Salinas I, Eichmann A, Thomas JL, Saltzman WM, Huttner A, Zeiss C, Ring A, Iwasaki A, Song E. Compartmentalized ocular lymphatic system mediates eye-brain immunity. Nature 2024; 628:204-211. [PMID: 38418880 PMCID: PMC10990932 DOI: 10.1038/s41586-024-07130-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/29/2024] [Indexed: 03/02/2024]
Abstract
The eye, an anatomical extension of the central nervous system (CNS), exhibits many molecular and cellular parallels to the brain. Emerging research demonstrates that changes in the brain are often reflected in the eye, particularly in the retina1. Still, the possibility of an immunological nexus between the posterior eye and the rest of the CNS tissues remains unexplored. Here, studying immune responses to herpes simplex virus in the brain, we observed that intravitreal immunization protects mice against intracranial viral challenge. This protection extended to bacteria and even tumours, allowing therapeutic immune responses against glioblastoma through intravitreal immunization. We further show that the anterior and posterior compartments of the eye have distinct lymphatic drainage systems, with the latter draining to the deep cervical lymph nodes through lymphatic vasculature in the optic nerve sheath. This posterior lymphatic drainage, like that of meningeal lymphatics, could be modulated by the lymphatic stimulator VEGFC. Conversely, we show that inhibition of lymphatic signalling on the optic nerve could overcome a major limitation in gene therapy by diminishing the immune response to adeno-associated virus and ensuring continued efficacy after multiple doses. These results reveal a shared lymphatic circuit able to mount a unified immune response between the posterior eye and the brain, highlighting an understudied immunological feature of the eye and opening up the potential for new therapeutic strategies in ocular and CNS diseases.
Collapse
Affiliation(s)
- Xiangyun Yin
- Department of Ophthalmology and Visual Science, Yale School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Sophia Zhang
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Ju Hyun Lee
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, CT, USA
| | - Huiping Dong
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - George Mourgkos
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Gordon Terwilliger
- Section of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Aurora Kraus
- Center of Evolutionary and Theoretical Immunology, Department of Biology, University of New Mexico, Albuquerque, NM, USA
| | - Luiz Henrique Geraldo
- Department of Internal Medicine, Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Mathilde Poulet
- Department of Internal Medicine, Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Suzanne Fischer
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Ting Zhou
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, China
| | - Farrah Shalima Mohammed
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, CT, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Jiangbing Zhou
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, CT, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Yongfu Wang
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Seth Malloy
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Nicolas Rohner
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Lokesh Sharma
- Section of Pulmonary and Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Irene Salinas
- Center of Evolutionary and Theoretical Immunology, Department of Biology, University of New Mexico, Albuquerque, NM, USA
| | - Anne Eichmann
- Department of Internal Medicine, Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
- Université de Paris, INSERM, PARCC, Paris, France
| | - Jean-Leon Thomas
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
- Department of Chemical & Environmental Engineering, Yale School of Engineering and Applied Science, New Haven, CT, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Anita Huttner
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Caroline Zeiss
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Aaron Ring
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Eric Song
- Department of Ophthalmology and Visual Science, Yale School of Medicine, New Haven, CT, USA.
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
11
|
Lian Z, Yu SR, Cui YX, Li SF, Su L, Song JX, Lee CY, Chen QX, Chen H. Rosuvastatin Enhances Lymphangiogenesis after Myocardial Infarction by Regulating the miRNAs/Vascular Endothelial Growth Factor Receptor 3 (miRNAs/VEGFR3) Pathway. ACS Pharmacol Transl Sci 2024; 7:335-347. [PMID: 38357274 PMCID: PMC10863446 DOI: 10.1021/acsptsci.3c00151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/24/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Several clinical studies have suggested that the early administration of statins could reduce the risk of in-hospital mortality in acute myocardial infarction (AMI) patients. Recently, some studies have identified that stimulating lymphangiogenesis after AMI could improve cardiac function by reducing myocardial edema and inflammation. This study aimed to identify the effect of rosuvastatin on postinfarct lymphangiogenesis and to identify the underlying mechanism of this effect. METHOD Myocardial infarction (MI) was induced by ligation of the left anterior descending coronary artery in mice orally administered rosuvastatin for 7 days. The changes in cardiac function, pathology, and lymphangiogenesis following MI were measured by echocardiography and immunostaining. EdU, Matrigel tube formation, and scratch wound assays were used to evaluate the effect of rosuvastatin on the proliferation, tube formation, and migration of the lymphatic endothelial cell line SVEC4-10. The expression of miR-107-3p, miR-491-5p, and VEGFR3 was measured by polymerase chain reaction (PCR) and Western blotting. A gain-of-function study was performed using miR-107-3p and miR-491-5p mimics. RESULTS The rosuvastatin-treated mice had a significantly improved ejection fraction and increased lymphatic plexus density 7 days after MI. Rosuvastatin also reduced myocardial edema and inflammatory response after MI. We used a VEGFR3 inhibitor to partially reverse these effects. Rosuvastatin promoted the proliferation, migration, and tube formation of SVEC4-10 cells. PCR and Western blot analyses revealed that rosuvastatin intervention downregulated miR-107-3p and miR-491-5p and promoted VEGFR3 expression. The gain-of-function study showed that miR-107-3p and miR-491-5p could inhibit the proliferation, migration, and tube formation of SVEC4-10 cells. CONCLUSION Rosuvastatin could improve heart function by promoting lymphangiogenesis after MI by regulating the miRNAs/VEGFR3 pathway.
Collapse
Affiliation(s)
- Zheng Lian
- Cardiovascular
Center, Beijing Tongren Hospital, Capital
Medical University, Xihuan South Road No. 2, Economic-Technological
Development Area, Beijing 100176, China
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Shi-Ran Yu
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Yu-Xia Cui
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Su-Fang Li
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Li−Na Su
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Jun-Xian Song
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Chong-Yoo Lee
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Qi-Xin Chen
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Hong Chen
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| |
Collapse
|
12
|
Ocskay Z, Bálint L, Christ C, Kahn ML, Jakus Z. CCBE1 regulates the development and prevents the age-dependent regression of meningeal lymphatics. Biomed Pharmacother 2024; 170:116032. [PMID: 38141283 DOI: 10.1016/j.biopha.2023.116032] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 12/25/2023] Open
Abstract
Recent studies have described the importance of lymphatics in numerous organ-specific physiological and pathological processes. The role of meningeal lymphatics in various neurological and cerebrovascular diseases has been suggested. It has also been shown that these structures develop postnatally and are altered by aging and that the vascular endothelial growth factor C (VEGFC)/ vascular endothelial growth factor receptor 3 (VEGFR3) signaling plays an essential role in the development and maintenance of them. However, the molecular mechanisms governing the development and maintenance of meningeal lymphatics are still poorly characterized. Recent in vitro cell culture-based experiments, and in vivo studies in zebrafish and mouse skin suggest that collagen and calcium binding EGF domains 1 (CCBE1) is involved in the processing of VEGFC. However, the organ-specific role of CCBE1 in developmental lymphangiogenesis and maintenance of lymphatics remains unclear. Here, we aimed to investigate the organ-specific functions of CCBE1 in developmental lymphangiogenesis and maintenance of meningeal lymphatics during aging. We demonstrate that inducible deletion of CCBE1 leads to impaired postnatal development of the meningeal lymphatics and decreased macromolecule drainage to deep cervical lymph nodes. The structural integrity and density of meningeal lymphatics are gradually altered during aging. Furthermore, the meningeal lymphatic structures in adults showed regression after inducible CCBE1 deletion. Collectively, our results indicate the importance of CCBE1-dependent mechanisms not only in the development, but also in the prevention of the age-related regression of meningeal lymphatics. Therefore, targeting CCBE1 may be a good therapeutic strategy to prevent age-related degeneration of meningeal lymphatics.
Collapse
Affiliation(s)
- Zsombor Ocskay
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
| | - László Bálint
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
| | - Carolin Christ
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
| | - Mark L Kahn
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA
| | - Zoltán Jakus
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary.
| |
Collapse
|
13
|
Jain A, Ang PS, Matrongolo MJ, Tischfield MA. Understanding the development, pathogenesis, and injury response of meningeal lymphatic networks through the use of animal models. Cell Mol Life Sci 2023; 80:332. [PMID: 37872442 PMCID: PMC11072018 DOI: 10.1007/s00018-023-04984-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/25/2023]
Abstract
Meningeal lymphatic vessels (MLVs) help maintain central nervous system (CNS) homeostasis via their ability to facilitate macromolecule waste clearance and neuroimmune trafficking. Although these vessels were overlooked for centuries, they have now been characterized in humans, non-human primates, and rodents. Recent studies in mice have explored the stereotyped growth and expansion of MLVs in dura mater, the various transcriptional, signaling, and environmental factors regulating their development and long-term maintenance, and the pathological changes these vessels undergo in injury, disease, or with aging. Key insights gained from these studies have also been leveraged to develop therapeutic approaches that help augment or restore MLV functions to improve brain health and cognition. Here, we review fundamental processes that control the development of peripheral lymphatic networks and how these might apply to the growth and expansion of MLVs in their unique meningeal environment. We also emphasize key findings in injury and disease models that may reveal additional insights into the plasticity of these vessels throughout the lifespan. Finally, we highlight unanswered questions and future areas of study that can further reveal the exciting therapeutic potential of meningeal lymphatics.
Collapse
Affiliation(s)
- Aditya Jain
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
- Child Health Institute of New Jersey, New Brunswick, NJ, 08901, USA
| | - Phillip S Ang
- University of Chicago Pritzker School of Medicine, Chicago, IL, 60637, USA
| | - Matthew J Matrongolo
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
- Child Health Institute of New Jersey, New Brunswick, NJ, 08901, USA
| | - Max A Tischfield
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA.
- Child Health Institute of New Jersey, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
14
|
Yang Y, Zhong J, Cui D, Jensen LD. Up-to-date molecular medicine strategies for management of ocular surface neovascularization. Adv Drug Deliv Rev 2023; 201:115084. [PMID: 37689278 DOI: 10.1016/j.addr.2023.115084] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 09/11/2023]
Abstract
Ocular surface neovascularization and its resulting pathological changes significantly alter corneal refraction and obstruct the light path to the retina, and hence is a major cause of vision loss. Various factors such as infection, irritation, trauma, dry eye, and ocular surface surgery trigger neovascularization via angiogenesis and lymphangiogenesis dependent on VEGF-related and alternative mechanisms. Recent advances in antiangiogenic drugs, nanotechnology, gene therapy, surgical equipment and techniques, animal models, and drug delivery strategies have provided a range of novel therapeutic options for the treatment of ocular surface neovascularization. In this review article, we comprehensively discuss the etiology and mechanisms of corneal neovascularization and other types of ocular surface neovascularization, as well as emerging animal models and drug delivery strategies that facilitate its management.
Collapse
Affiliation(s)
- Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Junmu Zhong
- Department of Ophthalmology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan 364000, Fujian Province, China
| | - Dongmei Cui
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen 518040, Guangdong Province, China
| | - Lasse D Jensen
- Department of Health, Medicine and Caring Sciences, Division of Diagnostics and Specialist Medicine, Unit of Cardiovascular Medicine, Linköping University, Linköping, Sweden.
| |
Collapse
|
15
|
Rauniyar K, Bokharaie H, Jeltsch M. Expansion and collapse of VEGF diversity in major clades of the animal kingdom. Angiogenesis 2023; 26:437-461. [PMID: 37017884 PMCID: PMC10328876 DOI: 10.1007/s10456-023-09874-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/17/2023] [Indexed: 04/06/2023]
Abstract
Together with the platelet-derived growth factors (PDGFs), the vascular endothelial growth factors (VEGFs) form the PDGF/VEGF subgroup among cystine knot growth factors. The evolutionary relationships within this subgroup have not been examined thoroughly to date. Here, we comprehensively analyze the PDGF/VEGF growth factors throughout all animal phyla and propose a phylogenetic tree. Vertebrate whole-genome duplications play a role in expanding PDGF/VEGF diversity, but several limited duplications are necessary to account for the temporal pattern of emergence. The phylogenetically oldest PDGF/VEGF-like growth factor likely featured a C-terminus with a BR3P signature, a hallmark of the modern-day lymphangiogenic growth factors VEGF-C and VEGF-D. Some younger VEGF genes, such as VEGFB and PGF, appeared completely absent in important vertebrate clades such as birds and amphibia, respectively. In contrast, individual PDGF/VEGF gene duplications frequently occurred in fish on top of the known fish-specific whole-genome duplications. The lack of precise counterparts for human genes poses limitations but also offers opportunities for research using organisms that diverge considerably from humans. Sources for the graphical abstract: 326 MYA and older [1]; 72-240 MYA [2]; 235-65 MYA [3].
Collapse
Affiliation(s)
- Khushbu Rauniyar
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Biocenter 2, (Viikinkaari 5E), P.O. Box. 56, 00790, Helsinki, Finland
| | - Honey Bokharaie
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Biocenter 2, (Viikinkaari 5E), P.O. Box. 56, 00790, Helsinki, Finland
| | - Michael Jeltsch
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Biocenter 2, (Viikinkaari 5E), P.O. Box. 56, 00790, Helsinki, Finland.
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Wihuri Research Institute, Helsinki, Finland.
- Helsinki One Health, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
16
|
Jannaway M, Iyer D, Mastrogiacomo DM, Li K, Sung DC, Yang Y, Kahn ML, Scallan JP. VEGFR3 is required for button junction formation in lymphatic vessels. Cell Rep 2023; 42:112777. [PMID: 37454290 PMCID: PMC10503778 DOI: 10.1016/j.celrep.2023.112777] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 06/01/2023] [Accepted: 06/25/2023] [Indexed: 07/18/2023] Open
Abstract
Lymphatic capillaries develop discontinuous cell-cell junctions that permit the absorption of large macromolecules, chylomicrons, and fluid from the interstitium. While excessive vascular endothelial growth factor 2 (VEGFR2) signaling can remodel and seal these junctions, whether and how VEGFR3 can alter lymphatic junctions remains incompletely understood. Here, we use lymphatic-specific Flt4 knockout mice to investigate VEGFR3 signaling in lymphatic junctions. We show that loss of Flt4 prevents specialized button junction formation in multiple tissues and impairs interstitial absorption. Knockdown of FLT4 in human lymphatic endothelial cells results in impaired NOTCH1 expression and activation, and overexpression of the NOTCH1 intracellular domain in Flt4 knockout vessels rescues the formation of button junctions and absorption of interstitial molecules. Together, our data reveal a requirement for VEGFR3 and NOTCH1 signaling in the development of button junctions during postnatal development and may hold clinical relevance to lymphatic diseases with impaired VEGFR3 signaling.
Collapse
Affiliation(s)
- Melanie Jannaway
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Drishya Iyer
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Diandra M Mastrogiacomo
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Kunyu Li
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Derek C Sung
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ying Yang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Mark L Kahn
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joshua P Scallan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
17
|
Salvador AFM, Dykstra T, Rustenhoven J, Gao W, Blackburn SM, Bhasiin K, Dong MQ, Guimarães RM, Gonuguntla S, Smirnov I, Kipnis J, Herz J. Age-dependent immune and lymphatic responses after spinal cord injury. Neuron 2023; 111:2155-2169.e9. [PMID: 37148871 PMCID: PMC10523880 DOI: 10.1016/j.neuron.2023.04.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 02/13/2023] [Accepted: 04/12/2023] [Indexed: 05/08/2023]
Abstract
Spinal cord injury (SCI) causes lifelong debilitating conditions. Previous works demonstrated the essential role of the immune system in recovery after SCI. Here, we explored the temporal changes of the response after SCI in young and aged mice in order to characterize multiple immune populations within the mammalian spinal cord. We revealed substantial infiltration of myeloid cells to the spinal cord in young animals, accompanied by changes in the activation state of microglia. In contrast, both processes were blunted in aged mice. Interestingly, we discovered the formation of meningeal lymphatic structures above the lesion site, and their role has not been examined after contusive injury. Our transcriptomic data predicted lymphangiogenic signaling between myeloid cells in the spinal cord and lymphatic endothelial cells (LECs) in the meninges after SCI. Together, our findings delineate how aging affects the immune response following SCI and highlight the participation of the spinal cord meninges in supporting vascular repair.
Collapse
Affiliation(s)
- Andrea Francesca M Salvador
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22903, USA
| | - Taitea Dykstra
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Justin Rustenhoven
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland 1023, New Zealand
| | - Wenqing Gao
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Susan M Blackburn
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kesshni Bhasiin
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Michael Q Dong
- Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Rafaela Mano Guimarães
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA; Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Sriharsha Gonuguntla
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Igor Smirnov
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Jonathan Kipnis
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA.
| | - Jasmin Herz
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
18
|
Ren Y, Okazaki T, Ngamnsae P, Hashimoto H, Ikeda R, Honkura Y, Suzuki J, Izumi SI. Anatomy and function of the lymphatic vessels in the parietal pleura and their plasticity under inflammation in mice. Microvasc Res 2023; 148:104546. [PMID: 37230165 DOI: 10.1016/j.mvr.2023.104546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/08/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023]
Abstract
Inflammatory pleuritis often causes pleural effusions, which are drained through lymphatic vessels (lymphatics) in the parietal pleura. The distribution of button- and zipper-like endothelial junctions can identify the subtypes of lymphatics, the initial, pre-collecting, and collecting lymphatics. Vascular endothelial growth factor receptor (VEGFR)-3 and its ligands VEGF-C/D are crucial lymphangiogenic factors. Currently, in the pleura covering the chest walls, the anatomy of the lymphatics and connecting networks of blood vessels are incompletely understood. Moreover, their pathological and functional plasticity under inflammation and the effects of VEGFR inhibition are unclear. This study aimed to learn the above-unanswered questions and immunostained mouse chest walls as whole-mount specimens. Confocal microscopic images and their 3-dimensional reconstruction analyzed the vasculatures. Repeated intra-pleural cavity lipopolysaccharide challenge induced pleuritis, which was also treated with VEGFR inhibition. Levels of vascular-related factors were evaluated by quantitative real-time polymerase chain reaction. We observed the initial lymphatics in the intercostals, collecting lymphatics under the ribs, and pre-collecting lymphatics connecting both. Arteries branched into capillaries and gathered into veins from the cranial to the caudal side. Lymphatics and blood vessels were in different layers with an adjacent distribution of the lymphatic layer to the pleural cavity. Inflammatory pleuritis elevated expression levels of VEGF-C/D and angiopoietin-2, induced lymphangiogenesis and blood vessel remodeling, and disorganized the lymphatic structures and subtypes. The disorganized lymphatics showed large sheet-like structures with many branches and holes inside. Such lymphatics were abundant in zipper-like endothelial junctions with some button-like junctions. The blood vessels were tortuous and had various diameters and complex networks. Stratified layers of lymphatics and blood vessels were disorganized, with impaired drainage function. VEGFR inhibition partially maintained their structures and drainage function. These findings demonstrate anatomy and pathological changes of the vasculatures in the parietal pleura and their potential as a novel therapeutic target.
Collapse
Affiliation(s)
- Yuzhuo Ren
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Tatsuma Okazaki
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan; Center for Dysphagia of Tohoku University Hospital, Sendai, Miyagi, Japan.
| | - Peerada Ngamnsae
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hikaru Hashimoto
- Center for Dysphagia of Tohoku University Hospital, Sendai, Miyagi, Japan; Department of Otolaryngology-Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-0872, Japan
| | - Ryoukichi Ikeda
- Center for Dysphagia of Tohoku University Hospital, Sendai, Miyagi, Japan; Department of Otolaryngology-Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-0872, Japan
| | - Yohei Honkura
- Center for Dysphagia of Tohoku University Hospital, Sendai, Miyagi, Japan; Department of Otolaryngology-Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-0872, Japan
| | - Jun Suzuki
- Center for Dysphagia of Tohoku University Hospital, Sendai, Miyagi, Japan; Department of Otolaryngology-Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-0872, Japan
| | - Shin-Ichi Izumi
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan; Center for Dysphagia of Tohoku University Hospital, Sendai, Miyagi, Japan; Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Biomedical Engineering, Sendai, Miyagi, Japan
| |
Collapse
|
19
|
Rütsche D, Nanni M, Rüdisser S, Biedermann T, Zenobi-Wong M. Enzymatically Crosslinked Collagen as a Versatile Matrix for In Vitro and In Vivo Co-Engineering of Blood and Lymphatic Vasculature. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2209476. [PMID: 36724374 DOI: 10.1002/adma.202209476] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/30/2022] [Indexed: 06/18/2023]
Abstract
Adequate vascularization is required for the successful translation of many in vitro engineered tissues. This study presents a novel collagen derivative that harbors multiple recognition peptides for orthogonal enzymatic crosslinking based on sortase A (SrtA) and Factor XIII (FXIII). SrtA-mediated crosslinking enables the rapid co-engineering of human blood and lymphatic microcapillaries and mesoscale capillaries in bulk hydrogels. Whereas tuning of gel stiffness determines the extent of neovascularization, the relative number of blood and lymphatic capillaries recapitulates the ratio of blood and lymphatic endothelial cells originally seeded into the hydrogel. Bioengineered capillaries readily form luminal structures and exhibit typical maturation markers both in vitro and in vivo. The secondary crosslinking enzyme Factor XIII is used for in situ tethering of the VEGF mimetic QK peptide to collagen. This approach supports the formation of blood and lymphatic capillaries in the absence of exogenous VEGF. Orthogonal enzymatic crosslinking is further used to bioengineer hydrogels with spatially defined polymer compositions with pro- and anti-angiogenic properties. Finally, macroporous scaffolds based on secondary crosslinking of microgels enable vascularization independent from supporting fibroblasts. Overall, this work demonstrates for the first time the co-engineering of mature micro- and meso-sized blood and lymphatic capillaries using a highly versatile collagen derivative.
Collapse
Affiliation(s)
- Dominic Rütsche
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zurich, Otto-Stern-Weg 7, Zurich, 8093, Switzerland
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, Schlieren, 8952, Switzerland
| | - Monica Nanni
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, Schlieren, 8952, Switzerland
- Institute for Mechanical Systems, Department of Mechanical and Process Engineering, ETH Zurich, Leonhardstrasse 21, Zurich, 8092, Switzerland
| | - Simon Rüdisser
- Biomolecular NMR Spectroscopy Platform, Department of Biology, ETH Zurich, Hönggerbergring 64, Zurich, 8093, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, Schlieren, 8952, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences & Technology, ETH Zurich, Otto-Stern-Weg 7, Zurich, 8093, Switzerland
| |
Collapse
|
20
|
Suarez AC, Hammel JH, Munson JM. Modeling lymphangiogenesis: Pairing in vitro and in vivo metrics. Microcirculation 2023; 30:e12802. [PMID: 36760223 PMCID: PMC10121924 DOI: 10.1111/micc.12802] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 01/20/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
Lymphangiogenesis is the mechanism by which the lymphatic system develops and expands new vessels facilitating fluid drainage and immune cell trafficking. Models to study lymphangiogenesis are necessary for a better understanding of the underlying mechanisms and to identify or test new therapeutic agents that target lymphangiogenesis. Across the lymphatic literature, multiple models have been developed to study lymphangiogenesis in vitro and in vivo. In vitro, lymphangiogenesis can be modeled with varying complexity, from monolayers to hydrogels to explants, with common metrics for characterizing proliferation, migration, and sprouting of lymphatic endothelial cells (LECs) and vessels. In comparison, in vivo models of lymphangiogenesis often use genetically modified zebrafish and mice, with in situ mouse models in the ear, cornea, hind leg, and tail. In vivo metrics, such as activation of LECs, number of new lymphatic vessels, and sprouting, mirror those most used in vitro, with the addition of lymphatic vessel hyperplasia and drainage. The impacts of lymphangiogenesis vary by context of tissue and pathology. Therapeutic targeting of lymphangiogenesis can have paradoxical effects depending on the pathology including lymphedema, cancer, organ transplant, and inflammation. In this review, we describe and compare lymphangiogenic outcomes and metrics between in vitro and in vivo studies, specifically reviewing only those publications in which both testing formats are used. We find that in vitro studies correlate well with in vivo in wound healing and development, but not in the reproductive tract or the complex tumor microenvironment. Considerations for improving in vitro models are to increase complexity with perfusable microfluidic devices, co-cultures with tissue-specific support cells, the inclusion of fluid flow, and pairing in vitro models of differing complexities. We believe that these changes would strengthen the correlation between in vitro and in vivo outcomes, giving more insight into lymphangiogenesis in healthy and pathological states.
Collapse
Affiliation(s)
- Aileen C. Suarez
- Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Department of Biomedical Engineering & Mechanics, Virginia Tech, Blacksburg, VA
| | - Jennifer H. Hammel
- Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Department of Biomedical Engineering & Mechanics, Virginia Tech, Blacksburg, VA
| | - Jennifer M. Munson
- Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Department of Biomedical Engineering & Mechanics, Virginia Tech, Blacksburg, VA
| |
Collapse
|
21
|
Promotion of Lymphangiogenesis by Targeted Delivery of VEGF-C Improves Diabetic Wound Healing. Cells 2023; 12:cells12030472. [PMID: 36766814 PMCID: PMC9913977 DOI: 10.3390/cells12030472] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/20/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
Chronic wounds represent a major therapeutic challenge. Lymphatic vessel function is impaired in chronic ulcers but the role of lymphangiogenesis in wound healing has remained unclear. We found that lymphatic vessels are largely absent from chronic human wounds as evaluated in patient biopsies. Excisional wound healing studies were conducted using transgenic mice with or without an increased number of cutaneous lymphatic vessels, as well as antibody-mediated inhibition of lymphangiogenesis. We found that a lack of lymphatic vessels mediated a proinflammatory wound microenvironment and delayed wound closure, and that the VEGF-C/VEGFR3 signaling axis is required for wound lymphangiogenesis. Treatment of diabetic mice (db/db mice) with the F8-VEGF-C fusion protein that targets the alternatively spliced extra domain A (EDA) of fibronectin, expressed in remodeling tissue, promoted wound healing, and potently induced wound lymphangiogenesis. The treatment also reduced tissue inflammation and exerted beneficial effects on the wound microenvironment, including myofibroblast density and collagen deposition. These findings indicate that activating the lymphatic vasculature might represent a new therapeutic strategy for treating chronic non-healing wounds.
Collapse
|
22
|
Biswas L, Chen J, De Angelis J, Singh A, Owen-Woods C, Ding Z, Pujol JM, Kumar N, Zeng F, Ramasamy SK, Kusumbe AP. Lymphatic vessels in bone support regeneration after injury. Cell 2023; 186:382-397.e24. [PMID: 36669473 DOI: 10.1016/j.cell.2022.12.031] [Citation(s) in RCA: 76] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 10/05/2022] [Accepted: 12/19/2022] [Indexed: 01/20/2023]
Abstract
Blood and lymphatic vessels form a versatile transport network and provide inductive signals to regulate tissue-specific functions. Blood vessels in bone regulate osteogenesis and hematopoiesis, but current dogma suggests that bone lacks lymphatic vessels. Here, by combining high-resolution light-sheet imaging and cell-specific mouse genetics, we demonstrate presence of lymphatic vessels in mouse and human bones. We find that lymphatic vessels in bone expand during genotoxic stress. VEGF-C/VEGFR-3 signaling and genotoxic stress-induced IL6 drive lymphangiogenesis in bones. During lymphangiogenesis, secretion of CXCL12 from proliferating lymphatic endothelial cells is critical for hematopoietic and bone regeneration. Moreover, lymphangiocrine CXCL12 triggers expansion of mature Myh11+ CXCR4+ pericytes, which differentiate into bone cells and contribute to bone and hematopoietic regeneration. In aged animals, such expansion of lymphatic vessels and Myh11-positive cells in response to genotoxic stress is impaired. These data suggest lymphangiogenesis as a therapeutic avenue to stimulate hematopoietic and bone regeneration.
Collapse
Affiliation(s)
- Lincoln Biswas
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK
| | - Junyu Chen
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jessica De Angelis
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK
| | - Amit Singh
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK; Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, Heidelberg D-69120, Germany
| | - Charlotte Owen-Woods
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK
| | - Zhangfan Ding
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Joan Mane Pujol
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK
| | - Naveen Kumar
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK
| | - Fanxin Zeng
- Department of Clinic Medical Center, Dazhou Central Hospital, Dazhou, China
| | - Saravana K Ramasamy
- MRC London Institute of Medical Sciences, Imperial College London, London W12 0NN, UK
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK.
| |
Collapse
|
23
|
Harris NR, Bálint L, Dy DM, Nielsen NR, Méndez HG, Aghajanian A, Caron KM. The ebb and flow of cardiac lymphatics: a tidal wave of new discoveries. Physiol Rev 2023; 103:391-432. [PMID: 35953269 PMCID: PMC9576179 DOI: 10.1152/physrev.00052.2021] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 06/16/2022] [Accepted: 07/18/2022] [Indexed: 12/16/2022] Open
Abstract
The heart is imbued with a vast lymphatic network that is responsible for fluid homeostasis and immune cell trafficking. Disturbances in the forces that regulate microvascular fluid movement can result in myocardial edema, which has profibrotic and proinflammatory consequences and contributes to cardiovascular dysfunction. This review explores the complex relationship between cardiac lymphatics, myocardial edema, and cardiac disease. It covers the revised paradigm of microvascular forces and fluid movement around the capillary as well as the arsenal of preclinical tools and animal models used to model myocardial edema and cardiac disease. Clinical studies of myocardial edema and their prognostic significance are examined in parallel to the recent elegant animal studies discerning the pathophysiological role and therapeutic potential of cardiac lymphatics in different cardiovascular disease models. This review highlights the outstanding questions of interest to both basic scientists and clinicians regarding the roles of cardiac lymphatics in health and disease.
Collapse
Affiliation(s)
- Natalie R Harris
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - László Bálint
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Danielle M Dy
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Natalie R Nielsen
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Hernán G Méndez
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Amir Aghajanian
- Division of Cardiology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
24
|
Mahapatra C, Kumar P, Paul MK, Kumar A. Angiogenic stimulation strategies in bone tissue regeneration. Tissue Cell 2022; 79:101908. [DOI: 10.1016/j.tice.2022.101908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/24/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022]
|
25
|
Arrigo A, Aragona E, Bandello F. VEGF-targeting drugs for the treatment of retinal neovascularization in diabetic retinopathy. Ann Med 2022; 54:1089-1111. [PMID: 35451900 PMCID: PMC9891228 DOI: 10.1080/07853890.2022.2064541] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Diabetic retinopathy (DR) is the most common microangiopathic complication of diabetes mellitus, representing a major cause of visual impairment in developed countries. Proliferative DR (PDR) represents the last stage of this extremely complex retinal disease, characterized by the development of neovascularization induced by the abnormal production and release of vascular endothelial growth factor (VEGF). The term VEGF includes different isoforms; VEGF-A represents one of the most important pathogenic factors of DR. Anti-VEGF intravitreal therapies radically changed the outcome of DR, due to combined anti-angiogenic and anti-edematous activities. Nowadays, several anti-VEGF molecules exist, characterized by different pharmacological features and duration. With respect to PDR, although anti-VEGF treatments represented a fundamental step forward in the management of this dramatic complication, a big debate is present in the literature regarding the role of anti-VEGF as substitute of panretinal photocoagulation or if these two approaches may be used in combination. In the present review, we provided an update on VEGF isoforms and their role in DR pathogenesis, on current anti-VEGF molecules and emerging new drugs, and on the current management strategies of PDR. There is an overall agreement regarding the relative advantage provided by anti-VEGF, especially looking at the management of PDR patients requiring vitrectomy, with respect to laser. Based on the current data, laser approaches might be avoided when a perfectly planned anti-VEGF therapeutic strategy can be adopted. Conversely, laser treatment may have a role for those patients unable to guarantee enough compliance to anti-VEGF injections.Key messagesVEGF increased production, stimulated by retinal hypoperfusion and ischaemia, is a major pathogenic factor of neovascular complication onset in diabetic retinopathy and of DR stages progression.Nowadays, several anti-VEGF molecules are available in clinical practice and other molecules are currently under investigation. Each anti-VEGF molecule is characterized by different targets and may interact with multiple biochemical pathways within the eye.All the data agreed in considering anti-VEGF molecules as a first line choice for the management of diabetic retinopathy. Laser treatments may have a role in selected advanced cases and for those patients unable to guarantee enough compliance to intravitreal treatments schemes.
Collapse
Affiliation(s)
- Alessandro Arrigo
- IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Emanuela Aragona
- IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco Bandello
- IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
26
|
Yang Y, Cao Y. The impact of VEGF on cancer metastasis and systemic disease. Semin Cancer Biol 2022; 86:251-261. [PMID: 35307547 DOI: 10.1016/j.semcancer.2022.03.011] [Citation(s) in RCA: 105] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 01/27/2023]
Abstract
Metastasis is the leading cause of cancer-associated mortality and the underlying mechanisms of cancer metastasis remain elusive. Both blood and lymphatic vasculatures are essential structures for mediating distal metastasis. The vasculature plays multiple functions, including accelerating tumor growth, sustaining the tumor microenvironment, supplying growth and invasive signals, promoting metastasis, and causing cancer-associated systemic disease. VEGF is one of the key angiogenic factors in tumors and participates in the initial stage of tumor development, progression and metastasis. Consequently, VEGF and its receptor-mediated signaling pathways have become one of the most important therapeutic targets for treating various cancers. Today, anti-VEGF-based antiangiogenic drugs (AADs) are widely used in the clinic for treating different types of cancer in human patients. Despite nearly 20-year clinical experience with AADs, the impact of these drugs on cancer metastasis and systemic disease remains largely unknown. In this review article, we focus our discussion on tumor VEGF in cancer metastasis and systemic disease and mechanisms underlying AADs in clinical benefits.
Collapse
Affiliation(s)
- Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Biomedicum, Karolinska Institute, 171 77 Stockholm, Sweden.
| |
Collapse
|
27
|
Thowsen IM, Reikvam T, Skogstrand T, Samuelsson AM, Müller DN, Tenstad O, Alitalo K, Karlsen T, Wiig H. Genetic Engineering of Lymphangiogenesis in Skin Does Not Affect Blood Pressure in Mouse Models of Salt-Sensitive Hypertension. Hypertension 2022; 79:2451-2462. [DOI: 10.1161/hypertensionaha.122.19777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Background:
Recent studies have indicated that sodium storage is influenced by macrophages that secrete VEGF-C (vascular endothelial growth factor) during salt stress thus stimulating lymphangiogenesis, thereby acting as a buffer against increased blood pressure (BP). We aimed to explore the role of dermal lymphatics in BP and sodium homeostasis. Our hypothesis was that mice with reduced dermal lymphatic vessels were more prone to develop salt-sensitive hypertension, and that mice with hyperplastic vessels were protected.
Methods:
Mice with either hypoplastic (Chy), absent (K14-VEGFR3 [vascular endothelial growth factor receptor 3]-Ig), or hyperplastic (K14-VEGF-C) dermal lymphatic vessels and littermate controls were given high-salt diet (4% NaCl in the chow), deoxycorticosterone acetate (DOCA)-salt diet and 1% saline to drink or nitric oxide blocker diet L-N
G
-nitro arginine methyl ester (followed by high salt diet). BP was measured by telemetric recording, and tissue sodium content by ion chromatography.
Results:
In contrast to previous studies, high salt diet did not induce an increase in BP or sodium storage in any of the mouse strains investigated. DOCA-salt, on the other hand, gave an increase in BP in Chy and K14-VEGFR3-Ig not different from their corresponding WT controls. DOCA induced salt storage in skin and muscle, but to the same extent in mice with dysfunctional lymphatic vessels and WT controls. Lymph flow as assessed by tracer washout was not affected by the diet in any of the mouse strains.
Conclusions:
Our results suggest that dermal lymphatic vessels are not involved in salt storage or blood pressure regulation in these mouse models of salt-sensitive hypertension.
Collapse
Affiliation(s)
- Irene Matre Thowsen
- Department of Biomedicine, University of Bergen, Norway (I.M.T., T.R., T.S., A.-M.S., O.T., T.K., H.W.)
| | - Tore Reikvam
- Department of Biomedicine, University of Bergen, Norway (I.M.T., T.R., T.S., A.-M.S., O.T., T.K., H.W.)
| | - Trude Skogstrand
- Department of Biomedicine, University of Bergen, Norway (I.M.T., T.R., T.S., A.-M.S., O.T., T.K., H.W.)
| | - Anne-Maj Samuelsson
- Department of Biomedicine, University of Bergen, Norway (I.M.T., T.R., T.S., A.-M.S., O.T., T.K., H.W.)
- Department of Medicine, Haukeland University Hospital, Bergen, Norway (A.-M.S.)
| | - Dominik N. Müller
- Experimental and Clinical Research Center, a cooperation of Charité - Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany (D.N.M.)
| | - Olav Tenstad
- Department of Biomedicine, University of Bergen, Norway (I.M.T., T.R., T.S., A.-M.S., O.T., T.K., H.W.)
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Medicine Program, Biomedicum Helsinki, University of Helsinki, Finland (K.A.)
| | - Tine Karlsen
- Department of Biomedicine, University of Bergen, Norway (I.M.T., T.R., T.S., A.-M.S., O.T., T.K., H.W.)
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, Norway (I.M.T., T.R., T.S., A.-M.S., O.T., T.K., H.W.)
| |
Collapse
|
28
|
Claudin-3 inhibits tumor-induced lymphangiogenesis via regulating the PI3K signaling pathway in lymphatic endothelial cells. Sci Rep 2022; 12:17440. [PMID: 36261482 PMCID: PMC9581975 DOI: 10.1038/s41598-022-22156-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 10/10/2022] [Indexed: 01/12/2023] Open
Abstract
Claudin-3 is a tight junction protein that has often been associated with the progression and metastasis of various tumors. Here, the role of claudin-3 in tumor-induced lymphangiogenesis is investigated. We found an increased lymphangiogenesis in the B16F10 tumor in claudin-3 knockout mice, accompanied by augmented melanoma cell metastasis into sentinel lymph nodes. In vitro, the overexpression of claudin-3 on lymphatic endothelial cells inhibited tube formation by suppressing cell migration, resulting in restricted lymphangiogenesis. Further experiments showed that claudin-3 inhibited lymphatic endothelial cell migration by regulating the PI3K signaling pathway. Interestingly, the expression of claudin-3 in lymphatic endothelial cells is down-regulated by vascular endothelial growth factor C that is often present in the tumor microenvironment. This study indicates that claudin-3 plays an important role as a signaling molecule in lymphatic endothelial cell activity associated with tumor lymphangiogenesis, which may further contribute to melanoma metastasis.
Collapse
|
29
|
Zhang M, Zhang H, Li Z, Bai L, Wang Q, Li J, Jiang M, Xue Q, Cheng N, Zhang W, Mao D, Chen Z, Huang J, Meng G, Chen Z, Chen SJ. Functional, structural, and molecular characterizations of the leukemogenic driver MEF2D-HNRNPUL1 fusion. Blood 2022; 140:1390-1407. [PMID: 35544603 PMCID: PMC9507012 DOI: 10.1182/blood.2022016241] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/03/2022] [Indexed: 12/02/2022] Open
Abstract
Recurrent MEF2D fusions with poor prognosis have been identified in B-cell precursor ALL (BCP-ALL). The molecular mechanisms underlying the pathogenic function of MEF2D fusions are poorly understood. Here, we show that MEF2D-HNRNPUL1 (MH) knock-in mice developed a progressive disease from impaired B-cell development at the pre-pro-B stage to pre-leukemia over 10 to 12 months. When cooperating with NRASG12D, MH drove an outbreak of BCP-ALL, with a more aggressive phenotype than the NRASG12D-induced leukemia. RNA-sequencing identified key networks involved in disease mechanisms. In chromatin immunoprecipitation-sequencing experiments, MH acquired increased chromatin-binding ability, mostly through MEF2D-responsive element (MRE) motifs in target genes, compared with wild-type MEF2D. Using X-ray crystallography, the MEF2D-MRE complex was characterized in atomic resolution, whereas disrupting the MH-DNA interaction alleviated the aberrant target gene expression and the B-cell differentiation arrest. The C-terminal moiety (HNRNPUL1 part) of MH was proven to contribute to the fusion protein's trans-regulatory activity, cofactor recruitment, and homodimerization. Furthermore, targeting MH-driven transactivation of the HDAC family by using the histone deacetylase inhibitor panobinostat in combination with chemotherapy improved the overall survival of MH/NRASG12D BCP-ALL mice. Altogether, these results not only highlight MH as an important driver in leukemogenesis but also provoke targeted intervention against BCP-ALL with MEF2D fusions.
Collapse
Affiliation(s)
- Ming Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai JiaoTong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai JiaoTong University, Shanghai, China
| | - Hao Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai JiaoTong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai JiaoTong University, Shanghai, China
| | - Zhihui Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai JiaoTong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai JiaoTong University, Shanghai, China
| | - Ling Bai
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai JiaoTong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai JiaoTong University, Shanghai, China
| | - Qianqian Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai JiaoTong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai JiaoTong University, Shanghai, China
| | - Jianfeng Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai JiaoTong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai JiaoTong University, Shanghai, China
| | - Minghao Jiang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai JiaoTong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai JiaoTong University, Shanghai, China
| | - Qing Xue
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai JiaoTong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai JiaoTong University, Shanghai, China
| | - Nuo Cheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai JiaoTong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai JiaoTong University, Shanghai, China
| | - Weina Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai JiaoTong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai JiaoTong University, Shanghai, China
| | - Dongdong Mao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai JiaoTong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai JiaoTong University, Shanghai, China
| | - Zhiming Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai JiaoTong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai JiaoTong University, Shanghai, China
| | - Jinyan Huang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai JiaoTong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai JiaoTong University, Shanghai, China
| | - Guoyu Meng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai JiaoTong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai JiaoTong University, Shanghai, China
| | - Zhu Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai JiaoTong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai JiaoTong University, Shanghai, China
| | - Sai-Juan Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai JiaoTong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai JiaoTong University, Shanghai, China
| |
Collapse
|
30
|
Matilla L, Martín-Núñez E, Garaikoetxea M, Navarro A, Vico JA, Arrieta V, García-Peña A, Fernández-Celis A, Gainza A, Álvarez V, Sádaba R, López-Andrés N, Jover E. Characterization of the sex-specific pattern of angiogenesis and lymphangiogenesis in aortic stenosis. Front Cardiovasc Med 2022; 9:971802. [PMID: 36172587 PMCID: PMC9510663 DOI: 10.3389/fcvm.2022.971802] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/11/2022] [Indexed: 11/25/2022] Open
Abstract
Objective We aim to analyze sex-related differences in angiogenesis and lymphangiogenesis in aortic valves (AVs) and valve interstitial cells (VICs) from aortic stenosis (AS) patients. Approach and Results Totally 230 patients (59% men) with severe AS undergoing surgical valve replacement were recruited. The density of total neovessels was higher in AVs from men as compared to women. Both small and medium neovessels were more abundant in men's AVs. Accordingly, male AVs exhibited higher CD31 and VE-cadherin expressions. The levels of the pro-angiogenic markers, such as vascular endothelial growth factor (VEGF)-A, VEGF receptor (VEGFR)1, VEGFR2, insulin-like growth factor-binding protein-2 (IGFBP-2), interleukin (IL)-8, chemerin, and fibroblast growth factor (FGF)-7, were increased in AVs from men. Transforming growth factor-β expression was higher in male AVs. The expression of antiangiogenic molecules thrombospondin (Tsp)-1, endostatin, and CD36 was upregulated in male AVs, although the levels of Tsp-2, IL-4, IL-12p70, and chondromodulin-1 were similar between both sexes. The number of lymphatic vessels and the expression of the lymphangiogenic markers Lyve-1 and D2-40 was higher in men's AV as well as VEGF-C, VEGF-D, and VEGFR3. Multivariate analyses adjusted for confounders further validated the sex-dependent expression of these targets. VICs isolated from men's AVs secreted higher amounts of the pro-angiogenic factors, VEGF-A, VEGFR1, IGFBP-2, and FGF-7, as well as the pro-lymphangiogenic factors, VEGF-C, VEGF-D, and VEGFR3, than women without changes in antiangiogenic markers. Conclusion Our data show that aberrant angiogenic and lymphangiogenic cues are over-represented in male AVs. Importantly, the VIC is a relevant source of multiple morphogens involved in angiogenesis and lymphangiogenesis likely endowing the AV of men with the predominant calcific AS phenotypes.
Collapse
|
31
|
Choi J, Choi E, Choi D. The ambivalent nature of the relationship between lymphatics and cancer. Front Cell Dev Biol 2022; 10:931335. [PMID: 36158182 PMCID: PMC9489845 DOI: 10.3389/fcell.2022.931335] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Do lymphatic vessels support cancer cells? Or are they vessels that help suppress cancer development? It is known that the lymphatic system is a vehicle for tumor metastasis and that the lymphangiogenic regulator VEGF-C supports the tumor. One such role of VEGF-C is the suppression of the immune response to cancer. The lymphatic system has also been correlated with an increase in interstitial fluid pressure of the tumor microenvironment. On the other hand, lymphatic vessels facilitate immune surveillance to mount an immune response against tumors with the support of VEGF-C. Furthermore, the activation of lymphatic fluid drainage may prove to filter and decrease tumor interstitial fluid pressure. In this review, we provide an overview of the dynamic between lymphatics, cancer, and tumor fluid pressure to suggest that lymphatic vessels may be used as an antitumor therapy due to their capabilities of immune surveillance and fluid pressure drainage. The application of this potential may help to prevent tumor proliferation or increase the efficacy of drugs that target cancer.
Collapse
|
32
|
Wasik A, Ratajczak-Wielgomas K, Badzinski A, Dziegiel P, Podhorska-Okolow M. The Role of Periostin in Angiogenesis and Lymphangiogenesis in Tumors. Cancers (Basel) 2022; 14:cancers14174225. [PMID: 36077762 PMCID: PMC9454705 DOI: 10.3390/cancers14174225] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Cancers are common diseases that affect people of all ages worldwide. For this reason, continuous attempts are being made to improve current therapeutic options. The formation of metastases significantly decreases patient survival. Therefore, understanding the mechanisms that are involved in this process seems to be crucial for effective cancer therapy. Cancer dissemination occurs mainly through blood and lymphatic vessels. As a result, many scientists have conducted a number of studies on the formation of new vessels. Many studies have shown that proangiogenic factors and the extracellular matrix protein, i.e., periostin, may be important in tumor angio- and lymphangiogenesis, thus contributing to metastasis formation and worsening of the prognosis. Abstract Periostin (POSTN) is a protein that is part of the extracellular matrix (ECM) and which significantly affects the control of intracellular signaling pathways (PI3K-AKT, FAK) through binding integrin receptors (αvβ3, αvβ5, α6β4). In addition, increased POSTN expression enhances the expression of VEGF family growth factors and promotes Erk phosphorylation. As a result, this glycoprotein controls the Erk/VEGF pathway. Therefore, it plays a crucial role in the formation of new blood and lymphatic vessels, which may be significant in the process of metastasis. Moreover, POSTN is involved in the proliferation, progression, migration and epithelial-mesenchymal transition (EMT) of tumor cells. Its increased expression has been detected in many cancers, including breast cancer, ovarian cancer, non-small cell lung carcinoma and glioblastoma. Many studies have shown that this protein may be an independent prognostic and predictive factor in many cancers, which may influence the choice of optimal therapy.
Collapse
Affiliation(s)
- Adrian Wasik
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Katarzyna Ratajczak-Wielgomas
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Correspondence:
| | - Arkadiusz Badzinski
- Silesian Nanomicroscopy Center, Silesia LabMed: Research and Implementation Center, Medical University of Silesia, 41-800 Zabrze, Poland
| | - Piotr Dziegiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Department of Human Biology, Wroclaw University of Health and Sport Sciences, 51-612 Wroclaw, Poland
| | - Marzenna Podhorska-Okolow
- Department of Human Biology, Wroclaw University of Health and Sport Sciences, 51-612 Wroclaw, Poland
- Department of Ultrastructural Research, Wroclaw Medical University, 50-368 Wroclaw, Poland
| |
Collapse
|
33
|
Sung C, Wang S, Hsu J, Yu R, Wong AK. Current Understanding of Pathological Mechanisms of Lymphedema. Adv Wound Care (New Rochelle) 2022; 11:361-373. [PMID: 34521256 PMCID: PMC9051876 DOI: 10.1089/wound.2021.0041] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Significance: Lymphedema is a common disease that affects hundreds of millions of people worldwide with significant financial and social burdens. Despite increasing prevalence and associated morbidities, the mainstay treatment of lymphedema is largely palliative without an effective cure due to incomplete understanding of the disease. Recent Advances: Recent studies have described key histological and pathological processes that contribute to the progression of lymphedema, including lymphatic stasis, inflammation, adipose tissue deposition, and fibrosis. This review aims to highlight cellular and molecular mechanisms involved in each of these pathological processes. Critical Issues: Despite recent advances in the understanding of the pathophysiology of lymphedema, cellular and molecular mechanisms underlying the disease remains elusive due to its complex nature. Future Directions: Additional research is needed to gain a better insight into the cellular and molecular mechanisms underlying the pathophysiology of lymphedema, which will guide the development of therapeutic strategies that target specific pathology of the disease.
Collapse
Affiliation(s)
- Cynthia Sung
- Keck School of Medicine of USC, Los Angeles, California, USA.,Division of Plastic Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Sarah Wang
- Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Jerry Hsu
- Division of Plastic Surgery, City of Hope National Medical Center, Duarte, California, USA.,Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, California, USA.,Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Roy Yu
- Keck School of Medicine of USC, Los Angeles, California, USA
| | - Alex K. Wong
- Division of Plastic Surgery, City of Hope National Medical Center, Duarte, California, USA.,Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, California, USA.,Correspondence: Division of Plastic Surgery, City of Hope National Medical Center, 1500 Duarte Road, Familian Science Building 1018, Duarte, CA 91010, USA.
| |
Collapse
|
34
|
Huang H, Chen T, Li F, Jin D, Li C, Yang Y, Liu X, Wang D, Di J. The functions, oncogenic roles, and clinical significance of circular RNAs in renal cell carcinoma. Med Oncol 2022; 39:72. [PMID: 35568747 DOI: 10.1007/s12032-022-01669-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 01/28/2022] [Indexed: 12/24/2022]
Abstract
Renal cell carcinoma (RCC) is the most common form of malignancy affecting the kidneys. Circular RNAs (circRNAs) are non-coding RNAs that are derived from exonic or intronic sequences through a selective shearing process. There is growing evidence that these circRNAs can influence a range of biological pathways by serving as protein decoys, microRNA sponges, regulators of transcriptional activity, or templates for protein translation. The dysregulation of circRNA expression patterns is a hallmark of RCC and other cancer types, and there is strong evidence that these RNA species can play central roles in the onset and progression of RCC tumors. In the present review, we summarized recent findings on the functional roles and clinical impacts of circRNAs in RCC. Further, we discussed their potential utility as diagnostic biomarkers or targets for therapeutic intervention.
Collapse
Affiliation(s)
- Hui Huang
- Department of Oncology, Kaizhou District People's Hospital of Chongqing, Chongqing, China
| | - Tao Chen
- Department of Osteology, Kaizhou District People's Hospital of Chongqing, Chongqing, China
| | - Fei Li
- Department of Osteology, Kaizhou District People's Hospital of Chongqing, Chongqing, China
| | - Dan Jin
- Department of Oncology, Kaizhou District People's Hospital of Chongqing, Chongqing, China
| | - Chuan Li
- Department of Oncology, Kaizhou District People's Hospital of Chongqing, Chongqing, China
| | - Yongbo Yang
- Department of Oncology, Kaizhou District People's Hospital of Chongqing, Chongqing, China
| | - Xuyang Liu
- Department of Oncology, Kaizhou District People's Hospital of Chongqing, Chongqing, China
| | - Dongmiao Wang
- Department of Oncology, Kaizhou District People's Hospital of Chongqing, Chongqing, China.
| | - Jiehui Di
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China. .,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China. .,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
35
|
Lymphatic Tissue Bioengineering for the Treatment of Postsurgical Lymphedema. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9040162. [PMID: 35447722 PMCID: PMC9025804 DOI: 10.3390/bioengineering9040162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/17/2022] [Accepted: 03/20/2022] [Indexed: 01/28/2023]
Abstract
Lymphedema is characterized by progressive and chronic tissue swelling and inflammation from local accumulation of interstitial fluid due to lymphatic injury or dysfunction. It is a debilitating condition that significantly impacts a patient's quality of life, and has limited treatment options. With better understanding of the molecular mechanisms and pathophysiology of lymphedema and advances in tissue engineering technologies, lymphatic tissue bioengineering and regeneration have emerged as a potential therapeutic option for postsurgical lymphedema. Various strategies involving stem cells, lymphangiogenic factors, bioengineered matrices and mechanical stimuli allow more precisely controlled regeneration of lymphatic tissue at the site of lymphedema without subjecting patients to complications or iatrogenic injuries associated with surgeries. This review provides an overview of current innovative approaches of lymphatic tissue bioengineering that represent a promising treatment option for postsurgical lymphedema.
Collapse
|
36
|
El-Sammak H, Yang B, Guenther S, Chen W, Marín-Juez R, Stainier DY. A Vegfc-Emilin2a-Cxcl8a Signaling Axis Required for Zebrafish Cardiac Regeneration. Circ Res 2022; 130:1014-1029. [PMID: 35264012 PMCID: PMC8976759 DOI: 10.1161/circresaha.121.319929] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Ischemic heart disease following the obstruction of coronary vessels leads to the death of cardiac tissue and the formation of a fibrotic scar. In contrast to adult mammals, zebrafish can regenerate their heart after injury, enabling the study of the underlying mechanisms. One of the earliest responses following cardiac injury in adult zebrafish is coronary revascularization. Defects in this process lead to impaired cardiomyocyte repopulation and scarring. Hence, identifying and investigating factors that promote coronary revascularization holds great therapeutic potential. METHODS We used wholemount imaging, immunohistochemistry and histology to assess various aspects of zebrafish cardiac regeneration. Deep transcriptomic analysis allowed us to identify targets and potential effectors of Vegfc (vascular endothelial growth factor C) signaling. We used newly generated loss- and gain-of-function genetic tools to investigate the role of Emilin2a (elastin microfibril interfacer 2a) and Cxcl8a (chemokine (C-X-C) motif ligand 8a)-Cxcr1 (chemokine (C-X-C) motif receptor 1) signaling in cardiac regeneration. RESULTS We first show that regenerating coronary endothelial cells upregulate vegfc upon cardiac injury in adult zebrafish and that Vegfc signaling is required for their proliferation during regeneration. Notably, blocking Vegfc signaling also significantly reduces cardiomyocyte dedifferentiation and proliferation. Using transcriptomic analyses, we identified emilin2a as a target of Vegfc signaling and found that manipulation of emilin2a expression can modulate coronary revascularization as well as cardiomyocyte proliferation. Mechanistically, Emilin2a induces the expression of the chemokine gene cxcl8a in epicardium-derived cells, while cxcr1, the Cxcl8a receptor gene, is expressed in coronary endothelial cells. We further show that Cxcl8a-Cxcr1 signaling is also required for coronary endothelial cell proliferation during cardiac regeneration. CONCLUSIONS These data show that after cardiac injury, coronary endothelial cells upregulate vegfc to promote coronary network reestablishment and cardiac regeneration. Mechanistically, Vegfc signaling upregulates epicardial emilin2a and cxcl8a expression to promote cardiac regeneration. These studies aid in understanding the mechanisms underlying coronary revascularization in zebrafish, with potential therapeutic implications to enhance revascularization and regeneration in injured human hearts.
Collapse
Affiliation(s)
- Hadil El-Sammak
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute, Frankfurt, Germany
| | - Bingyuan Yang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Stefan Guenther
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute, Frankfurt, Germany
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
| | - Wenbiao Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Rubén Marín-Juez
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Current address: Centre Hospitalier Universitaire Sainte-Justine Research Center, 3175 Chemin de la Côte-Sainte-Catherine, H3T 1C5 Montréal, QC, Canada, Department of Pathology and Cell Biology, University of Montreal, Montréal, QC H3T 1J4, Canada
| | - Didier Y.R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute, Frankfurt, Germany
| |
Collapse
|
37
|
Dupont L, Joannes L, Morfoisse F, Blacher S, Monseur C, Deroanne CF, Noël A, Colige AC. ADAMTS2 and ADAMTS14 substitute ADAMTS3 in adults for proVEGFC activation and lymphatic homeostasis. JCI Insight 2022; 7:151509. [PMID: 35316211 PMCID: PMC9089798 DOI: 10.1172/jci.insight.151509] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 03/16/2022] [Indexed: 11/17/2022] Open
Abstract
The capacity of ADAMTS3 to cleave pro-VEGFC into active VEGFC able to bind its receptors and to stimulate lymphangiogenesis has been clearly established during embryonic life. However, this function of ADAMTS3 is unlikely to persist in adulthood because of its restricted expression pattern after birth. Because ADAMTS2 and ADAMTS14 are closely related to ADAMTS3 and are mainly expressed in connective tissues where the lymphatic network extends, we hypothesized that they could substitute for ADAMTS3 during adulthood in mammals allowing proteolytic activation of pro-VEGFC. Here, we demonstrated that ADAMTS2 and ADAMTS14 are able to process pro-VEGFC into active VEGFC as efficiently as ADAMTS3. In vivo, adult mice lacking Adamts2 developed skin lymphedema due to a reduction of the density and diameter of lymphatic vessels, leading to a decrease of lymphatic functionality, while genetic ablation of Adamts14 had no impact. In a model of thermal cauterization of cornea, lymphangiogenesis was significantly reduced in Adamts2- and Adamts14-KO mice and further repressed in Adamts2/Adamts14 double-KO mice. In summary, we have demonstrated that ADAMTS2 and ADAMTS14 are as efficient as ADAMTS3 in activation of pro-VEGFC and are involved in the homeostasis of the lymphatic vasculature in adulthood, both in physiological and pathological processes.
Collapse
Affiliation(s)
- Laura Dupont
- Laboratory of Tumor and Developmental Biology, University of Liege, Liège, Belgium
| | - Loïc Joannes
- Laboratory of Connective Tissues Biology, University of Liege, Liège, Belgium
| | - Florent Morfoisse
- Laboratory of Tumor and Developmental Biology, University of Liege, Liège, Belgium
| | - Silvia Blacher
- Laboratory of Tumor and Development Biology, University of Liege, Liège, Belgium
| | - Christine Monseur
- Laboratory of Connective Tissues Biology, University of Liege, Liège, Belgium
| | | | - Agnès Noël
- Laboratory of Tumor and Development Biology, University of Liege, Liège, Belgium
| | - Alain Cma Colige
- Laboratory of Connective Tissues Biology, University of Liege, Liège, Belgium
| |
Collapse
|
38
|
Kolarzyk AM, Wong G, Lee E. Lymphatic Tissue and Organ Engineering for In Vitro Modeling and In Vivo Regeneration. Cold Spring Harb Perspect Med 2022; 12:a041169. [PMID: 35288402 PMCID: PMC9435571 DOI: 10.1101/cshperspect.a041169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The lymphatic system has an important role in maintaining fluid homeostasis and transporting immune cells and biomolecules, such as dietary fat, metabolic products, and antigens in different organs and tissues. Therefore, impaired lymphatic vessel function and/or lymphatic vessel deficiency can lead to numerous human diseases. The discovery of lymphatic endothelial markers and prolymphangiogenic growth factors, along with a growing number of in vitro and in vivo models and technologies has expedited research in lymphatic tissue and organ engineering, advancing therapeutic strategies. In this article, we describe lymphatic tissue and organ engineering in two- and three-dimensional culture systems and recently developed microfluidics and organ-on-a-chip systems in vitro. Next, we discuss advances in lymphatic tissue and organ engineering in vivo, focusing on biomaterial and scaffold engineering and their applications for lymphatic vessels and lymphoid organ regeneration. Last, we provide expert perspective and prospects in the field of lymphatic tissue engineering.
Collapse
Affiliation(s)
- Anna M Kolarzyk
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biomedical and Biological Sciences PhD Program, Ithaca, New York 14853, USA
| | - Gigi Wong
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biological Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biomedical and Biological Sciences PhD Program, Ithaca, New York 14853, USA
| |
Collapse
|
39
|
Eldrid C, Zloh M, Fotinou C, Yelland T, Yu L, Mota F, Selwood DL, Djordjevic S. VEGFA, B, C: Implications of the C-Terminal Sequence Variations for the Interaction with Neuropilins. Biomolecules 2022; 12:biom12030372. [PMID: 35327564 PMCID: PMC8945599 DOI: 10.3390/biom12030372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 11/23/2022] Open
Abstract
Vascular endothelial growth factors (VEGFs) are the key regulators of blood and lymphatic vessels’ formation and function. Each of the proteins from the homologous family VEGFA, VEGFB, VEGFC and VEGFD employs a core cysteine-knot structural domain for the specific interaction with one or more of the cognate tyrosine kinase receptors. Additional diversity is exhibited by the involvement of neuropilins–transmembrane co-receptors, whose b1 domain contains the binding site for the C-terminal sequence of VEGFs. Although all relevant isoforms of VEGFs that interact with neuropilins contain the required C-terminal Arg residue, there is selectivity of neuropilins and VEGF receptors for the VEGF proteins, which is reflected in the physiological roles that they mediate. To decipher the contribution made by the C-terminal sequences of the individual VEGF proteins to that functional differentiation, we determined structures of molecular complexes of neuropilins and VEGF-derived peptides and examined binding interactions for all neuropilin-VEGF pairs experimentally and computationally. While X-ray crystal structures and ligand-binding experiments highlighted similarities between the ligands, the molecular dynamics simulations uncovered conformational preferences of VEGF-derived peptides beyond the C-terminal arginine that contribute to the ligand selectivity of neuropilins. The implications for the design of the selective antagonists of neuropilins’ functions are discussed.
Collapse
Affiliation(s)
- Charles Eldrid
- Structural and Molecular Biology, ISMB, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK; (C.E.); (C.F.); (T.Y.); (L.Y.)
| | - Mire Zloh
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK;
- Faculty of Pharmacy, University Business Academy, 2100 Novi Sad, Serbia
| | - Constantina Fotinou
- Structural and Molecular Biology, ISMB, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK; (C.E.); (C.F.); (T.Y.); (L.Y.)
| | - Tamas Yelland
- Structural and Molecular Biology, ISMB, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK; (C.E.); (C.F.); (T.Y.); (L.Y.)
| | - Lefan Yu
- Structural and Molecular Biology, ISMB, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK; (C.E.); (C.F.); (T.Y.); (L.Y.)
| | - Filipa Mota
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK; (F.M.); (D.L.S.)
| | - David L. Selwood
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK; (F.M.); (D.L.S.)
| | - Snezana Djordjevic
- Structural and Molecular Biology, ISMB, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK; (C.E.); (C.F.); (T.Y.); (L.Y.)
- Correspondence: ; Tel.: +44-(0)20-7679-2230
| |
Collapse
|
40
|
Hashiguchi S, Tanaka T, Mano R, Kondo S, Kodama S. CCN2-induced lymphangiogenesis is mediated by the integrin αvβ5-ERK pathway and regulated by DUSP6. Sci Rep 2022; 12:926. [PMID: 35042954 PMCID: PMC8766563 DOI: 10.1038/s41598-022-04988-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 01/05/2022] [Indexed: 12/20/2022] Open
Abstract
Lymphangiogenesis is essential for the development of the lymphatic system and is important for physiological processes such as homeostasis, metabolism and immunity. Cellular communication network factor 2 (CCN2, also known as CTGF), is a modular and matricellular protein and a well-known angiogenic factor in physiological and pathological angiogenesis. However, its roles in lymphangiogenesis and intracellular signaling in lymphatic endothelial cells (LECs) remain unclear. Here, we investigated the effects of CCN2 on lymphangiogenesis. In in vivo Matrigel plug assays, exogenous CCN2 increased the number of Podoplanin-positive vessels. Subsequently, we found that CCN2 induced phosphorylation of ERK in primary cultured LECs, which was almost completely inhibited by the blockade of integrin αvβ5 and partially decreased by the blockade of integrin αvβ3. CCN2 promoted direct binding of ERK to dual-specific phosphatase 6 (DUSP6), which regulated the activation of excess ERK by dephosphorylating ERK. In vitro, CCN2 promoted tube formation in LECs, while suppression of Dusp6 further increased tube formation. In vivo, immunohistochemistry also detected ERK phosphorylation and DUSP6 expression in Podoplanin-positive cells on CCN2-supplemented Matrigel. These results indicated that CCN2 promotes lymphangiogenesis by enhancing integrin αvβ5-mediated phosphorylation of ERK and demonstrated that DUSP6 is a negative regulator of excessive lymphangiogenesis by CCN2.
Collapse
Affiliation(s)
- Shiho Hashiguchi
- Department of Oral Surgery, Faculty of Medicine, Fukuoka University, Fukuoka, Japan.,Department of Regenerative Medicine and Transplantation, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Tomoko Tanaka
- Department of Regenerative Medicine and Transplantation, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Ryosuke Mano
- Department of Oral Surgery, Faculty of Medicine, Fukuoka University, Fukuoka, Japan.,Department of Regenerative Medicine and Transplantation, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Seiji Kondo
- Department of Oral Surgery, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Shohta Kodama
- Department of Regenerative Medicine and Transplantation, Faculty of Medicine, Fukuoka University, Fukuoka, Japan.
| |
Collapse
|
41
|
Schiavo RK, Tamplin OJ. Vascular endothelial growth factor c regulates hematopoietic stem cell fate in the dorsal aorta. Development 2022; 149:dev199498. [PMID: 34919128 PMCID: PMC8917412 DOI: 10.1242/dev.199498] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 12/06/2021] [Indexed: 01/21/2023]
Abstract
Hematopoietic stem and progenitor cells (HSPCs) are multipotent cells that self-renew or differentiate to establish the entire blood hierarchy. HSPCs arise from the hemogenic endothelium of the dorsal aorta (DA) during development in a process called endothelial-to-hematopoietic transition. The factors and signals that control HSPC fate decisions from the hemogenic endothelium are not fully understood. We found that Vegfc has a role in HSPC emergence from the zebrafish DA. Using time-lapse live imaging, we show that some HSPCs in the DA of vegfc loss-of-function embryos display altered cellular behavior. Instead of typical budding from the DA, emergent HSPCs exhibit crawling behavior similar to myeloid cells. This was confirmed by increased myeloid cell marker expression in the ventral wall of the DA and the caudal hematopoietic tissue. This increase in myeloid cells corresponded with a decrease in HSPCs that persisted into larval stages. Together, our data suggest that Vegfc regulates HSPC emergence in the hemogenic endothelium, in part by suppressing a myeloid cell fate. Our study provides a potential signal for modulation of HSPC fate in stem cell differentiation protocols.
Collapse
|
42
|
Abstract
Adipose tissue, once thought to be an inert receptacle for energy storage, is now recognized as a complex tissue with multiple resident cell populations that actively collaborate in response to diverse local and systemic metabolic, thermal, and inflammatory signals. A key participant in adipose tissue homeostasis that has only recently captured broad scientific attention is the lymphatic vasculature. The lymphatic system's role in lipid trafficking and mediating inflammation makes it a natural partner in regulating adipose tissue, and evidence supporting a bidirectional relationship between lymphatics and adipose tissue has accumulated in recent years. Obesity is now understood to impair lymphatic function, whereas altered lymphatic function results in aberrant adipose tissue deposition, though the molecular mechanisms governing these phenomena have yet to be fully elucidated. We will review our current understanding of the relationship between adipose tissue and the lymphatic system here, focusing on known mechanisms of lymphatic-adipose crosstalk.
Collapse
Affiliation(s)
- Gregory P Westcott
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Joslin Diabetes Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA
| | - Evan D Rosen
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA
- Broad Institute, Cambridge, MA 02142, USA
- Correspondence: Evan D. Rosen, MD, PhD, Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, USA.
| |
Collapse
|
43
|
Oliver G. Lymphatic endothelial cell fate specification in the mammalian embryo: An historical perspective. Dev Biol 2021; 482:44-54. [PMID: 34915023 DOI: 10.1016/j.ydbio.2021.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023]
Abstract
Development of the mammalian lymphatic vasculature is a stepwise process requiring the specification of lymphatic endothelial cell progenitors in the embryonic veins, and their subsequent budding to give rise to most of the mature lymphatic vasculature. In mice, formation of the lymphatic vascular network starts inside the cardinal vein at around E9.5 when a subpopulation of venous endothelial cells gets committed into the lymphatic lineage by their acquisition of Prox1 expression. Identification of critical genes regulating lymphatic development facilitated the detailed cellular and molecular characterization of some of the cellular and molecular mechanisms regulating the early steps leading to the formation of the mammalian lymphatic vasculature. A better understanding of basic aspects of early lymphatic development, and the availability of novel tools and animal models has been instrumental in the identification of important novel functional roles of this vasculature network.
Collapse
Affiliation(s)
- Guillermo Oliver
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
44
|
Buso G, Favre L, Maufus M, Honorati M, Lessert C, Depairon M, Raffoul W, Tomson D, Mazzolai L. Indocyanine green lymphography as novel tool to assess lymphatics in patients with lipedema. Microvasc Res 2021; 140:104298. [PMID: 34896377 DOI: 10.1016/j.mvr.2021.104298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/11/2021] [Accepted: 12/02/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Lipedema is a chronic and progressive disease associated with lymphatic impairment at later stages. The aim of our study was to describe the functional status and anatomy of lower limb superficial lymphatic system using indocyanine green (ICG) lymphography in patients with lipedema. METHODS Following ICG injection at the dorsum of the foot, distance (cm) covered by the dye at 10 (T10') and 25 min (T25') was measured and normalized for limb length. If the dye did not reach the groin within 25 min, patients were classified as "drainage-needing" group (DNG). Values of fat and lean distribution assessed by dual-energy X-ray absorptiometry were extracted, and correlation analysis was performed. Furthermore, anatomical patterns of superficial lymphatics were assessed. RESULTS Overall, 45 women were included, 25 (56%) of whom were classified as DNG. Symptoms duration was significantly associated with DNG status at multivariate analysis (odds ratio 1.07; 95% CI 1.01-1.14; p = 0.047). Moreover, Spearman's analysis showed a negative correlation between symptoms duration and T25' dye migration (r = -0.469; p = 0.037). Overall, no major anatomical lymphatic changes were found. CONCLUSIONS Present study suggests that lymphatic functioning in patients with lipedema correlates with symptoms duration. Further research on larger cohorts should verify our findings and clarify their potential therapeutic implications. Overall, ICG lymphography may be promising technique to assess both lymphatic anatomy and functioning in patients with lipedema.
Collapse
Affiliation(s)
- Giacomo Buso
- Angiology Division, Heart and Vessel Department, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Lucie Favre
- Endocrinology, Diabetology and Metabolism Division, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Mario Maufus
- Angiology Division, Heart and Vessel Department, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Marcella Honorati
- Angiology Division, Heart and Vessel Department, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Claudia Lessert
- Angiology Division, Heart and Vessel Department, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Michèle Depairon
- Angiology Division, Heart and Vessel Department, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Wassim Raffoul
- Plastic surgery Division, Locomotor system Department, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Didier Tomson
- Angiology Division, Heart and Vessel Department, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Lucia Mazzolai
- Angiology Division, Heart and Vessel Department, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
45
|
Liu H, Hiremath C, Patterson Q, Vora S, Shang Z, Jamieson AR, Fiolka R, Dean KM, Dellinger MT, Marciano DK. Heterozygous Mutation of Vegfr3 Reduces Renal Lymphatics without Renal Dysfunction. J Am Soc Nephrol 2021; 32:3099-3113. [PMID: 34551997 PMCID: PMC8638391 DOI: 10.1681/asn.2021010061] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 08/29/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Lymphatic abnormalities are observed in several types of kidney disease, but the relationship between the renal lymphatic system and renal function is unclear. The discovery of lymphatic-specific proteins, advances in microscopy, and available genetic mouse models provide the tools to help elucidate the role of renal lymphatics in physiology and disease. METHODS We utilized a mouse model containing a missense mutation in Vegfr3 (dubbed Chy ) that abrogates its kinase ability. Vegfr3 Chy/+ mice were examined for developmental abnormalities and kidney-specific outcomes. Control and Vegfr3 Chy/+ mice were subjected to cisplatin-mediated injury. We characterized renal lymphatics using tissue-clearing, light-sheet microscopy, and computational analyses. RESULTS In the kidney, VEGFR3 is expressed not only in lymphatic vessels but also, in various blood capillaries. Vegfr3 Chy/+ mice had severely reduced renal lymphatics with 100% penetrance, but we found no abnormalities in BP, serum creatinine, BUN, albuminuria, and histology. There was no difference in the degree of renal injury after low-dose cisplatin (5 mg/kg), although Vegfr3 Chy/+ mice developed perivascular inflammation. Cisplatin-treated controls had no difference in total cortical lymphatic volume and length but showed increased lymphatic density due to decreased cortical volume. CONCLUSIONS We demonstrate that VEGFR3 is required for development of renal lymphatics. Our studies reveal that reduced lymphatic density does not impair renal function at baseline and induces only modest histologic changes after mild injury. We introduce a novel quantification method to evaluate renal lymphatics in 3D and demonstrate that accurate measurement of lymphatic density in CKD requires assessment of changes to cortical volume.
Collapse
Affiliation(s)
- Hao Liu
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chitkale Hiremath
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Quinten Patterson
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Saumya Vora
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Zhiguo Shang
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Andrew R. Jamieson
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Reto Fiolka
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas,Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kevin M. Dean
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Michael T. Dellinger
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Denise K. Marciano
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
46
|
Isolating and characterizing lymphatic endothelial progenitor cells for potential therapeutic lymphangiogenic applications. Acta Biomater 2021; 135:191-202. [PMID: 34384911 DOI: 10.1016/j.actbio.2021.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 12/14/2022]
Abstract
Lymphatic dysfunction is associated with the progression of several vascular disorders, though currently, there are limited strategies to promote new lymphatic vasculature (i.e., lymphangiogenesis) to restore lost lymphatic function. One promising approach to stimulate lymphangiogenesis involves delivering endothelial progenitor cells (EPCs), which are naturally involved in de novo blood vessel formation and have recently been identified to include a lymphatic subpopulation. However, the contribution of lymphatic EPCs in lymphangiogenesis is not clear and challenges with maintaining the activity of transplanted EPCs remain. Thus, the objective of this study was to isolate lymphatic EPCs from human umbilical cord blood and characterize their role in the initial stages of blood or lymphatic vasculature formation. Furthermore, this study also tested the applicability of alginate hydrogels to deliver lymphatic EPCs for a possible therapeutic application. We postulated and confirmed that blood and lymphatic EPC colonies could be isolated from human umbilical cord blood. Additionally, EPC populations responded to either angiogenic or lymphangiogenic growth factors and could stimulate their respective mature endothelial cells in vasculature models in vitro. Finally, lymphatic EPCs maintained their ability to promote lymphatic sprouts after prolonged interactions with the alginate hydrogel microenvironment. These results suggest EPCs have both a blood and a lymphatic population that have specific roles in promoting revascularization and highlight the potential of alginate hydrogels for the delivery of lymphatic EPCs. STATEMENT OF SIGNIFICANCE: Despite the potential therapeutic benefit of promoting lymphatic vasculature, lymphangiogenesis remains understudied. One appealing strategy for promoting lymphangiogenesis involves delivering lymphatic endothelial progenitor cells (EPCs), which are a subpopulation of EPCs involved in de novo vessel formation. Here, we investigate the role of isolated blood and lymphatic EPC subpopulations in promoting the early stages of vascularization and the utility of alginate hydrogels to deliver lymphatic EPCs. We determined that EPCs had two populations that expressed either blood or lymphatic markers, could stimulate their respective mature vasculature in tissue constructs and that alginate hydrogels maintained the therapeutic potential of lymphatic EPCs. We anticipate this work could support promising biomaterial applications of EPCs to promote revascularization, which could have many therapeutic applications.
Collapse
|
47
|
Lee HK, Lee SM, Lee DI. Corneal Lymphangiogenesis: Current Pathophysiological Understandings and Its Functional Role in Ocular Surface Disease. Int J Mol Sci 2021; 22:ijms222111628. [PMID: 34769057 PMCID: PMC8583961 DOI: 10.3390/ijms222111628] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/19/2021] [Accepted: 10/23/2021] [Indexed: 12/23/2022] Open
Abstract
The cornea is a transparent and avascular tissue that plays a central role in light refraction and provides a physical barrier to the external environment. Corneal avascularity is a unique histological feature that distinguishes it from the other parts of the body. Functionally, corneal immune privilege critically relies on corneal avascularity. Corneal lymphangiogenesis is now recognized as a general pathological feature in many pathologies, including dry eye disease (DED), corneal allograft rejection, ocular allergy, bacterial and viral keratitis, and transient corneal edema. Currently, sizable data from clinical and basic research have accumulated on the pathogenesis and functional role of ocular lymphangiogenesis. However, because of the invisibility of lymphatic vessels, ocular lymphangiogenesis has not been studied as much as hemangiogenesis. We reviewed the basic mechanisms of lymphangiogenesis and summarized recent advances in the pathogenesis of ocular lymphangiogenesis, focusing on corneal allograft rejection and DED. In addition, we discuss future directions for lymphangiogenesis research.
Collapse
Affiliation(s)
- Hyung-Keun Lee
- Department of Ophthalmology, Institute of Vision Research, Yonsei University College of Medicine, Seoul 06273, Korea
- Correspondence: ; Tel.: +82-2-2019-3444
| | - Sang-Mok Lee
- Department of Ophthalmology, HanGil Eye Hospital, Catholic Kwandong University College of Medicine, Incheon 21388, Korea;
| | - Dong-Ihll Lee
- Medical School, Capital Medical University, Beijing 100069, China;
| |
Collapse
|
48
|
Pankova MN, Lobov GI. Lymphangiogenesis and Features of Lymphatic Drainage in Different Organs: the Significance for Allograft Fate. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021050100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
49
|
Lugo-Cintrón KM, Ayuso JM, Humayun M, Gong MM, Kerr SC, Ponik SM, Harari PM, Virumbrales-Muñoz M, Beebe DJ. Primary head and neck tumour-derived fibroblasts promote lymphangiogenesis in a lymphatic organotypic co-culture model. EBioMedicine 2021; 73:103634. [PMID: 34673450 PMCID: PMC8528684 DOI: 10.1016/j.ebiom.2021.103634] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND In head and neck cancer, intratumour lymphatic density and tumour lymphangiogenesis have been correlated with lymphatic metastasis, making lymphangiogenesis a promising therapeutic target. However, inter-patient tumour heterogeneity makes it challenging to predict tumour progression and lymph node metastasis. Understanding the lymphangiogenic-promoting factors leading to metastasis (e.g., tumour-derived fibroblasts or TDF), would help develop strategies to improve patient outcomes. METHODS A microfluidic in vitro model of a tubular lymphatic vessel was co-cultured with primary TDF from head and neck cancer patients to evaluate the effect of TDF on lymphangiogenesis. We assessed the length and number of lymphangiogenic sprouts and vessel permeability via microscopy and image analysis. Finally, we characterised lymphatic vessel conditioning by TDF via RT-qPCR. FINDINGS Lymphatic vessels were conditioned by the TDF in a patient-specific manner. Specifically, the presence of TDF induced sprouting, altered vessel permeability, and increased the expression of pro-lymphangiogenic genes. Gene expression and functional responses in the fibroblast-conditioned lymphatic vessels were consistent with the patient tumour stage and lymph node status. IGF-1, upregulated among patients, was targeted to validate our personalised medicine approach. Interestingly, IGF-1 blockade was not effective across different patients. INTERPRETATION The use of lymphatic organotypic models incorporating head and neck TDF provides insight into the pathways leading to lymphangiogenesis in each patient. This model provided a platform to test anti-angiogenic therapeutics and inform of their effectiveness for individual patients. FUNDING NIH R33CA225281. Wisconsin Head and Neck SPORE NIH P50DE026787. NIH R01AI34749.
Collapse
Affiliation(s)
- Karina M Lugo-Cintrón
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA; Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - José M Ayuso
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Mouhita Humayun
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA; Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Max M Gong
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Biomedical Engineering, Trine University, Angola, IN, USA
| | - Sheena C Kerr
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Suzanne M Ponik
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Paul M Harari
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - María Virumbrales-Muñoz
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA; Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
50
|
Black LM, Winfree S, Khochare SD, Kamocka MM, Traylor AM, Esman SK, Khan S, Zarjou A, Agarwal A, El-Achkar TM. Quantitative 3-dimensional imaging and tissue cytometry reveals lymphatic expansion in acute kidney injury. J Transl Med 2021; 101:1186-1196. [PMID: 34017058 PMCID: PMC8373805 DOI: 10.1038/s41374-021-00609-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/16/2021] [Accepted: 04/16/2021] [Indexed: 12/27/2022] Open
Abstract
The lymphatic system plays an integral role in physiology and has recently been identified as a key player in disease progression. Tissue injury stimulates lymphatic expansion, or lymphangiogenesis (LA), though its precise role in disease processes remains unclear. LA is associated with inflammation, which is a key component of acute kidney injury (AKI), for which there are no approved therapies. While LA research has gained traction in the last decade, there exists a significant lack of understanding of this process in the kidney. Though innovative studies have elucidated markers and models with which to study LA, the field is still evolving with ways to visualize lymphatics in vivo. Prospero-related homeobox-1 (Prox-1) is the master regulator of LA and determines lymphatic cell fate through its action on vascular endothelial growth factor receptor expression. Here, we investigate the consequences of AKI on the abundance and distribution of lymphatic endothelial cells using Prox1-tdTomato reporter mice (ProxTom) coupled with large-scale three-dimensional quantitative imaging and tissue cytometry (3DTC). Using these technologies, we describe the spatial dynamics of lymphatic vasculature in quiescence and post-AKI. We also describe the use of lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1) as a marker of lymphatic vessels using 3DTC in the absence of the ProxTom reporter mice as an alternative approach. The use of 3DTC for lymphatic research presents a new avenue with which to study the origin and distribution of renal lymphatic vessels. These findings will enhance our understanding of renal lymphatic function during injury and could inform the development of novel therapeutics for intervention in AKI.
Collapse
Affiliation(s)
- Laurence M Black
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Seth Winfree
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Biological Microscopy, Indianapolis, IN, USA
| | - Suraj D Khochare
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Biological Microscopy, Indianapolis, IN, USA
| | - Malgorzata M Kamocka
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Biological Microscopy, Indianapolis, IN, USA
| | - Amie M Traylor
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stephanie K Esman
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shehnaz Khan
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Biological Microscopy, Indianapolis, IN, USA
| | - Abolfazl Zarjou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Veterans Affairs, Birmingham, AL, USA.
| | - Tarek M El-Achkar
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Center for Biological Microscopy, Indianapolis, IN, USA.
- Indianapolis Veterans Affairs Medical Center, Indianapolis, IN, USA.
| |
Collapse
|