1
|
Zhou Z, Chen W, Cao Y, Abdi R, Tao W. Nanomedicine-based strategies for the treatment of vein graft disease. Nat Rev Cardiol 2025; 22:255-272. [PMID: 39501093 PMCID: PMC11925677 DOI: 10.1038/s41569-024-01094-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: 09/27/2024] [Indexed: 01/03/2025]
Abstract
Autologous saphenous veins are the most frequently used conduits for coronary and peripheral artery bypass grafting. However, vein graft failure rates of 40-50% within 10 years of the implantation lead to poor long-term outcomes after bypass surgery. Currently, only a few therapeutic approaches for vein graft disease have been successfully translated into clinical practice. Building on the past two decades of advanced understanding of vein graft biology and the pathophysiological mechanisms underlying vein graft disease, nanomedicine-based strategies offer promising opportunities to address this important unmet clinical need. In this Review, we provide deep insight into the latest developments in the rational design and applications of nanoparticles that have the potential to target specific cells during various pathophysiological stages of vein graft disease, including early endothelial dysfunction, intermediate intimal hyperplasia and late-stage accelerated atherosclerosis. Additionally, we underscore the convergence of nanofabricated biomaterials, with a particular focus on hydrogels, external graft support devices and cell-based therapies, alongside bypass surgery to improve local delivery efficiency and therapeutic efficacy. Finally, we provide a specific discussion on the considerations, challenges and novel perspectives for the future clinical translation of nanomedicine for the treatment of vein graft disease.
Collapse
Affiliation(s)
- Zhuoming Zhou
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Chen
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Yihai Cao
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Reza Abdi
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wei Tao
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Iwata A, Chelvanambi S, Asano T, Whelan M, Nakamura Y, Aikawa E, Sasaki Y, Aikawa M. Gene expression profiles of precursor cells identify compounds that reduce NRP1 surface expression in macrophages: Implication for drug repositioning for COVID-19. Front Cardiovasc Med 2024; 11:1438396. [PMID: 39512370 PMCID: PMC11541348 DOI: 10.3389/fcvm.2024.1438396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 09/23/2024] [Indexed: 11/15/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) is transitioning from a pandemic to an endemic phase through recurring mutations. Initial efforts focused on developing strategies to mitigate infection of lung epithelial cells which are the primary targets of the SARS-CoV-2 virus using the affinity of the spike protein to human ACE2 receptor. SARS-CoV-2, however, infects additional cell types present in the lung such as macrophages through the alternate entry receptor Neuropilin 1 (NRP1). Developing novel therapeutic strategies to prevent SARS-CoV-2 infection of cells crucial for immunosurveillance could thus be integral to treat post-acute sequelae of COVID-19 (PASC). Since traditional drug development process takes a long time, it is imperative to establish new strategies that can be rapidly deployed to combat the dynamic nature of COVID-19 evolution and to contribute to prevention of future pandemics. We obtained the gene expression profiles of THP-1 monocytes from L1000-based Connectivity Map using CLUE, cloud- based software platform for the analysis of perturbational datasets to identify compounds that could reduce the expression level of NRP1. Out of 33,590 compounds, we analyzed the profiles of 45 compounds for their ability to reduce NRP1 expression. We selected the top five small molecule inhibitors predicted to decrease the expression of NRP1 for validation studies. All five selected compounds showed low cytotoxicity at tested doses and their ability to reduce NRP1 surface expression was evaluated in THP-1 monocytes, THP-1-derived macrophage like cells and human peripheral blood mononuclear cell (PBMC)-derived primary macrophages. Five compounds with the largest predicted reduction of NRP1 expression decreased macrophage NRP1 surface expression measured using flow cytometry and fluorescent microscopy assays in both cell line and primary macrophages. Using our computational approach, we identified 45 compounds that could potentially decrease NRP1 surface expression in macrophages based on their effect on THP-1 monocytes. Validation studies showed that such an approach can help to identify compounds for drug repositioning in target cells that are absent in the L1000 database. Our proposed approach can be applicable for the rapid compound exploration to combat novel cell types that SARS-CoV-2 targets for infection and could provide molecular bases for the development of new drugs.
Collapse
Affiliation(s)
- Akira Iwata
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Sarvesh Chelvanambi
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Takaharu Asano
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Mary Whelan
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Yuto Nakamura
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Yusuke Sasaki
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
3
|
Yuwen Y, Wang X, Liu J, Liu Z, Zhu H. Delta- like ligand 4- expressing macrophages and human diseases: Insights into pathophysiology and therapeutic opportunities. Heliyon 2023; 9:e20777. [PMID: 37842562 PMCID: PMC10569996 DOI: 10.1016/j.heliyon.2023.e20777] [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: 05/28/2023] [Revised: 08/20/2023] [Accepted: 10/06/2023] [Indexed: 10/17/2023] Open
Abstract
Macrophages are key players in the immune response and have been implicated in various human diseases, including atherosclerosis, cancer, and chronic inflammatory disorders. While numerous studies have delved into the nuances of macrophage behavior in these conditions, there remains a gap in understanding the specific role of Delta-like ligand 4 (Dll4)-expressing macrophages and their overarching implications across these diseases. Among the plethora of factors expressed by macrophages, Dll4 has emerged as a molecule of particular interest. Recent studies have highlighted its unique role in modulating macrophage functions and its potential implications in various diseases. This review seeks to consolidate existing knowledge, address this gap, and present a comprehensive overview of Dll4-expressing macrophages in the context of these disorders and highlight their potential as therapeutic targets. We examined the involvement of Dll4-expressing macrophages in multiple human diseases such as atherosclerosis, cancer and chronic inflammatory diseases, emphasizing their influence on disease progression. We also discussed the challenges, limitations, and emerging research areas in targeting Dll4-expressing macrophages and provide an outlook on potential therapeutic strategies for the treatment of these diseases. By addressing the previously existing research gap, we've provided a roadmap that brings together fragmented insights, paving the way for more holistic research and potentially more effective therapeutic strategies centered on Dll4-expressing macrophages.
Collapse
Affiliation(s)
- Ya Yuwen
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, China
- Medical School, Xizang Minzu University, Xianyang, China
- Integrative Chinese and Western Medicine Key Laboratory of Atherosclerosis, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, China
| | - Xiqiang Wang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, China
- Integrative Chinese and Western Medicine Key Laboratory of Atherosclerosis, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, China
| | - Jing Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, China
- Integrative Chinese and Western Medicine Key Laboratory of Atherosclerosis, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, China
| | - Zhongwei Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, China
- Integrative Chinese and Western Medicine Key Laboratory of Atherosclerosis, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, China
| | - Haitao Zhu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, China
- Department of Pediatrics (No. 3 Ward), Northwest Women's and Children's Hospital, Xi'an, China
| |
Collapse
|
4
|
Franco-Acevedo A, Comes J, Mack JJ, Valenzuela NM. New insights into maladaptive vascular responses to donor specific HLA antibodies in organ transplantation. FRONTIERS IN TRANSPLANTATION 2023; 2:1146040. [PMID: 38993843 PMCID: PMC11235244 DOI: 10.3389/frtra.2023.1146040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/03/2023] [Indexed: 07/13/2024]
Abstract
Transplant vasculopathy (TV) causes thickening of donor blood vessels in transplanted organs, and is a significant cause of graft loss and mortality in allograft recipients. It is known that patients with repeated acute rejection and/or donor specific antibodies are predisposed to TV. Nevertheless, the exact molecular mechanisms by which alloimmune injury culminates in this disease have not been fully delineated. As a result of this incomplete knowledge, there is currently a lack of effective therapies for this disease. The immediate intracellular signaling and the acute effects elicited by anti-donor HLA antibodies are well-described and continuing to be revealed in deeper detail. Further, advances in rejection diagnostics, including intragraft gene expression, provide clues to the inflammatory changes within allografts. However, mechanisms linking these events with long-term outcomes, particularly the maladaptive vascular remodeling seen in transplant vasculopathy, are still being delineated. New evidence demonstrates alterations in non-coding RNA profiles and the occurrence of endothelial to mesenchymal transition (EndMT) during acute antibody-mediated graft injury. EndMT is also readily apparent in numerous settings of non-transplant intimal hyperplasia, and lessons can be learned from advances in those fields. This review will provide an update on these recent developments and remaining questions in our understanding of HLA antibody-induced vascular damage, framed within a broader consideration of manifestations and implications across transplanted organ types.
Collapse
Affiliation(s)
- Adriana Franco-Acevedo
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, United States
| | - Johanna Comes
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Julia J Mack
- Department of Medicine, Division of Cardiology, University of California, Los Angeles, CA, United States
| | - Nicole M Valenzuela
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, United States
| |
Collapse
|
5
|
Da Silva Sanchez AJ, Zhao K, Huayamares SG, Hatit MZC, Lokugamage MP, Loughrey D, Dobrowolski C, Wang S, Kim H, Paunovska K, Kuzminich Y, Dahlman JE. Substituting racemic ionizable lipids with stereopure ionizable lipids can increase mRNA delivery. J Control Release 2023; 353:270-277. [PMID: 36423872 PMCID: PMC9897220 DOI: 10.1016/j.jconrel.2022.11.037] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 12/03/2022]
Abstract
Lipid nanoparticles (LNPs) have delivered siRNA and mRNA drugs in humans, underscoring the potential impact of improving the therapeutic window of next-generation LNPs. To increase the LNP therapeutic window, we applied lessons from small-molecule chemistry to ionizable lipid design. Specifically, given that stereochemistry often influences small-molecule safety and pharmacokinetics, we hypothesized that the stereochemistry of lipids within an LNP would influence mRNA delivery. We tested this hypothesis in vivo using 128 novel LNPs that included stereopure derivatives of C12-200, an ionizable lipid that when formulated into LNPs delivers RNA in mice and non-human primates but is not used clinically due to its poor tolerability. We found that a novel C12-200-S LNP delivered up to 2.8-fold and 6.1-fold more mRNA in vivo than its racemic and C12-200-R controls, respectively. To identify the potential causes leading to increased delivery, we quantified LNP biophysical traits and concluded that these did not change with stereochemistry. Instead, we found that stereopure LNPs were better tolerated than racemic LNPs in vivo. These data suggest that LNP-mediated mRNA delivery can be improved by designing LNPs to include stereopure ionizable lipids.
Collapse
Affiliation(s)
- Alejandro J Da Silva Sanchez
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA; Department of Chemical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Kun Zhao
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Sebastian G Huayamares
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Marine Z C Hatit
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Melissa P Lokugamage
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - David Loughrey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Curtis Dobrowolski
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Shuaishuai Wang
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | - Hyejin Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Kalina Paunovska
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Yanina Kuzminich
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| |
Collapse
|
6
|
Katsuki S, K. Jha P, Lupieri A, Nakano T, Passos LS, Rogers MA, Becker-Greene D, Le TD, Decano JL, Ho Lee L, Guimaraes GC, Abdelhamid I, Halu A, Muscoloni A, V. Cannistraci C, Higashi H, Zhang H, Vromman A, Libby P, Keith Ozaki C, Sharma A, Singh SA, Aikawa E, Aikawa M. Proprotein Convertase Subtilisin/Kexin 9 (PCSK9) Promotes Macrophage Activation via LDL Receptor-Independent Mechanisms. Circ Res 2022; 131:873-889. [PMID: 36263780 PMCID: PMC9973449 DOI: 10.1161/circresaha.121.320056] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Activated macrophages contribute to the pathogenesis of vascular disease. Vein graft failure is a major clinical problem with limited therapeutic options. PCSK9 (proprotein convertase subtilisin/kexin 9) increases low-density lipoprotein (LDL)-cholesterol levels via LDL receptor (LDLR) degradation. The role of PCSK9 in macrophage activation and vein graft failure is largely unknown, especially through LDLR-independent mechanisms. This study aimed to explore a novel mechanism of macrophage activation and vein graft disease induced by circulating PCSK9 in an LDLR-independent fashion. METHODS We used Ldlr-/- mice to examine the LDLR-independent roles of circulating PCSK9 in experimental vein grafts. Adeno-associated virus (AAV) vector encoding a gain-of-function mutant of PCSK9 (rAAV8/D377Y-mPCSK9) induced hepatic PCSK9 overproduction. To explore novel inflammatory targets of PCSK9, we used systems biology in Ldlr-/- mouse macrophages. RESULTS In Ldlr-/- mice, AAV-PCSK9 increased circulating PCSK9, but did not change serum cholesterol and triglyceride levels. AAV-PCSK9 promoted vein graft lesion development when compared with control AAV. In vivo molecular imaging revealed that AAV-PCSK9 increased macrophage accumulation and matrix metalloproteinase activity associated with decreased fibrillar collagen, a molecular determinant of atherosclerotic plaque stability. AAV-PCSK9 induced mRNA expression of the pro-inflammatory mediators IL-1β (interleukin-1 beta), TNFα (tumor necrosis factor alpha), and MCP-1 (monocyte chemoattractant protein-1) in peritoneal macrophages underpinned by an in vitro analysis of Ldlr-/- mouse macrophages stimulated with endotoxin-free recombinant PCSK9. A combination of unbiased global transcriptomics and new network-based hyperedge entanglement prediction analysis identified the NF-κB (nuclear factor-kappa B) signaling molecules, lectin-like oxidized LOX-1 (LDL receptor-1), and SDC4 (syndecan-4) as potential PCSK9 targets mediating pro-inflammatory responses in macrophages. CONCLUSIONS Circulating PCSK9 induces macrophage activation and vein graft lesion development via LDLR-independent mechanisms. PCSK9 may be a potential target for pharmacologic treatment for this unmet medical need.
Collapse
Affiliation(s)
- Shunsuke Katsuki
- The Center for Excellence in Vascular Biology, Cardiovascular Division (S.K., P.K.J., A.L., T.N., L.S.A.P., D.B.-G., T.-D.L., G.C.G., A.V., P.L., E.A., M.A.)
| | - Prabhash K. Jha
- The Center for Excellence in Vascular Biology, Cardiovascular Division (S.K., P.K.J., A.L., T.N., L.S.A.P., D.B.-G., T.-D.L., G.C.G., A.V., P.L., E.A., M.A.)
| | - Adrien Lupieri
- The Center for Excellence in Vascular Biology, Cardiovascular Division (S.K., P.K.J., A.L., T.N., L.S.A.P., D.B.-G., T.-D.L., G.C.G., A.V., P.L., E.A., M.A.)
| | - Toshiaki Nakano
- The Center for Excellence in Vascular Biology, Cardiovascular Division (S.K., P.K.J., A.L., T.N., L.S.A.P., D.B.-G., T.-D.L., G.C.G., A.V., P.L., E.A., M.A.)
| | - Livia S.A. Passos
- The Center for Excellence in Vascular Biology, Cardiovascular Division (S.K., P.K.J., A.L., T.N., L.S.A.P., D.B.-G., T.-D.L., G.C.G., A.V., P.L., E.A., M.A.)
| | - Maximillian A. Rogers
- The Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (M.A.R., J.L.D., L.H.L., I.A., A.H., H.H., H.Z., A.S., S.A.S., E.A., M.A.)
| | - Dakota Becker-Greene
- The Center for Excellence in Vascular Biology, Cardiovascular Division (S.K., P.K.J., A.L., T.N., L.S.A.P., D.B.-G., T.-D.L., G.C.G., A.V., P.L., E.A., M.A.)
| | - Thanh-Dat Le
- The Center for Excellence in Vascular Biology, Cardiovascular Division (S.K., P.K.J., A.L., T.N., L.S.A.P., D.B.-G., T.-D.L., G.C.G., A.V., P.L., E.A., M.A.)
| | - Julius L. Decano
- The Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (M.A.R., J.L.D., L.H.L., I.A., A.H., H.H., H.Z., A.S., S.A.S., E.A., M.A.)
| | - Lang Ho Lee
- The Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (M.A.R., J.L.D., L.H.L., I.A., A.H., H.H., H.Z., A.S., S.A.S., E.A., M.A.)
| | - Gabriel C. Guimaraes
- The Center for Excellence in Vascular Biology, Cardiovascular Division (S.K., P.K.J., A.L., T.N., L.S.A.P., D.B.-G., T.-D.L., G.C.G., A.V., P.L., E.A., M.A.)
| | - Ilyes Abdelhamid
- The Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (M.A.R., J.L.D., L.H.L., I.A., A.H., H.H., H.Z., A.S., S.A.S., E.A., M.A.)
- Channing Division of Network Medicine (I.A., A.H., A.S., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Arda Halu
- The Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (M.A.R., J.L.D., L.H.L., I.A., A.H., H.H., H.Z., A.S., S.A.S., E.A., M.A.)
- Channing Division of Network Medicine (I.A., A.H., A.S., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Alessandro Muscoloni
- The Biomedical Cybernetics Group, Biotechnology Center, Center for Molecular and Cellular Bioengineering, Center for Systems Biology Dresden, Cluster of Excellence Physics of Life, Department of Physics, Technical University Dresden, Dresden, Germany (A.M., C.V.C)
- Center for Complex Network Intelligence at the Tsinghua Laboratory of Brain and Intelligence, Department of Bioengineering, Tsinghua University, Beijing, China (A.M., C.V.C.)
| | - Carlo V. Cannistraci
- The Center for Excellence in Vascular Biology, Cardiovascular Division (S.K., P.K.J., A.L., T.N., L.S.A.P., D.B.-G., T.-D.L., G.C.G., A.V., P.L., E.A., M.A.)
- Center for Complex Network Intelligence at the Tsinghua Laboratory of Brain and Intelligence, Department of Bioengineering, Tsinghua University, Beijing, China (A.M., C.V.C.)
| | - Hideyuki Higashi
- The Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (M.A.R., J.L.D., L.H.L., I.A., A.H., H.H., H.Z., A.S., S.A.S., E.A., M.A.)
| | - Hengmin Zhang
- The Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (M.A.R., J.L.D., L.H.L., I.A., A.H., H.H., H.Z., A.S., S.A.S., E.A., M.A.)
| | - Amélie Vromman
- The Center for Excellence in Vascular Biology, Cardiovascular Division (S.K., P.K.J., A.L., T.N., L.S.A.P., D.B.-G., T.-D.L., G.C.G., A.V., P.L., E.A., M.A.)
| | - Peter Libby
- The Center for Excellence in Vascular Biology, Cardiovascular Division (S.K., P.K.J., A.L., T.N., L.S.A.P., D.B.-G., T.-D.L., G.C.G., A.V., P.L., E.A., M.A.)
| | - C. Keith Ozaki
- Center for Complex Network Intelligence at the Tsinghua Laboratory of Brain and Intelligence, Department of Bioengineering, Tsinghua University, Beijing, China (A.M., C.V.C.)
| | - Amitabh Sharma
- The Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (M.A.R., J.L.D., L.H.L., I.A., A.H., H.H., H.Z., A.S., S.A.S., E.A., M.A.)
- Channing Division of Network Medicine (I.A., A.H., A.S., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Sasha A. Singh
- The Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (M.A.R., J.L.D., L.H.L., I.A., A.H., H.H., H.Z., A.S., S.A.S., E.A., M.A.)
| | - Elena Aikawa
- The Center for Excellence in Vascular Biology, Cardiovascular Division (S.K., P.K.J., A.L., T.N., L.S.A.P., D.B.-G., T.-D.L., G.C.G., A.V., P.L., E.A., M.A.)
- The Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (M.A.R., J.L.D., L.H.L., I.A., A.H., H.H., H.Z., A.S., S.A.S., E.A., M.A.)
| | - Masanori Aikawa
- The Center for Excellence in Vascular Biology, Cardiovascular Division (S.K., P.K.J., A.L., T.N., L.S.A.P., D.B.-G., T.-D.L., G.C.G., A.V., P.L., E.A., M.A.)
- The Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (M.A.R., J.L.D., L.H.L., I.A., A.H., H.H., H.Z., A.S., S.A.S., E.A., M.A.)
- Channing Division of Network Medicine (I.A., A.H., A.S., M.A.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Tang Y, Chen Y, Zhang Z, Tang B, Zhou Z, Chen H. Nanoparticle-Based RNAi Therapeutics Targeting Cancer Stem Cells: Update and Prospective. Pharmaceutics 2021; 13:pharmaceutics13122116. [PMID: 34959397 PMCID: PMC8708448 DOI: 10.3390/pharmaceutics13122116] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/19/2021] [Accepted: 12/02/2021] [Indexed: 02/05/2023] Open
Abstract
Cancer stem cells (CSCs) are characterized by intrinsic self-renewal and tumorigenic properties, and play important roles in tumor initiation, progression, and resistance to diverse forms of anticancer therapy. Accordingly, targeting signaling pathways that are critical for CSC maintenance and biofunctions, including the Wnt, Notch, Hippo, and Hedgehog signaling cascades, remains a promising therapeutic strategy in multiple cancer types. Furthermore, advances in various cancer omics approaches have largely increased our knowledge of the molecular basis of CSCs, and provided numerous novel targets for anticancer therapy. However, the majority of recently identified targets remain ‘undruggable’ through small-molecule agents, whereas the implications of exogenous RNA interference (RNAi, including siRNA and miRNA) may make it possible to translate our knowledge into therapeutics in a timely manner. With the recent advances of nanomedicine, in vivo delivery of RNAi using elaborate nanoparticles can potently overcome the intrinsic limitations of RNAi alone, as it is rapidly degraded and has unpredictable off-target side effects. Herein, we present an update on the development of RNAi-delivering nanoplatforms in CSC-targeted anticancer therapy and discuss their potential implications in clinical trials.
Collapse
Affiliation(s)
- Yongquan Tang
- Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu 610041, China;
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (Z.Z.)
| | - Yan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (Z.Z.)
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (Z.Z.)
| | - Bo Tang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Zongguang Zhou
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (Z.Z.)
- Correspondence: (Z.Z.); (H.C.)
| | - Haining Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.C.); (Z.Z.)
- Correspondence: (Z.Z.); (H.C.)
| |
Collapse
|
8
|
Matsubara Y, Gonzalez L, Kiwan G, Liu J, Langford J, Gao M, Gao X, Taniguchi R, Yatsula B, Furuyama T, Matsumoto T, Komori K, Mori M, Dardik A. PD-L1 (Programmed Death Ligand 1) Regulates T-Cell Differentiation to Control Adaptive Venous Remodeling. Arterioscler Thromb Vasc Biol 2021; 41:2909-2922. [PMID: 34670406 PMCID: PMC8664128 DOI: 10.1161/atvbaha.121.316380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Patients with end-stage renal disease depend on hemodialysis for survival. Although arteriovenous fistulae (AVF) are the preferred vascular access for hemodialysis, the primary success rate of AVF is only 30% to 50% within 6 months, showing an urgent need for improvement. PD-L1 (programmed death ligand 1) is a ligand that regulates T-cell activity. Since T cells have an important role during AVF maturation, we hypothesized that PD-L1 regulates T cells to control venous remodeling that occurs during AVF maturation. Approach and results: In the mouse aortocaval fistula model, anti-PD-L1 antibody (200 mg, 3×/wk intraperitoneal) was given to inhibit PD-L1 activity during AVF maturation. Inhibition of PD-L1 increased T-helper type 1 cells and T-helper type 2 cells but reduced regulatory T cells to increase M1-type macrophages and reduce M2-type macrophages; these changes were associated with reduced vascular wall thickening and reduced AVF patency. Inhibition of PD-L1 also inhibited smooth muscle cell proliferation and increased endothelial dysfunction. The effects of anti-PD-L1 antibody on adaptive venous remodeling were diminished in nude mice; however, they were restored after T-cell transfer into nude mice, indicating the effects of anti-PD-L1 antibody on venous remodeling were dependent on T cells. CONCLUSIONS Regulation of PD-L1 activity may be a potential therapeutic target for clinical translation to improve AVF maturation.
Collapse
Affiliation(s)
- Yutaka Matsubara
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
- Department of Surgery and Sciences, Kyushu University, Fukuoka, Japan
| | - Luis Gonzalez
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - Gathe Kiwan
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - Jia Liu
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - John Langford
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - Mingjie Gao
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xixiang Gao
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ryosuke Taniguchi
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
- Division of Vascular Surgery, The University of Tokyo, Tokyo, Japan
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - Tadashi Furuyama
- Department of Surgery and Sciences, Kyushu University, Fukuoka, Japan
| | - Takuya Matsumoto
- Department of Vascular Surgery, Kyushu Central Hospital, Fukuoka, Japan
| | - Kimihiro Komori
- Division of Vascular Surgery, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masaki Mori
- Department of Surgery and Sciences, Kyushu University, Fukuoka, Japan
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
- Division of Vascular and Endovascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT
| |
Collapse
|
9
|
Wen J, Chen C, Luo M, Liu X, Guo J, Wei T, Gu X, Gu S, Ning Y, Li Y. Notch Signaling Ligand Jagged1 Enhances Macrophage-Mediated Response to Helicobacter pylori. Front Microbiol 2021; 12:692832. [PMID: 34305857 PMCID: PMC8297740 DOI: 10.3389/fmicb.2021.692832] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022] Open
Abstract
Helicobacter pylori (H. pylori) is one of the gram-negative bacteria that mainly colonize the stomach mucosa and cause many gastrointestinal diseases, such as gastritis, peptic ulcer, and gastric cancer. Macrophages play a key role in eradicating H. pylori. Recent data have shown that Notch signaling could modulate the activation and bactericidal activities of macrophages. However, the role of Notch signaling in macrophages against H. pylori remains unclear. In the present study, in the co-culture model of macrophages with H. pylori, the inhibition of Notch signaling using γ-secretase decreased the expression of inducible nitric oxide synthase (iNOS) and its product, nitric oxide (NO), and downregulated the secretion of pro-inflammatory cytokine and attenuated phagocytosis and bactericidal activities of macrophages to H. pylori. Furthermore, we identified that Jagged1, one of Notch signaling ligands, was both upregulated in mRNA and protein level in activated macrophages induced by H. pylori. Clinical specimens showed that the number of Jagged1+ macrophages in the stomach mucosa from H. pylori-infected patients was significantly higher than that in healthy control. The overexpression of Jagged1 promoted bactericidal activities of macrophages against H. pylori and siRNA-Jagged1 presented the opposite effect. Besides, the addition of exogenous rJagged1 facilitated the pro-inflammatory mediators of macrophages against H. pylori, but the treatment of anti-Jagged1 neutralizing antibody attenuated it. Taken together, these results suggest that Jagged1 is a promoting molecule for macrophages against H. pylori, which will provide insight for exploring Jagged1 as a novel therapeutic target for the control of H. pylori infection.
Collapse
Affiliation(s)
- Junjie Wen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Chuxi Chen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Meiqun Luo
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xiaocong Liu
- The First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Jiading Guo
- The First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Tingting Wei
- The First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Xinyi Gu
- The First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Sinan Gu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yunshan Ning
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yan Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| |
Collapse
|
10
|
Decano JL, Singh SA, Gasparotto Bueno C, Ho Lee L, Halu A, Chelvanambi S, Matamalas JT, Zhang H, Mlynarchik AK, Qiao J, Sharma A, Mukai S, Wang J, Anderson DG, Ozaki CK, Libby P, Aikawa E, Aikawa M. Systems Approach to Discovery of Therapeutic Targets for Vein Graft Disease: PPARα Pivotally Regulates Metabolism, Activation, and Heterogeneity of Macrophages and Lesion Development. Circulation 2021; 143:2454-2470. [PMID: 33821665 PMCID: PMC8212880 DOI: 10.1161/circulationaha.119.043724] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Supplemental Digital Content is available in the text. Vein graft failure remains a common clinical challenge. We applied a systems approach in mouse experiments to discover therapeutic targets for vein graft failure.
Collapse
Affiliation(s)
- Julius L Decano
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., S.A.S., C.G.B., L.H.L., A.H., S.C., J.T.M., H.Z., A.K.M., J.Q., A.S., S.M., J.W., E.A., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., S.A.S., C.G.B., L.H.L., A.H., S.C., J.T.M., H.Z., A.K.M., J.Q., A.S., S.M., J.W., E.A., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Cauê Gasparotto Bueno
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., S.A.S., C.G.B., L.H.L., A.H., S.C., J.T.M., H.Z., A.K.M., J.Q., A.S., S.M., J.W., E.A., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Lang Ho Lee
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., S.A.S., C.G.B., L.H.L., A.H., S.C., J.T.M., H.Z., A.K.M., J.Q., A.S., S.M., J.W., E.A., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Arda Halu
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., S.A.S., C.G.B., L.H.L., A.H., S.C., J.T.M., H.Z., A.K.M., J.Q., A.S., S.M., J.W., E.A., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Channing Division of Network Medicine (A.H., A.S., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sarvesh Chelvanambi
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., S.A.S., C.G.B., L.H.L., A.H., S.C., J.T.M., H.Z., A.K.M., J.Q., A.S., S.M., J.W., E.A., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Joan T Matamalas
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., S.A.S., C.G.B., L.H.L., A.H., S.C., J.T.M., H.Z., A.K.M., J.Q., A.S., S.M., J.W., E.A., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hengmin Zhang
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., S.A.S., C.G.B., L.H.L., A.H., S.C., J.T.M., H.Z., A.K.M., J.Q., A.S., S.M., J.W., E.A., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Andrew K Mlynarchik
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., S.A.S., C.G.B., L.H.L., A.H., S.C., J.T.M., H.Z., A.K.M., J.Q., A.S., S.M., J.W., E.A., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Jiao Qiao
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., S.A.S., C.G.B., L.H.L., A.H., S.C., J.T.M., H.Z., A.K.M., J.Q., A.S., S.M., J.W., E.A., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Amitabh Sharma
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., S.A.S., C.G.B., L.H.L., A.H., S.C., J.T.M., H.Z., A.K.M., J.Q., A.S., S.M., J.W., E.A., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Channing Division of Network Medicine (A.H., A.S., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Shin Mukai
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., S.A.S., C.G.B., L.H.L., A.H., S.C., J.T.M., H.Z., A.K.M., J.Q., A.S., S.M., J.W., E.A., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Jianguo Wang
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., S.A.S., C.G.B., L.H.L., A.H., S.C., J.T.M., H.Z., A.K.M., J.Q., A.S., S.M., J.W., E.A., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Daniel G Anderson
- Institutes for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge (D.G.A.)
| | - C Keith Ozaki
- Department of Medicine, Division of Vascular and Endovascular Surgery, Department of Surgery (C.K.O.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Peter Libby
- Center for Excellence in Vascular Biology (P.L., E.A., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., S.A.S., C.G.B., L.H.L., A.H., S.C., J.T.M., H.Z., A.K.M., J.Q., A.S., S.M., J.W., E.A., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Center for Excellence in Vascular Biology (P.L., E.A., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Department of Human Pathology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health, Russia (E.A., M.A.)
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division (J.L.D., S.A.S., C.G.B., L.H.L., A.H., S.C., J.T.M., H.Z., A.K.M., J.Q., A.S., S.M., J.W., E.A., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Channing Division of Network Medicine (A.H., A.S., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Center for Excellence in Vascular Biology (P.L., E.A., M.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Department of Human Pathology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health, Russia (E.A., M.A.)
| |
Collapse
|
11
|
Katsuki S, Koga JI, Matoba T, Umezu R, Nakashiro S, Nakano K, Tsutsui H, Egashira K. Nanoparticle-Mediated Delivery of Pitavastatin to Monocytes/Macrophages Inhibits Angiotensin II-Induced Abdominal Aortic Aneurysm Formation in Apoe -/- Mice. J Atheroscler Thromb 2021; 29:111-125. [PMID: 33455994 PMCID: PMC8737070 DOI: 10.5551/jat.54379] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Aim:
Abdominal aortic aneurysm (AAA) is a lethal and multifactorial disease. To prevent a rupture and dissection of enlarged AAA, prophylactic surgery and stenting are currently available. There are, however, no medical therapies preventing these complications of AAA. Statin is one of the candidates, but its efficacy on AAA formation/progression remains controversial. We have previously demonstrated that nanoparticles (NPs) incorporating pitavastatin (Pitava-NPs)—clinical trials using these nanoparticles have been already conducted—suppressed progression of atherosclerosis in apolipoprotein E-deficient (
Apoe−/−
) mice. Therefore, we have tested a hypothesis that monocytes/macrophages-targeting delivery of pitavastatin prevents the progression of AAA.
Methods:
Angiotensin II was intraperitoneally injected by osmotic mini-pumps to induce AAA formation in
Apoe−/−
mice. NPs consisting of poly(lactic-co-glycolic acid) were used for
in vivo
delivery of pitavastatin to monocytes/macrophages.
Results:
Intravenously administered Pitava-NPs (containing 0.012 mg/kg/week pitavastatin) inhibited AAA formation accompanied with reduction of macrophage accumulation and monocyte chemoattractant protein-1 (MCP-1) expression.
Ex vivo
molecular imaging revealed that Pitava-NPs not only reduced macrophage accumulation but also attenuated matrix metalloproteinase activity in the abdominal aorta, which was underpinned by attenuated elastin degradation.
Conclusion:
These results suggest that Pitava-NPs inhibit AAA formation associated with reduced macrophage accumulation and MCP-1 expression. This clinically feasible nanomedicine could be an innovative therapeutic strategy that prevents devastating complications of AAA.
Collapse
Affiliation(s)
- Shunsuke Katsuki
- The Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University
| | - Jun-Ichiro Koga
- The Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University
| | - Tetsuya Matoba
- The Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University
| | - Ryuta Umezu
- The Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University
| | - Soichi Nakashiro
- The Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University
| | - Kaku Nakano
- The Department of Cardiovascular Research, Development, and Translational Medicine, Center for Disruptive Cardiovascular Innovation, Kyushu University
| | - Hiroyuki Tsutsui
- The Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University
| | - Kensuke Egashira
- The Department of Cardiovascular Research, Development, and Translational Medicine, Center for Disruptive Cardiovascular Innovation, Kyushu University.,The Department of Translational Medicine, Kyushu University Graduate School of Pharmaceutical Sciences
| |
Collapse
|
12
|
Brahmbhatt AN, Misra S. Stem Cell Delivery for the Treatment of Arteriovenous Fistula Failure. STEM CELL THERAPY FOR VASCULAR DISEASES 2021:281-297. [DOI: 10.1007/978-3-030-56954-9_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
13
|
Stassen OMJA, Ristori T, Sahlgren CM. Notch in mechanotransduction - from molecular mechanosensitivity to tissue mechanostasis. J Cell Sci 2020; 133:133/24/jcs250738. [PMID: 33443070 DOI: 10.1242/jcs.250738] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tissue development and homeostasis are controlled by mechanical cues. Perturbation of the mechanical equilibrium triggers restoration of mechanostasis through changes in cell behavior, while defects in these restorative mechanisms lead to mechanopathologies, for example, osteoporosis, myopathies, fibrosis or cardiovascular disease. Therefore, sensing mechanical cues and integrating them with the biomolecular cell fate machinery is essential for the maintenance of health. The Notch signaling pathway regulates cell and tissue fate in nearly all tissues. Notch activation is directly and indirectly mechanosensitive, and regulation of Notch signaling, and consequently cell fate, is integral to the cellular response to mechanical cues. Fully understanding the dynamic relationship between molecular signaling, tissue mechanics and tissue remodeling is challenging. To address this challenge, engineered microtissues and computational models play an increasingly large role. In this Review, we propose that Notch takes on the role of a 'mechanostat', maintaining the mechanical equilibrium of tissues. We discuss the reciprocal role of Notch in the regulation of tissue mechanics, with an emphasis on cardiovascular tissues, and the potential of computational and engineering approaches to unravel the complex dynamic relationship between mechanics and signaling in the maintenance of cell and tissue mechanostasis.
Collapse
Affiliation(s)
- Oscar M J A Stassen
- Faculty of Science and Engineering, Biosciences, Åbo Akademi University, 20500 Turku, Finland.,Turku Bioscience Centre, Åbo Akademi University and University of Turku, 20520 Turku, Finland.,Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Tommaso Ristori
- Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.,Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Cecilia M Sahlgren
- Faculty of Science and Engineering, Biosciences, Åbo Akademi University, 20500 Turku, Finland .,Turku Bioscience Centre, Åbo Akademi University and University of Turku, 20520 Turku, Finland.,Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
14
|
Spadaccio C, Antoniades C, Nenna A, Chung C, Will R, Chello M, Gaudino MFL. Preventing treatment failures in coronary artery disease: what can we learn from the biology of in-stent restenosis, vein graft failure, and internal thoracic arteries? Cardiovasc Res 2020; 116:505-519. [PMID: 31397850 DOI: 10.1093/cvr/cvz214] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 06/01/2019] [Accepted: 08/08/2019] [Indexed: 12/18/2022] Open
Abstract
Coronary artery disease (CAD) remains one of the most important causes of morbidity and mortality worldwide, and the availability of percutaneous or surgical revascularization procedures significantly improves survival. However, both strategies are daunted by complications which limit long-term effectiveness. In-stent restenosis (ISR) is a major drawback for intracoronary stenting, while graft failure is the limiting factor for coronary artery bypass graft surgery (CABG), especially using veins. Conversely, internal thoracic artery (ITA) is known to maintain long-term patency in CABG. Understanding the biology and pathophysiology of ISR and vein graft failure (VGF) and mechanisms behind ITA resistance to failure is crucial to combat these complications in CAD treatment. This review intends to provide an overview of the biological mechanisms underlying stent and VGF and of the potential therapeutic strategy to prevent these complications. Interestingly, despite being different modalities of revascularization, mechanisms of failure of stent and saphenous vein grafts are very similar from the biological standpoint.
Collapse
Affiliation(s)
- Cristiano Spadaccio
- Department of Cardiac Surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank, G81 4DY Glasgow, UK
| | | | - Antonio Nenna
- Department of Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Calvin Chung
- Department of Cardiac Surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank, G81 4DY Glasgow, UK
| | - Ricardo Will
- Department of Cardiac Surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank, G81 4DY Glasgow, UK
| | - Massimo Chello
- Department of Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Mario F L Gaudino
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
15
|
Matsubara Y, Kiwan G, Fereydooni A, Langford J, Dardik A. Distinct subsets of T cells and macrophages impact venous remodeling during arteriovenous fistula maturation. JVS Vasc Sci 2020; 1:207-218. [PMID: 33748787 PMCID: PMC7971420 DOI: 10.1016/j.jvssci.2020.07.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Patients with end-stage renal failure depend on hemodialysis indefinitely without renal transplantation, requiring a long-term patent vascular access. While the arteriovenous fistula (AVF) remains the preferred vascular access for hemodialysis because of its longer patency and fewer complications compared with other vascular accesses, the primary patency of AVF is only 50-60%, presenting a clinical need for improvement. AVF mature by developing a thickened vascular wall and increased diameter to adapt to arterial blood pressure and flow volume. Inflammation plays a critical role during vascular remodeling and fistula maturation; increased shear stress triggers infiltration of T-cells and macrophages that initiate inflammation, with involvement of several different subsets of T-cells and macrophages. We review the literature describing distinct roles of the various subsets of T-cells and macrophages during vascular remodeling. Immunosuppression with sirolimus or prednisolone reduces neointimal hyperplasia during AVF maturation, suggesting novel approaches to enhance vascular remodeling. However, M2 macrophages and CD4+ T-cells play essential roles during AVF maturation, suggesting that total immunosuppression may suppress adaptive vascular remodeling. Therefore it is likely that regulation of inflammation during fistula maturation will require a balanced approach to coordinate the various inflammatory cell subsets. Advances in immunosuppressive drug development and delivery systems may allow for more targeted regulation of inflammation to improve vascular remodeling and enhance AVF maturation.
Collapse
Affiliation(s)
- Yutaka Matsubara
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT.,Department of Surgery and Sciences, Kyushu University, Fukuoka, Japan
| | - Gathe Kiwan
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - Arash Fereydooni
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - John Langford
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT.,Division of Vascular and Endovascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT.,Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT
| |
Collapse
|
16
|
Baganha F, de Jong A, Jukema JW, Quax PHA, de Vries MR. The Role of Immunomodulation in Vein Graft Remodeling and Failure. J Cardiovasc Transl Res 2020; 14:100-109. [PMID: 32542547 PMCID: PMC7892738 DOI: 10.1007/s12265-020-10001-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/01/2020] [Indexed: 12/18/2022]
Abstract
Obstructive arterial disease is a major cause of morbidity and mortality in the developed world. Venous bypass graft surgery is one of the most frequently used revascularization strategies despite its considerable short and long time failure rate. Due to vessel wall remodeling, inflammation, intimal hyperplasia, and accelerated atherosclerosis, vein grafts may (ultimately) fail to revascularize tissues downstream to occlusive atherosclerotic lesions. In the past decades, little has changed in the prevention of vein graft failure (VGF) although new insights in the role of innate and adaptive immunity in VGF have emerged. In this review, we discuss the pathophysiological mechanisms underlying the development of VGF, emphasizing the role of immune response and associated factors related to VG remodeling and failure. Moreover, we discuss potential therapeutic options that can improve patency based on data from both preclinical studies and the latest clinical trials. This review contributes to the insights in the role of immunomodulation in vein graft failure in humans. We describe the effects of immune cells and related factors in early (thrombosis), intermediate (inward remodeling and intimal hyperplasia), and late (intimal hyperplasia and accelerated atherosclerosis) failure based on both preclinical (mouse) models and clinical data.
Collapse
Affiliation(s)
- Fabiana Baganha
- Department of Vascular Surgery, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.,Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, Aberdeen University, Aberdeen, UK
| | - Alwin de Jong
- Department of Vascular Surgery, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Paul H A Quax
- Department of Vascular Surgery, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Margreet R de Vries
- Department of Vascular Surgery, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands. .,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
17
|
Umezu R, Koga JI, Matoba T, Katsuki S, Wang L, Hasuzawa N, Nomura M, Tsutsui H, Egashira K. Macrophage (Drp1) Dynamin-Related Protein 1 Accelerates Intimal Thickening After Vascular Injury. Arterioscler Thromb Vasc Biol 2020; 40:e214-e226. [PMID: 32493171 DOI: 10.1161/atvbaha.120.314383] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Mitochondria consistently change their morphology in a process regulated by proteins, including Drp1 (dynamin-related protein 1), a protein promoting mitochondrial fission. Drp1 is involved in the mechanisms underlying various cardiovascular diseases, such as myocardial ischemia/reperfusion injury, heart failure, and pulmonary arterial hypertension. However, its role in macrophages, which promote various vascular diseases, is poorly understood. We therefore tested our hypothesis that macrophage Drp1 promotes vascular remodeling after injury. METHOD AND RESULTS To explore the selective role of macrophage Drp1, we created macrophage-selective Drp1-deficient mice and performed femoral arterial wire injury. In these mice, intimal thickening and negative remodeling were attenuated at 4 weeks after injury when compared with control mice. Deletion of macrophage Drp1 also attenuated the macrophage accumulation and cell proliferation in the injured arteries. Gain- and loss-of-function experiments using cultured macrophages indicated that Drp1 induces the expression of molecules associated with inflammatory macrophages. Morphologically, mitochondrial fission was induced in inflammatory macrophages, whereas mitochondrial fusion was induced in less inflammatory/reparative macrophages. Pharmacological inhibition or knockdown of Drp1 decreased the mitochondrial reactive oxygen species and chemotactic activity in cultured macrophages. Co-culture experiments of macrophages with vascular smooth muscle cells indicated that deletion of macrophage Drp1 suppresses growth and migration of vascular smooth muscle cells induced by macrophage-derived soluble factors. CONCLUSIONS Macrophage Drp1 accelerates intimal thickening after vascular injury by promoting macrophage-mediated inflammation. Macrophage Drp1 may be a potential therapeutic target of vascular diseases.
Collapse
Affiliation(s)
- Ryuta Umezu
- From the Department of Cardiovascular Medicine, Graduate School of Medical Sciences (R.U., J.K., T.M., S.K., H.T.), Kyushu University, Fukuoka, Japan
| | - Jun-Ichiro Koga
- From the Department of Cardiovascular Medicine, Graduate School of Medical Sciences (R.U., J.K., T.M., S.K., H.T.), Kyushu University, Fukuoka, Japan.,the Department of Cardiovascular Research, Development, and Translational Medicine, Center for Cardiovascular Disruptive Innovation (J.K., K.E.), Kyushu University, Fukuoka, Japan
| | - Tetsuya Matoba
- From the Department of Cardiovascular Medicine, Graduate School of Medical Sciences (R.U., J.K., T.M., S.K., H.T.), Kyushu University, Fukuoka, Japan
| | - Shunsuke Katsuki
- From the Department of Cardiovascular Medicine, Graduate School of Medical Sciences (R.U., J.K., T.M., S.K., H.T.), Kyushu University, Fukuoka, Japan
| | - Lixiang Wang
- The Department of Medicine and Bioregulatory Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan (L.W., N.H., M.N.).,Department of Medical Biochemistry (L.W.), Kurume University School of Medicine, Japan
| | - Nao Hasuzawa
- The Department of Medicine and Bioregulatory Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan (L.W., N.H., M.N.).,Division of Endocrinology and Metabolism, Department of Internal Medicine (N.H., M.N.), Kurume University School of Medicine, Japan
| | - Masatoshi Nomura
- The Department of Medicine and Bioregulatory Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan (L.W., N.H., M.N.).,Division of Endocrinology and Metabolism, Department of Internal Medicine (N.H., M.N.), Kurume University School of Medicine, Japan
| | - Hiroyuki Tsutsui
- From the Department of Cardiovascular Medicine, Graduate School of Medical Sciences (R.U., J.K., T.M., S.K., H.T.), Kyushu University, Fukuoka, Japan
| | - Kensuke Egashira
- the Department of Cardiovascular Research, Development, and Translational Medicine, Center for Cardiovascular Disruptive Innovation (J.K., K.E.), Kyushu University, Fukuoka, Japan.,Department of Translational Medicine, Kyushu University Graduate School of Pharmaceutical Sciences, Fukuoka, Japan (K.E.)
| |
Collapse
|
18
|
Kawakami R, Katsuki S, Travers R, Romero DC, Becker-Greene D, Passos LSA, Higashi H, Blaser MC, Sukhova GK, Buttigieg J, Kopriva D, Schmidt AM, Anderson DG, Singh SA, Cardoso L, Weinbaum S, Libby P, Aikawa M, Croce K, Aikawa E. S100A9-RAGE Axis Accelerates Formation of Macrophage-Mediated Extracellular Vesicle Microcalcification in Diabetes Mellitus. Arterioscler Thromb Vasc Biol 2020; 40:1838-1853. [PMID: 32460581 DOI: 10.1161/atvbaha.118.314087] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Vascular calcification is a cardiovascular risk factor and accelerated in diabetes mellitus. Previous work has established a role for calcification-prone extracellular vesicles in promoting vascular calcification. However, the mechanisms by which diabetes mellitus provokes cardiovascular events remain incompletely understood. Our goal was to identify that increased S100A9 promotes the release of calcification-prone extracellular vesicles from human macrophages in diabetes mellitus. Approach and Results: Human primary macrophages exposed to high glucose (25 mmol/L) increased S100A9 secretion and the expression of receptor for advanced glycation end products (RAGE) protein. Recombinant S100A9 induced the expression of proinflammatory and osteogenic factors, as well as the number of extracellular vesicles with high calcific potential (alkaline phosphatase activity, P<0.001) in macrophages. Treatment with a RAGE antagonist or silencing with S100A9 siRNA in macrophages abolished these responses, suggesting that stimulation of the S100A9-RAGE axis by hyperglycemia favors a procalcific environment. We further showed that an imbalance between Nrf-2 (nuclear factor 2 erythroid related factor 2) and NF-κB (nuclear factor-κB) pathways contributes to macrophage activation and promotes a procalcific environment. In addition, streptozotocin-induced diabetic Apoe-/-S100a9-/- mice and mice treated with S100a9 siRNA encapsulated in macrophage-targeted lipid nanoparticles showed decreased inflammation and microcalcification in atherosclerotic plaques, as gauged by molecular imaging and comprehensive histological analysis. In human carotid plaques, comparative proteomics in patients with diabetes mellitus and histological analysis showed that the S100A9-RAGE axis associates with osteogenic activity and the formation of microcalcification. CONCLUSIONS Under hyperglycemic conditions, macrophages release calcific extracellular vesicles through mechanisms involving the S100A9-RAGE axis, thus contributing to the formation of microcalcification within atherosclerotic plaques.
Collapse
Affiliation(s)
- Ryo Kawakami
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Shunsuke Katsuki
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Richard Travers
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Dayanna Carolina Romero
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Dakota Becker-Greene
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Livia Silva Araujo Passos
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hideyuki Higashi
- Center for Interdisciplinary Cardiovascular Sciences (H.H., M.C.B., S.A.S., M.A., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Mark C Blaser
- Center for Interdisciplinary Cardiovascular Sciences (H.H., M.C.B., S.A.S., M.A., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Galina K Sukhova
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Josef Buttigieg
- Department of Biology, University of Regina, Saskatchewan, Canada (J.B.)
| | - David Kopriva
- Regina Qu'Appelle Health Region, University of Saskatchewan, Regina, Canada (D.K.)
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University (A.M.S.)
| | - Daniel G Anderson
- Institutes for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge (D.G.A.)
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences (H.H., M.C.B., S.A.S., M.A., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Luis Cardoso
- Department of Biomedical Engineering, The City College of New York (L.C., S.W.)
| | - Sheldon Weinbaum
- Department of Biomedical Engineering, The City College of New York (L.C., S.W.)
| | - Peter Libby
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Masanori Aikawa
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Center for Interdisciplinary Cardiovascular Sciences (H.H., M.C.B., S.A.S., M.A., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Kevin Croce
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Elena Aikawa
- From the Center for Excellence in Vascular Biology (R.K., S.K., R.T., D.C.R., D.B.-G., L.S.A.P., G.K.S., P.L., M.A., K.C., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Center for Interdisciplinary Cardiovascular Sciences (H.H., M.C.B., S.A.S., M.A., E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Department of Human Pathology, Sechenov First Moscow State Medical University, Russia (E.A.)
| |
Collapse
|
19
|
Wang Y, Dubland JA, Allahverdian S, Asonye E, Sahin B, Jaw JE, Sin DD, Seidman MA, Leeper NJ, Francis GA. Smooth Muscle Cells Contribute the Majority of Foam Cells in ApoE (Apolipoprotein E)-Deficient Mouse Atherosclerosis. Arterioscler Thromb Vasc Biol 2020; 39:876-887. [PMID: 30786740 DOI: 10.1161/atvbaha.119.312434] [Citation(s) in RCA: 244] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objective- Smooth muscle cells (SMCs) are the most abundant cells in human atherosclerotic lesions and are suggested to contribute at least 50% of atheroma foam cells. In mice, SMCs contribute fewer total lesional cells. The purpose of this study was to determine the contribution of SMCs to total foam cells in apolipoprotein E-deficient (ApoE-/-) mice, and the utility of these mice to model human SMC foam cell biology and interventions. Approach and Results- Using flow cytometry, foam cells in the aortic arch of ApoE-/- mice were characterized based on the expression of leukocyte-specific markers. Nonleukocyte foam cells increased from 37% of total foam cells in 27-week-old to 75% in 57-week-old male ApoE-/- mice fed a chow diet and were ≈70% in male and female ApoE-/- mice following 6 weeks of Western diet feeding. A similar contribution to total foam cells by SMCs was found using SMC-lineage tracing ApoE-/- mice fed the Western diet for 6 or 12 weeks. Nonleukocyte foam cells contributed a similar percentage of total atheroma cholesterol and exhibited lower expression of the cholesterol exporter ABCA1 (ATP-binding cassette transporter A1) when compared with leukocyte-derived foam cells. Conclusions- Consistent with previous studies of human atheromas, we present evidence that SMCs contribute the majority of atheroma foam cells in ApoE-/- mice fed a Western diet and a chow diet for longer periods. Reduced expression of ABCA1, also seen in human intimal SMCs, suggests a common mechanism for formation of SMC foam cells across species, and represents a novel target to enhance atherosclerosis regression.
Collapse
Affiliation(s)
- Ying Wang
- From the Departments of Medicine (Y.W., J.A.D., S.A., E.A., B.S., J.E.J., D.D.S., G.A.F.), Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St Paul's Hospital, University of British Columbia, Vancouver, Canada.,Division of Vascular Surgery, Stanford University, CA (Y.W., N.J.L.)
| | - Joshua A Dubland
- From the Departments of Medicine (Y.W., J.A.D., S.A., E.A., B.S., J.E.J., D.D.S., G.A.F.), Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St Paul's Hospital, University of British Columbia, Vancouver, Canada
| | - Sima Allahverdian
- From the Departments of Medicine (Y.W., J.A.D., S.A., E.A., B.S., J.E.J., D.D.S., G.A.F.), Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St Paul's Hospital, University of British Columbia, Vancouver, Canada
| | - Enyinnaya Asonye
- From the Departments of Medicine (Y.W., J.A.D., S.A., E.A., B.S., J.E.J., D.D.S., G.A.F.), Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St Paul's Hospital, University of British Columbia, Vancouver, Canada
| | - Basak Sahin
- From the Departments of Medicine (Y.W., J.A.D., S.A., E.A., B.S., J.E.J., D.D.S., G.A.F.), Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St Paul's Hospital, University of British Columbia, Vancouver, Canada
| | - Jen Erh Jaw
- From the Departments of Medicine (Y.W., J.A.D., S.A., E.A., B.S., J.E.J., D.D.S., G.A.F.), Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St Paul's Hospital, University of British Columbia, Vancouver, Canada
| | - Don D Sin
- From the Departments of Medicine (Y.W., J.A.D., S.A., E.A., B.S., J.E.J., D.D.S., G.A.F.), Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St Paul's Hospital, University of British Columbia, Vancouver, Canada
| | - Michael A Seidman
- Pathology and Laboratory Medicine (M.A.S.), Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St Paul's Hospital, University of British Columbia, Vancouver, Canada
| | - Nicholas J Leeper
- Division of Vascular Surgery, Stanford University, CA (Y.W., N.J.L.)
| | - Gordon A Francis
- From the Departments of Medicine (Y.W., J.A.D., S.A., E.A., B.S., J.E.J., D.D.S., G.A.F.), Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St Paul's Hospital, University of British Columbia, Vancouver, Canada
| |
Collapse
|
20
|
Affiliation(s)
- Ziad Mallat
- From the Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, United Kingdom; and Institut National de la Santé et de la Recherche Médicale, Paris, France.
| |
Collapse
|
21
|
The Use of Nutraceuticals to Counteract Atherosclerosis: The Role of the Notch Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5470470. [PMID: 31915510 PMCID: PMC6935452 DOI: 10.1155/2019/5470470] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 03/13/2019] [Indexed: 12/13/2022]
Abstract
Despite the currently available pharmacotherapies, today, thirty percent of worldwide deaths are due to cardiovascular diseases (CVDs), whose primary cause is atherosclerosis, an inflammatory disorder characterized by the buildup of lipid deposits on the inside of arteries. Multiple cellular signaling pathways have been shown to be involved in the processes underlying atherosclerosis, and evidence has been accumulating for the crucial role of Notch receptors in regulating the functions of the diverse cell types involved in atherosclerosis onset and progression. Several classes of nutraceuticals have potential benefits for the prevention and treatment of atherosclerosis and CVDs, some of which could in part be due to their ability to modulate the Notch pathway. In this review, we summarize the current state of knowledge on the role of Notch in vascular health and its modulation by nutraceuticals for the prevention of atherosclerosis and/or treatment of related CVDs.
Collapse
|
22
|
Rehfuss JP, DeSart KM, Rozowsky JM, O'Malley KA, Moldawer LL, Baker HV, Wang Y, Wu R, Nelson PR, Berceli SA. Hyperacute Monocyte Gene Response Patterns Are Associated With Lower Extremity Vein Bypass Graft Failure. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019. [PMID: 29530886 DOI: 10.1161/circgen.117.001970] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Despite being the definitive treatment for lower extremity peripheral arterial disease, vein bypass grafts fail in half of all cases. Early repair mechanisms after implantation, governed largely by the immune environment, contribute significantly to long-term outcomes. The current study investigates the early response patterns of circulating monocytes as a determinant of graft outcome. METHODS In 48 patients undergoing infrainguinal vein bypass grafting, the transcriptomes of circulating monocytes were analyzed preoperatively and at 1, 7, and 28 days post-operation. RESULTS Dynamic clustering algorithms identified 50 independent gene response patterns. Three clusters (64 genes) were differentially expressed, with a hyperacute response pattern defining those patients with failed versus patent grafts 12 months post-operation. A second independent data set, comprised of 96 patients subjected to major trauma, confirmed the value of these 64 genes in predicting an uncomplicated versus complicated recovery. Causal network analysis identified 8 upstream elements that regulate these mediator genes, and Bayesian analysis with a priori knowledge of the biological interactions was integrated to create a functional network describing the relationships among the regulatory elements and downstream mediator genes. Linear models predicted the removal of either STAT3 (signal transducer and activator of transcription 3) or MYD88 (myeloid differentiation primary response 88) to shift mediator gene expression levels toward those seen in successful grafts. CONCLUSIONS A novel combination of dynamic gene clustering, linear models, and Bayesian network analysis has identified a core set of regulatory genes whose manipulations could migrate vein grafts toward a more favorable remodeling phenotype.
Collapse
Affiliation(s)
- Jonathan P Rehfuss
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.)
| | - Kenneth M DeSart
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.)
| | - Jared M Rozowsky
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.)
| | - Kerri A O'Malley
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.)
| | - Lyle L Moldawer
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.)
| | - Henry V Baker
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.)
| | - Yaqun Wang
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.)
| | - Rongling Wu
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.)
| | - Peter R Nelson
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.)
| | - Scott A Berceli
- From the Malcom Randall Veterans Affairs Medical Center, Gainesville, FL (J.P.R., K.M.D., J.M.R., K.A.O., S.A.B.); Department of Surgery (J.P.R., K.M.D., J.M.R., K.A.O., L.L.M., S.A.B.) and Department of Molecular Genetics and Microbiology (H.V.B.), University of Florida, Gainesville; Department of Biostatistics, Rutgers University, New Brunswick, NJ (Y.W.); Center for Statistical Genetics, Pennsylvania State University, Hershey (R.W.); and Department of Surgery, University of South Florida, Tampa (P.R.N.).
| |
Collapse
|
23
|
Nakano T, Katsuki S, Chen M, Decano JL, Halu A, Lee LH, Pestana DVS, Kum AST, Kuromoto RK, Golden WS, Boff MS, Guimaraes GC, Higashi H, Kauffman KJ, Maejima T, Suzuki T, Iwata H, Barabási AL, Aster JC, Anderson DG, Sharma A, Singh SA, Aikawa E, Aikawa M. Uremic Toxin Indoxyl Sulfate Promotes Proinflammatory Macrophage Activation Via the Interplay of OATP2B1 and Dll4-Notch Signaling. Circulation 2019; 139:78-96. [PMID: 30586693 PMCID: PMC6311723 DOI: 10.1161/circulationaha.118.034588] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Chronic kidney disease (CKD) increases cardiovascular risk. Underlying mechanisms, however, remain obscure. The uremic toxin indoxyl sulfate is an independent cardiovascular risk factor in CKD. We explored the potential impact of indoxyl sulfate on proinflammatory activation of macrophages and its underlying mechanisms. METHODS We examined in vitro the effects of clinically relevant concentrations of indoxyl sulfate on proinflammatory responses of macrophages and the roles of organic anion transporters and organic anion transporting polypeptides (OATPs). A systems approach, involving unbiased global proteomics, bioinformatics, and network analysis, then explored potential key pathways. To address the role of Delta-like 4 (Dll4) in indoxyl sulfate-induced macrophage activation and atherogenesis in CKD in vivo, we used 5/6 nephrectomy and Dll4 antibody in low-density lipoprotein receptor-deficient (Ldlr-/-) mice. To further determine the relative contribution of OATP2B1 or Dll4 to proinflammatory activation of macrophages and atherogenesis in vivo, we used siRNA delivered by macrophage-targeted lipid nanoparticles in mice. RESULTS We found that indoxyl sulfate-induced proinflammatory macrophage activation is mediated by its uptake through transporters, including OATP2B1, encoded by the SLCO2B1 gene. The global proteomics identified potential mechanisms, including Notch signaling and the ubiquitin-proteasome pathway, that mediate indoxyl sulfate-triggered proinflammatory macrophage activation. We chose the Notch pathway as an example of key candidates for validation of our target discovery platform and for further mechanistic studies. As predicted computationally, indoxyl sulfate triggered Notch signaling, which was preceded by the rapid induction of Dll4 protein. Dll4 induction may result from inhibition of the ubiquitin-proteasome pathway, via the deubiquitinating enzyme USP5. In mice, macrophage-targeted OATP2B1/Slco2b1 silencing and Dll4 antibody inhibited proinflammatory activation of peritoneal macrophages induced by indoxyl sulfate. In low-density lipoprotein receptor-deficient mice, Dll4 antibody abolished atherosclerotic lesion development accelerated in Ldlr-/- mice. Moreover, coadministration of indoxyl sulfate and OATP2B1/Slco2b1 or Dll4 siRNA encapsulated in macrophage-targeted lipid nanoparticles in Ldlr-/- mice suppressed lesion development. CONCLUSIONS These results suggest that novel crosstalk between OATP2B1 and Dll4-Notch signaling in macrophages mediates indoxyl sulfate-induced vascular inflammation in CKD.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Animals
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Calcium-Binding Proteins
- Disease Models, Animal
- Humans
- Indican/toxicity
- Inflammation Mediators/metabolism
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Macrophage Activation/drug effects
- Macrophages/drug effects
- Macrophages/metabolism
- Macrophages/pathology
- Male
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Knockout
- Organic Anion Transporters/genetics
- Organic Anion Transporters/metabolism
- Phenotype
- Plaque, Atherosclerotic
- RAW 264.7 Cells
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Signal Transduction/drug effects
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
Collapse
Affiliation(s)
- Toshiaki Nakano
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Shunsuke Katsuki
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Mingxian Chen
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Julius L. Decano
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Arda Halu
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Lang Ho Lee
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Diego V. S. Pestana
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Angelo S. T. Kum
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Rodrigo K. Kuromoto
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Whitney S. Golden
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Mario S. Boff
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Gabriel C. Guimaraes
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Hideyuki Higashi
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Kevin J. Kauffman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA
| | - Takashi Maejima
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Takehiro Suzuki
- Division of Medical Science, Tohoku University Graduate School of Biomedical Engineering, Sendai, Japan
| | - Hiroshi Iwata
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Albert-László Barabási
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Center for Complex Network Research, Northeastern University, Boston, MA
| | - Jon C. Aster
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Daniel G. Anderson
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA
| | - Amitabh Sharma
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Sasha A. Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Elena Aikawa
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Masanori Aikawa
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
24
|
Wolf K, Hu H, Isaji T, Dardik A. Molecular identity of arteries, veins, and lymphatics. J Vasc Surg 2019; 69:253-262. [PMID: 30154011 PMCID: PMC6309638 DOI: 10.1016/j.jvs.2018.06.195] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/25/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Arteries, veins, and lymphatic vessels are distinguished by structural differences that correspond to their different functions. Each of these vessels is also defined by specific molecular markers that persist throughout adult life; these markers are some of the molecular determinants that control the differentiation of embryonic undifferentiated cells into arteries, veins, or lymphatics. METHODS This is a review of experimental literature. RESULTS The Eph-B4 receptor and its ligand, ephrin-B2, are critical molecular determinants of vessel identity, arising on endothelial cells early in embryonic development. Eph-B4 and ephrin-B2 continue to be expressed on adult vessels and mark vessel identity. However, after vascular surgery, vessel identity can change and is marked by altered Eph-B4 and ephrin-B2 expression. Vein grafts show loss of venous identity, with less Eph-B4 expression. Arteriovenous fistulas show gain of dual arterial-venous identity, with both Eph-B4 and ephrin-B2 expression, and manipulation of Eph-B4 improves arteriovenous fistula patency. Patches used to close arteries and veins exhibit context-dependent gain of identity, that is, patches in the arterial environment gain arterial identity, whereas patches in the venous environment gain venous identity; these results show the importance of the host infiltrating cells in determining vascular identity after vascular surgery. CONCLUSIONS Changes in the vessel's molecular identity after vascular surgery correspond to structural changes that depend on the host's postsurgical environment. Regulation of vascular identity and the underlying molecular mechanisms may allow new therapeutic approaches to improve vascular surgical procedures.
Collapse
Affiliation(s)
- Katharine Wolf
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Haidi Hu
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Toshihiko Isaji
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Alan Dardik
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn; VA Connecticut Healthcare System, West Haven, Conn.
| |
Collapse
|
25
|
Sago CD, Lokugamage MP, Islam FZ, Krupczak BR, Sato M, Dahlman JE. Nanoparticles That Deliver RNA to Bone Marrow Identified by in Vivo Directed Evolution. J Am Chem Soc 2018; 140:17095-17105. [PMID: 30394729 PMCID: PMC6556374 DOI: 10.1021/jacs.8b08976] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bone marrow endothelial cells (BMECs) regulate their microenvironment, which includes hematopoietic stem cells. This makes BMECs an important target cell type for siRNA or gene editing (e.g., CRISPR) therapies. However, siRNA and sgRNA have not been delivered to BMECs using systemically administered nanoparticles. Given that in vitro nanoparticle screens have not identified nanoparticles with BMEC tropism, we developed a system to quantify how >100 different nanoparticles deliver siRNA in a single mouse. This is the first barcoding system capable of quantifying functional cytosolic siRNA delivery (where the siRNA drug is active), distinguishing it from in vivo screens that quantify biodistribution (where the siRNA drug went). Combining this approach with bioinformatics, we performed in vivo directed evolution, and identified BM1, a lipid nanoparticle (LNP) that delivers siRNA and sgRNA to BMECs. Interestingly, chemical analysis revealed BMEC tropism was not related to LNP size; tropism changed with the structure of poly(ethylene glycol), as well as the presence of cholesterol. These results suggest that significant changes to vascular targeting can be imparted to a LNP by making simple changes to its chemical composition, rather than using active targeting ligands. BM1 is the first nanoparticle to efficiently deliver siRNA and sgRNA to BMECs in vivo, demonstrating that this functional in vivo screen can identify nanoparticles with novel tropism in vivo. More generally, in vivo screening may help reveal the complex relationship between nanoparticle structure and tropism, thereby helping scientists understand how simple chemical changes control nanoparticle targeting.
Collapse
Affiliation(s)
- Cory D Sago
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - Melissa P Lokugamage
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - Fatima Z Islam
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - Brandon R Krupczak
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - Manaka Sato
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| |
Collapse
|
26
|
Paunovska K, Gil CJ, Lokugamage MP, Sago CD, Sato M, Lando GN, Gamboa Castro M, Bryksin AV, Dahlman JE. Analyzing 2000 in Vivo Drug Delivery Data Points Reveals Cholesterol Structure Impacts Nanoparticle Delivery. ACS NANO 2018; 12:8341-8349. [PMID: 30016076 PMCID: PMC6115295 DOI: 10.1021/acsnano.8b03640] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Lipid nanoparticles (LNPs) are formulated using unmodified cholesterol. However, cholesterol is naturally esterified and oxidized in vivo, and these cholesterol variants are differentially trafficked in vivo via lipoproteins including LDL and VLDL. We hypothesized that incorporating the same cholesterol variants into LNPs-which can be structurally similar to LDL and VLDL-would alter nanoparticle targeting in vivo. To test this hypothesis, we quantified how >100 LNPs made with six cholesterol variants delivered DNA barcodes to 18 cell types in wild-type, LDLR-/-, and VLDLR-/- mice that were both age-matched and female. By analyzing ∼2000 in vivo drug delivery data points, we found that LNPs formulated with esterified cholesterol delivered nucleic acids more efficiently than LNPs formulated with regular or oxidized cholesterol when compared across all tested cell types in the mouse. We also identified an LNP containing cholesteryl oleate that efficiently delivered siRNA and sgRNA to liver endothelial cells in vivo. Delivery was as-or more-efficient as the same LNP made with unmodified cholesterol. Moreover, delivery to liver endothelial cells was 3 times more efficient than delivery to hepatocytes, distinguishing this oleate LNP from hepatocyte-targeting LNPs. RNA delivery can be improved by rationally selecting cholesterol variants, allowing optimization of nanoparticle targeting.
Collapse
Affiliation(s)
- Kalina Paunovska
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory School of Medicine , Atlanta , Georgia 30332 , United States
| | - Carmen J Gil
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory School of Medicine , Atlanta , Georgia 30332 , United States
| | - Melissa P Lokugamage
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory School of Medicine , Atlanta , Georgia 30332 , United States
| | - Cory D Sago
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory School of Medicine , Atlanta , Georgia 30332 , United States
| | - Manaka Sato
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory School of Medicine , Atlanta , Georgia 30332 , United States
| | - Gwyn N Lando
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory School of Medicine , Atlanta , Georgia 30332 , United States
| | - Marielena Gamboa Castro
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory School of Medicine , Atlanta , Georgia 30332 , United States
| | - Anton V Bryksin
- Petit Institute for Bioengineering and Bioscience , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory School of Medicine , Atlanta , Georgia 30332 , United States
| |
Collapse
|
27
|
Decano JL, Aikawa M. Dynamic Macrophages: Understanding Mechanisms of Activation as Guide to Therapy for Atherosclerotic Vascular Disease. Front Cardiovasc Med 2018; 5:97. [PMID: 30123798 PMCID: PMC6086112 DOI: 10.3389/fcvm.2018.00097] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 07/02/2018] [Indexed: 12/13/2022] Open
Abstract
An emerging theory is that macrophages are heterogenous; an attribute that allows them to change behavior and execute specific functions in disease processes. This review aims to describe the current understanding on factors that govern their phenotypic changes, and provide insights for intervention beyond managing classical risk factors. Evidence suggests that metabolic reprogramming of macrophages triggers either a pro-inflammatory, anti-inflammatory or pro-resolving behavior. Dynamic changes in bioenergetics, metabolome or influence from bioactive lipids may promote resolution or aggravation of inflammation. Direct cell-to-cell interactions with other immune cells can also influence macrophage activation. Both paracrine signaling and intercellular molecular interactions either co-stimulate or co-inhibit activation of macrophages as well as their paired immune cell collaborator. More pathways of activation can even be uncovered by inspecting macrophages in the single cell level, since differential expression in key gene regulators can be screened in higher resolution compared to conventional averaged gene expression readouts. All these emerging macrophage activation mechanisms may be further explored and consolidated by using approaches in network biology. Integrating these insights can unravel novel and safer drug targets through better understanding of the pro-inflammatory activation circuitry.
Collapse
Affiliation(s)
- Julius L. Decano
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, MA, United States
- Channing Division of Network Medicine, Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
28
|
Khan OF, Kowalski PS, Doloff JC, Tsosie JK, Bakthavatchalu V, Winn CB, Haupt J, Jamiel M, Langer R, Anderson DG. Endothelial siRNA delivery in nonhuman primates using ionizable low-molecular weight polymeric nanoparticles. SCIENCE ADVANCES 2018; 4:eaar8409. [PMID: 29963629 PMCID: PMC6021147 DOI: 10.1126/sciadv.aar8409] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 05/18/2018] [Indexed: 05/19/2023]
Abstract
Dysfunctional endothelial cells contribute to the pathophysiology of many diseases, including vascular disease, stroke, hypertension, atherosclerosis, organ failure, diabetes, retinopathy, and cancer. Toward the goal of creating a new RNA-based therapy to correct aberrant endothelial cell gene expression in humans, efficient gene silencing in the endothelium of nonhuman primates was achieved by delivering small interfering RNA (siRNA) with 7C1, a low-molecular weight, ionizable polymer that forms nanoparticles. After a single intravenous administration of 1 mg of siRNA per kilogram of animal, 7C1 nanoparticles delivering Tie2 siRNA caused Tie2 mRNA levels to decrease by approximately 80% in the endothelium of the lung. Significant decreases in Tie2 mRNA were also found in the heart, retina, kidney, pancreas, and bone. Blood chemistry and liver function analysis before and after treatment all showed protein and enzyme concentrations within the normal reference ranges. Furthermore, after controlling for siRNA-specific effects, no significant increases in inflammatory cytokine concentrations were found in the serum. Similarly, no gross lesions or significant underlying pathologies were observed after histological examination of nonhuman primate tissues. This study is the first demonstration of endothelial gene silencing in multiple nonhuman primate organs using systemically administered siRNA nanoparticles and highlights the potential of this approach for the treatment of disease in humans.
Collapse
Affiliation(s)
- Omar F. Khan
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Piotr S. Kowalski
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Joshua C. Doloff
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Jonathan K. Tsosie
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
| | - Vasudevan Bakthavatchalu
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA 02139
| | - Caroline Bodi Winn
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA 02139
| | - Jennifer Haupt
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA 02139
| | - Morgan Jamiel
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA 02139
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Division of Health Science Technology, Massachusetts Institute of Technology, MA 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel G. Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, MA 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
- Department of Anesthesiology, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Division of Health Science Technology, Massachusetts Institute of Technology, MA 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Corresponding author.
| |
Collapse
|
29
|
Bai H, Guo J, Liu S, Guo X, Hu H, Wang T, Isaji T, Ono S, Yatsula B, Xing Y, Dardik A. Autologous tissue patches acquire vascular identity depending on the environment. VASCULAR INVESTIGATION AND THERAPY 2018; 1:14-23. [PMID: 31406962 DOI: 10.4103/vit.vit_9_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Vascular identity is genetically determined, but can be altered during surgical procedures. We hypothesized that the environment of the procedure critically alters the identity of autologous tissue patches implanted into the arterial or venous environment. Autologous jugular vein or carotid artery was used as a patch to repair a rat aorta or inferior vena cava. In the aortic environment patches contained neointimal cells that were CD34/Ephrin-B2-dual positive but not CD34/Eph-B4-dual positive; patches expressed Ephrin-B2, notch-4 and dll-4 but not Eph-B4 and COUP-TFII. In the venous environment patches contained neointimal cells that were CD34/Eph-B4-dual positive but not CD34/Ephrin-B2-dual positive; patches expressed Eph-B4 and COUP-TFII but not Ephrin-B2, notch-4 and dll-4. These data show that autologous tissue patches heal by acquisition of the vascular identity determined by the environment into which they are implanted, suggesting some plasticity of adult vascular identity.
Collapse
Affiliation(s)
- Hualong Bai
- Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China.,The Vascular Biology and Therapeutics Program and the Department of Surgery, Yale University School of Medicine, New Haven, CT, 06520 USA.,Department of Physiology, Basic Medical College of Zhengzhou University, Henan, China
| | - Jianming Guo
- The Vascular Biology and Therapeutics Program and the Department of Surgery, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Shirley Liu
- The Vascular Biology and Therapeutics Program and the Department of Surgery, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Xiangjiang Guo
- The Vascular Biology and Therapeutics Program and the Department of Surgery, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Haidi Hu
- The Vascular Biology and Therapeutics Program and the Department of Surgery, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Tun Wang
- The Vascular Biology and Therapeutics Program and the Department of Surgery, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Toshihiko Isaji
- The Vascular Biology and Therapeutics Program and the Department of Surgery, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Shun Ono
- The Vascular Biology and Therapeutics Program and the Department of Surgery, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Bogdan Yatsula
- The Vascular Biology and Therapeutics Program and the Department of Surgery, Yale University School of Medicine, New Haven, CT, 06520 USA
| | - Ying Xing
- Department of Physiology, Basic Medical College of Zhengzhou University, Henan, China
| | - Alan Dardik
- Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China.,Department of Surgery, VA Connecticut Healthcare System, West Haven, CT 06515 USA
| |
Collapse
|
30
|
Jones EA, Lehoux S. Shear stress, arterial identity and atherosclerosis. Thromb Haemost 2018; 115:467-73. [DOI: 10.1160/th15-10-0791] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 12/01/2015] [Indexed: 01/23/2023]
Abstract
SummaryIn the developing embryo, the vasculature first takes the form of a web-like network called the vascular plexus. Arterial and venous differentiation is subsequently guided by the specific expression of genes in the endothelial cells that provide spatial and temporal cues for development. Notch1/4, Notch ligand delta-like 4 (Dll4), and Notch downstream effectors are typically expressed in arterial cells along with EphrinB2, whereas chicken ovalbumin upstream promoter transcription factor II (COUP-TFII) and EphB4 characterise vein endothelial cells. Haemodynamic forces (blood pressure and blood flow) also contribute importantly to vascular remodelling. Early arteriovenous differentiation and local blood flow may hold the key to future inflammatory diseases. Indeed, despite the fact that atherosclerosis risk factors such as smoking, hypertension, hypercholesterolaemia, and diabetes all induce endothelial cell dysfunction throughout the vasculature, plaques develop only in arteries, and they localise essentially in vessel branch points, curvatures and bifurcations, where blood flow (and consequently shear stress) is low or oscillatory. Arterial segments exposed to high blood flow (and high laminar shear stress) tend to remain plaque-free. These observations have led many to investigate what particular properties of arterial or venous endothelial cells confer susceptibility or protection from plaque formation, and how that might interact with a particular shear stress environment.
Collapse
|
31
|
Paunovska K, Sago CD, Monaco CM, Hudson WH, Castro MG, Rudoltz TG, Kalathoor S, Vanover DA, Santangelo PJ, Ahmed R, Bryksin AV, Dahlman JE. A Direct Comparison of in Vitro and in Vivo Nucleic Acid Delivery Mediated by Hundreds of Nanoparticles Reveals a Weak Correlation. NANO LETTERS 2018; 18:2148-2157. [PMID: 29489381 PMCID: PMC6054134 DOI: 10.1021/acs.nanolett.8b00432] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Endothelial cells and macrophages play active roles in disease and as a result are important targets for nucleic acid therapies. While thousands of chemically distinct lipid nanoparticles (LNPs) can be synthesized to deliver nucleic acids, studying more than a few LNPs in vivo is challenging. As a result, it is difficult to understand how nanoparticles target these cells in vivo. Using high throughput LNP barcoding, we quantified how well LNPs delivered DNA barcodes to endothelial cells and macrophages in vitro, as well as endothelial cells and macrophages isolated from the lung, heart, and bone marrow in vivo. We focused on two fundamental questions in drug delivery. First, does in vitro LNP delivery predict in vivo LNP delivery? By comparing how 281 LNPs delivered barcodes to endothelial cells and macrophages in vitro and in vivo, we found in vitro delivery did not predict in vivo delivery. Second, does LNP delivery change within the microenvironment of a tissue? We quantified how 85 LNPs delivered barcodes to eight splenic cell populations, and found that cell types derived from myeloid progenitors tended to be targeted by similar LNPs, relative to cell types derived from lymphoid progenitors. These data demonstrate that barcoded LNPs can elucidate fundamental questions about in vivo nanoparticle delivery.
Collapse
Affiliation(s)
- Kalina Paunovska
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Cory D Sago
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Christopher M Monaco
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
- School of Biological Sciences , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - William H Hudson
- Emory Vaccine Center and Department of Microbiology and Immunology , Emory University School of Medicine , Atlanta , Georgia 30317 , United States
| | - Marielena Gamboa Castro
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Tobi G Rudoltz
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Sujay Kalathoor
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Daryll A Vanover
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Philip J Santangelo
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| | - Rafi Ahmed
- Emory Vaccine Center and Department of Microbiology and Immunology , Emory University School of Medicine , Atlanta , Georgia 30317 , United States
| | - Anton V Bryksin
- Parker H. Petit Institute for Bioengineering and Bioscience , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering , Georgia Institute of Technology and Emory University School of Medicine , Atlanta , Georgia 30332 , United States
| |
Collapse
|
32
|
Roostalu U, Wong JK. Arterial smooth muscle dynamics in development and repair. Dev Biol 2018; 435:109-121. [PMID: 29397877 DOI: 10.1016/j.ydbio.2018.01.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 01/08/2018] [Accepted: 01/24/2018] [Indexed: 12/11/2022]
Abstract
Arterial vasculature distributes blood from early embryonic development and provides a nutrient highway to maintain tissue viability. Atherosclerosis, peripheral artery diseases, stroke and aortic aneurysm represent the most frequent causes of death and are all directly related to abnormalities in the function of arteries. Vascular intervention techniques have been established for the treatment of all of these pathologies, yet arterial surgery can itself lead to biological changes in which uncontrolled arterial wall cell proliferation leads to restricted blood flow. In this review we describe the intricate cellular composition of arteries, demonstrating how a variety of distinct cell types in the vascular walls regulate the function of arteries. We provide an overview of the developmental origin of arteries and perivascular cells and focus on cellular dynamics in arterial repair. We summarize the current knowledge of the molecular signaling pathways that regulate vascular smooth muscle differentiation in the embryo and in arterial injury response. Our review aims to highlight the similarities as well as differences between cellular and molecular mechanisms that control arterial development and repair.
Collapse
Affiliation(s)
- Urmas Roostalu
- Manchester Academic Health Science Centre, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, UK.
| | - Jason Kf Wong
- Manchester Academic Health Science Centre, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, UK; Department of Plastic Surgery, Manchester University NHS Foundation Trust, Wythenshawe Hospital, Manchester, UK.
| |
Collapse
|
33
|
de Vries MR, Quax PHA. Inflammation in Vein Graft Disease. Front Cardiovasc Med 2018; 5:3. [PMID: 29417051 PMCID: PMC5787541 DOI: 10.3389/fcvm.2018.00003] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/08/2018] [Indexed: 12/23/2022] Open
Abstract
Bypass surgery is one of the most frequently used strategies to revascularize tissues downstream occlusive atherosclerotic lesions. For venous bypass surgery the great saphenous vein is the most commonly used vessel. Unfortunately, graft efficacy is low due to the development of vascular inflammation, intimal hyperplasia and accelerated atherosclerosis. Moreover, failure of grafts leads to significant adverse outcomes and even mortality. The last couple of decades not much has changed in the treatment of vein graft disease (VGD). However, insight is the cellular and molecular mechanisms of VGD has increased. In this review, we discuss the latest insights on VGD and the role of inflammation in this. We discuss vein graft pathophysiology including hemodynamic changes, the role of vessel wall constitutions and vascular remodeling. We show that profound systemic and local inflammatory responses, including inflammation of the perivascular fat, involve both the innate and adaptive immune system.
Collapse
Affiliation(s)
- Margreet R de Vries
- Department of Surgery, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Paul H A Quax
- Department of Surgery, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
34
|
Pagie S, Gérard N, Charreau B. Notch signaling triggered via the ligand DLL4 impedes M2 macrophage differentiation and promotes their apoptosis. Cell Commun Signal 2018; 16:4. [PMID: 29321062 PMCID: PMC5764024 DOI: 10.1186/s12964-017-0214-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 12/28/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Notch signaling controls many cellular processes, including cell fate determination, cell differentiation, proliferation and apoptosis. In mammals, four Notch receptors (Notch 1-4) can interact with five distinct ligands [Jagged1, Jagged2, Delta-like 1 (DLL1), DLL3, and DLL4]. We previously reported that Notch activation is modulated in endothelial cells and monocytes during inflammation and showed that inflammation upregulates DLL4 on endothelial cells. DLL4 promotes differentiation of blood monocytes into proinflammatory M1 macrophages. Here, we further investigated the ability of DLL4 to interfere with the polarization of blood monocytes into immunosuppressive M2 macrophages. METHODS Human blood monocytes were differentiated in vitro into M0 macrophages and then polarized into M1 or M2 macrophages with LPS/IFNγ and IL-4, respectively. Polarization steps were performed in the presence of immobilized recombinant DLL4. Immune phenotype and apoptosis of macrophage subsets were analyzed and quantified by flow cytometry. Regulatory effects of DLL4 on gene expression, cell signaling and apoptotic pathways were investigated by QPCR and western blots. RESULTS The phenotype of M2 macrophages was subject to specific alterations in the presence of recombinant DLL4. DLL4 inhibits the upregulation of IL-4 induced M2 markers such as CD11b, CD206, and CD200R. Survival of macrophages upon M2 polarization was also strongly reduced in the presence of DLL4. DLL4 induces a caspase3/7-dependent apoptosis during M2 but not M1 macrophage polarization. The Notch ligand DLL1 has no apoptotic effect. Both DLL4 signaling via Notch1 as well as DLL4-mediated apoptosis are Notch-dependent. Fully differentiated M2 macrophages became resistant to DLL4 action. Mechanistically, DLL4 selectively upregulates gene expression in macrophages upon M2 polarization, thereby affecting the Notch pattern (Notch1, 3, Jag1), activity (HES1), and transcription (IRF5, STAT1). The pro-apoptotic effectors Bax and Bak and the BH3-only proteins Bid and Bim seem to convey DLL4 apoptotic signal. CONCLUSION Interplay between the DLL4/Notch and IL-4/IL-4R signaling pathways impairs M2 differentiation. Thus, DLL4 may drive a Notch-dependent selection process not only by promoting M1 macrophage differentiation but also by preventing M2 macrophage differentiation through inhibition of M2-specific gene expression and apoptotic cell death.
Collapse
Affiliation(s)
- Sylvain Pagie
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, 30 bd J. Monnet, 44093, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Nathalie Gérard
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, 30 bd J. Monnet, 44093, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Béatrice Charreau
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, 30 bd J. Monnet, 44093, Nantes, France. .,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.
| |
Collapse
|
35
|
Katsuki S, Matoba T, Koga JI, Nakano K, Egashira K. Anti-inflammatory Nanomedicine for Cardiovascular Disease. Front Cardiovasc Med 2017; 4:87. [PMID: 29312961 PMCID: PMC5743792 DOI: 10.3389/fcvm.2017.00087] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/12/2017] [Indexed: 12/11/2022] Open
Abstract
Coronary artery disease, in the development of which inflammation mediated by innate immune cells plays a critical role, is one of the leading causes of death worldwide. The 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) are a widely used lipid-lowering drug that has lipid-independent vasculoprotective effects, such as improvement of endothelial dysfunction, antioxidant properties, and inhibitory effects on inflammation. Despite recent advances in lipid-lowering therapy, clinical trials of statins suggest that anti-inflammatory therapy beyond lipid-lowering therapy is indispensible to further reduce cardiovascular events. One possible therapeutic option to the residual risk is to directly intervene in the inflammatory process by utilizing a nanotechnology-based drug delivery system (nano-DDS). Various nano-sized materials are currently developed as DDS, including micelles, liposomes, polymeric nanoparticles, dendrimers, carbon nanotubes, and metallic nanoparticles. The application of nano-DDS to coronary artery disease is a feasible strategy since the inflammatory milieu enhances incorporation of nano-sized materials into mononuclear phagocytic system and permeability of target lesions, which confers nano-DDS on “passive-targeting” property. Recently, we have developed a polymeric nanoparticle-incorporating statin to maximize its anti-inflammatory property. This statin nanoparticle has been tested in various disease models, including plaque destabilization and rupture, myocardial ischemia-reperfusion injury, and ventricular remodeling after acute myocardial infarction, and its clinical application is in progress. In this review, we present current development of DDS and future perspective on the application of anti-inflammatory nanomedicine to treat life-threatening cardiovascular diseases.
Collapse
Affiliation(s)
- Shunsuke Katsuki
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan.,Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Tetsuya Matoba
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Jun-Ichiro Koga
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan.,Department of Cardiovascular Research, Development, and Translational Medicine, Center for Cardiovascular Disruptive Innovation, Kyushu University, Fukuoka, Japan
| | - Kaku Nakano
- Department of Cardiovascular Research, Development, and Translational Medicine, Center for Cardiovascular Disruptive Innovation, Kyushu University, Fukuoka, Japan
| | - Kensuke Egashira
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan.,Department of Cardiovascular Research, Development, and Translational Medicine, Center for Cardiovascular Disruptive Innovation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
36
|
Stassen OMJA, Muylaert DEP, Bouten CVC, Hjortnaes J. Current Challenges in Translating Tissue-Engineered Heart Valves. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2017; 19:71. [PMID: 28782083 PMCID: PMC5545463 DOI: 10.1007/s11936-017-0566-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Heart valve disease is a major health burden, treated by either valve repair or valve replacement, depending on the affected valve. Nearly 300,000 valve replacements are performed worldwide per year. Valve replacement is lifesaving, but not without complications. The in situ tissue-engineered heart valve is a promising alternative to current treatments, but the translation of this novel technology to the clinic still faces several challenges. These challenges originate from the variety encountered in the patient population, the conversion of an implant into a living tissue, the highly mechanical nature of the heart valve, the complex homeostatic tissue that has to be reached at the end stage of the regenerating heart valve, and all the biomaterial properties that can be controlled to obtain this tissue. Many of these challenges are multidimensional and multiscalar, and both the macroscopic properties of the complete heart valve and the microscopic properties of the patient’s cells interacting with the materials have to be optimal. Using newly developed in vitro models, or bioreactors, where variables of interest can be controlled tightly and complex mixtures of cell populations similar to those encountered in the regenerating valve can be cultured, it is likely that the challenges can be overcome.
Collapse
Affiliation(s)
- O M J A Stassen
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands.
| | - D E P Muylaert
- Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - C V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - J Hjortnaes
- Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, The Netherlands.,Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
37
|
Tian DY, Jin XR, Zeng X, Wang Y. Notch Signaling in Endothelial Cells: Is It the Therapeutic Target for Vascular Neointimal Hyperplasia? Int J Mol Sci 2017; 18:ijms18081615. [PMID: 28757591 PMCID: PMC5578007 DOI: 10.3390/ijms18081615] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 07/05/2017] [Accepted: 07/21/2017] [Indexed: 01/09/2023] Open
Abstract
Blood vessels respond to injury through a healing process that includes neointimal hyperplasia. The vascular endothelium is a monolayer of cells that separates the outer vascular wall from the inner circulating blood. The disruption and exposure of endothelial cells (ECs) to subintimal components initiate the neointimal formation. ECs not only act as a highly selective barrier to prevent early pathological changes of neointimal hyperplasia, but also synthesize and release molecules to maintain vascular homeostasis. After vascular injury, ECs exhibit varied responses, including proliferation, regeneration, apoptosis, phenotypic switching, interacting with other cells by direct contact or secreted molecules and the change of barrier function. This brief review presents the functional role of the evolutionarily-conserved Notch pathway in neointimal hyperplasia, notably by regulating endothelial cell functions (proliferation, regeneration, apoptosis, differentiation, cell-cell interaction). Understanding endothelial cell biology should help us define methods to prompt cell proliferation, prevent cell apoptosis and dysfunction, block neointimal hyperplasia and vessel narrowing.
Collapse
Affiliation(s)
- Ding-Yuan Tian
- Trainee Brigade, Third Military Medical University, Chongqing 400038, China.
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China.
| | - Xu-Rui Jin
- Trainee Brigade, Third Military Medical University, Chongqing 400038, China.
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China.
| | - Xi Zeng
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China.
| | - Yun Wang
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
38
|
Nakano T, Fukuda D, Koga JI, Aikawa M. Delta-Like Ligand 4-Notch Signaling in Macrophage Activation. Arterioscler Thromb Vasc Biol 2016; 36:2038-47. [PMID: 27562914 PMCID: PMC5033717 DOI: 10.1161/atvbaha.116.306926] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 08/09/2016] [Indexed: 12/20/2022]
Abstract
The Notch signaling pathway regulates the development of various cell types and organs, and also contributes to disease mechanisms in adults. Accumulating evidence suggests its role in cardiovascular and metabolic diseases. Notch signaling components also control the phenotype of immune cells. Delta-like ligand 4 (Dll4) of the Notch pathway promotes proinflammatory activation of macrophages in vitro and in vivo. Dll4 blockade attenuates chronic atherosclerosis, vein graft disease, vascular calcification, insulin resistance, and fatty liver in mice. The Dll4-Notch axis may, thus, participate in the shared mechanisms for cardiometabolic disorders, serving as a potential therapeutic target for ameliorating these global health problems.
Collapse
Affiliation(s)
- Toshiaki Nakano
- From The Center for Excellence in Vascular Biology (T.N., D.F., J.K., M.A.), The Center for Interdisciplinary Cardiovascular Sciences (M.A.), Cardiovascular Division (T.N., D.F., J.K., M.A.), and Channing Division of Network Medicine (M.A.), Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Daiju Fukuda
- From The Center for Excellence in Vascular Biology (T.N., D.F., J.K., M.A.), The Center for Interdisciplinary Cardiovascular Sciences (M.A.), Cardiovascular Division (T.N., D.F., J.K., M.A.), and Channing Division of Network Medicine (M.A.), Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Jun-Ichiro Koga
- From The Center for Excellence in Vascular Biology (T.N., D.F., J.K., M.A.), The Center for Interdisciplinary Cardiovascular Sciences (M.A.), Cardiovascular Division (T.N., D.F., J.K., M.A.), and Channing Division of Network Medicine (M.A.), Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Masanori Aikawa
- From The Center for Excellence in Vascular Biology (T.N., D.F., J.K., M.A.), The Center for Interdisciplinary Cardiovascular Sciences (M.A.), Cardiovascular Division (T.N., D.F., J.K., M.A.), and Channing Division of Network Medicine (M.A.), Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW Notch signaling is an evolutionary conserved pathway critical for cardiovascular development and angiogenesis. More recently, the contribution of Notch signaling to the homeostasis of the adult vasculature has emerged as an important novel paradigm, but much remains to be understood. RECENT FINDINGS Recent findings shed light on the impact of Notch in vascular and immune responses to microenvironmental signals as well as on the onset of atherosclerosis. In the past year, studies in human and mice explored the role of Notch in the maintenance of a nonactivated endothelium. Novel pieces of evidence suggest that this pathway is sensitive to environmental factors, including inflammatory mediators and diet-derived by-products. SUMMARY An emerging theme is the ability of Notch to respond to changes in the microenvironment, including glucose and lipid metabolites. In turn, alterations in Notch enable an important link between metabolism and transcriptional changes, thus this receptor appears to function as a metabolic sensor with direct implications to gene expression.
Collapse
Affiliation(s)
- Anaïs Briot
- I2MC, Institute of Metabolic and Cardiovascular Diseases, Université de Toulouse, INSERM, Team 1, Toulouse, France
| | - Anne Bouloumié
- I2MC, Institute of Metabolic and Cardiovascular Diseases, Université de Toulouse, INSERM, Team 1, Toulouse, France
| | - M. Luisa Iruela-Arispe
- Department of Molecular, Cell, and Developmental Biology; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
40
|
Cai Z, Zhao B, Deng Y, Shangguan S, Zhou F, Zhou W, Li X, Li Y, Chen G. Notch signaling in cerebrovascular diseases (Review). Mol Med Rep 2016; 14:2883-98. [PMID: 27574001 PMCID: PMC5042775 DOI: 10.3892/mmr.2016.5641] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 07/22/2016] [Indexed: 12/30/2022] Open
Abstract
The Notch signaling pathway is a crucial regulator of numerous fundamental cellular processes. Increasing evidence suggests that Notch signaling is involved in inflammation and oxidative stress, and thus in the progress of cerebrovascular diseases. In addition, Notch signaling in cerebrovascular diseases is associated with apoptosis, angiogenesis and the function of blood-brain barrier. Despite the contradictory results obtained to date as to whether Notch signaling is harmful or beneficial, the regulation of Notch signaling may provide a novel strategy for the treatment of cerebrovascular diseases.
Collapse
Affiliation(s)
- Zhiyou Cai
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Bin Zhao
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Yanqing Deng
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Shouqin Shangguan
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Faming Zhou
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Wenqing Zhou
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Xiaoli Li
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Yanfeng Li
- Department of Neurology, Peking Union Medical College Hospital, Beijing 100730, P.R. China
| | - Guanghui Chen
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| |
Collapse
|
41
|
de Vries MR, Simons KH, Jukema JW, Braun J, Quax PHA. Vein graft failure: from pathophysiology to clinical outcomes. Nat Rev Cardiol 2016; 13:451-70. [PMID: 27194091 DOI: 10.1038/nrcardio.2016.76] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Occlusive arterial disease is a leading cause of morbidity and mortality worldwide. Aside from balloon angioplasty, bypass graft surgery is the most commonly performed revascularization technique for occlusive arterial disease. Coronary artery bypass graft surgery is performed in patients with left main coronary artery disease and three-vessel coronary disease, whereas peripheral artery bypass graft surgery is used to treat patients with late-stage peripheral artery occlusive disease. The great saphenous veins are commonly used conduits for surgical revascularization; however, they are associated with a high failure rate. Therefore, preservation of vein graft patency is essential for long-term surgical success. With the exception of 'no-touch' techniques and lipid-lowering and antiplatelet (aspirin) therapy, no intervention has hitherto unequivocally proven to be clinically effective in preventing vein graft failure. In this Review, we describe both preclinical and clinical studies evaluating the pathophysiology underlying vein graft failure, and the latest therapeutic options to improve patency for both coronary and peripheral grafts.
Collapse
Affiliation(s)
- Margreet R de Vries
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Karin H Simons
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - J Wouter Jukema
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands.,Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Jerry Braun
- Department of Cardiothoracic Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Paul H A Quax
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| |
Collapse
|
42
|
Decano JL, Mattson PC, Aikawa M. Macrophages in Vascular Inflammation: Origins and Functions. Curr Atheroscler Rep 2016; 18:34. [PMID: 27125207 DOI: 10.1007/s11883-016-0585-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
43
|
Koga JI, Matoba T, Egashira K. Anti-inflammatory Nanoparticle for Prevention of Atherosclerotic Vascular Diseases. J Atheroscler Thromb 2016; 23:757-65. [PMID: 27108537 DOI: 10.5551/jat.35113] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Recent technical innovation has enabled chemical modifications of small materials and various kinds of nanoparticles have been created. In clinical settings, nanoparticle-mediated drug delivery systems have been used in the field of cancer care to deliver therapeutic agents specifically to cancer tissues and to enhance the efficacy of drugs by gradually releasing their contents. In addition, nanotechnology has enabled the visualization of various molecular processes by targeting proteinases or inflammation. Nanoparticles that consist of poly (lactic-co-glycolic) acid (PLGA) deliver therapeutic agents to monocytes/macrophages and function as anti-inflammatory nanoparticles in combination with statins, angiotensin receptor antagonists, or agonists of peroxisome proliferator-activated receptor-γ (PPARγ). PLGA nanoparticle-mediated delivery of pitavastatin has been shown to prevent inflammation and ameliorated features associated with plaque ruptures in hyperlipidemic mice. PLGA nanoparticles were also delivered to tissues with increased vascular permeability and nanoparticles incorporating pitavastatin, injected intramuscularly, were retained in ischemic tissues and induced therapeutic arteriogenesis. This resulted in attenuation of hind limb ischemia. Ex vivo treatment of vein grafts with imatinib nanoparticles before graft implantation has been demonstrated to inhibit lesion development. These results suggest that nanoparticle-mediated drug delivery system can be a promising strategy as a next generation therapy for atherosclerotic vascular diseases.
Collapse
Affiliation(s)
- Jun-Ichiro Koga
- The Department of Cardiovascular Research, Development, and Translational Medicine, Center for Disruptive Cardiovascular Medicine, Kyushu University
| | | | | |
Collapse
|
44
|
Hashimoto T, Tsuneki M, Foster TR, Santana JM, Bai H, Wang M, Hu H, Hanisch JJ, Dardik A. Membrane-mediated regulation of vascular identity. BIRTH DEFECTS RESEARCH. PART C, EMBRYO TODAY : REVIEWS 2016; 108:65-84. [PMID: 26992081 PMCID: PMC5310768 DOI: 10.1002/bdrc.21123] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 02/22/2016] [Indexed: 02/06/2023]
Abstract
Vascular diseases span diverse pathology, but frequently arise from aberrant signaling attributed to specific membrane-associated molecules, particularly the Eph-ephrin family. Originally recognized as markers of embryonic vessel identity, Eph receptors and their membrane-associated ligands, ephrins, are now known to have a range of vital functions in vascular physiology. Interactions of Ephs with ephrins at cell-to-cell interfaces promote a variety of cellular responses such as repulsion, adhesion, attraction, and migration, and frequently occur during organ development, including vessel formation. Elaborate coordination of Eph- and ephrin-related signaling among different cell populations is required for proper formation of the embryonic vessel network. There is growing evidence supporting the idea that Eph and ephrin proteins also have postnatal interactions with a number of other membrane-associated signal transduction pathways, coordinating translation of environmental signals into cells. This article provides an overview of membrane-bound signaling mechanisms that define vascular identity in both the embryo and the adult, focusing on Eph- and ephrin-related signaling. We also discuss the role and clinical significance of this signaling system in normal organ development, neoplasms, and vascular pathologies.
Collapse
Affiliation(s)
- Takuya Hashimoto
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, Connecticut
- Department of Vascular Surgery, The University of Tokyo, Tokyo, Japan
| | - Masayuki Tsuneki
- Division of Cancer Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Trenton R. Foster
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
| | - Jeans M. Santana
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
| | - Hualong Bai
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
- Department of Vascular Surgery, The 1st Affiliated Hospital of Zhengzhou University, Henan, China
| | - Mo Wang
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
| | - Haidi Hu
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
| | - Jesse J. Hanisch
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
| | - Alan Dardik
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, Connecticut
| |
Collapse
|