1
|
Jiao J, Lu L, Yin M, Wang T, Wang Z, Zhu Y, Zhao J, Wang X, Hu B, Dai F, Xue L, Qiao J, Liu Y, Chen Q. A multifunctional biosensor for linked monitoring of inflammation indicators in hypertension drug evaluation and companion diagnostics. Talanta 2025; 291:127882. [PMID: 40056652 DOI: 10.1016/j.talanta.2025.127882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/20/2025] [Accepted: 03/01/2025] [Indexed: 03/10/2025]
Abstract
Hypertension, often called the "silent killer", is a prevalent chronic disease closely linked to inflammation. However, most current methods monitor only single indicator, providing a limited view of inflammation in hypertension progression. To address this, we developed a multifunctional biosensor featuring a dual target linked monitoring (DTLM) Probe for the simultaneous detection of IL-6 and CRP, two key inflammatory markers in hypertension progression. The DTLM Probe, based on NH2-UiO-66@AuNPs with mutually non-interfering signal chains, was optimized for high performance in tracking both indicators simultaneously. The dual outputs operate independently, enabling IL-6 and CRP to be detected together or individually within a single sample injection. Under optimized conditions, the biosensor demonstrated excellent specificity and sensitivity, with detection limits of 355 fg/mL for IL-6 and 367 fg/mL for CRP. Applied to a rat model, the biosensor effectively explored the anti-inflammatory effects of Qishenyiqi, a traditional Chinese medicine, assessing its efficacy in reducing hypertensive heart damage. Additionally, it distinguished IL-6 and CRP levels between healthy and hypertensive individuals, capturing subtle changes after treatments. This ensured targeted anti-inflammatory therapies for patients who would benefit most. This biosensor provides a powerful and versatile platform for dual markers tracking, supporting both drug evaluation and companion diagnostics for tailor treatments in hypertension management.
Collapse
Affiliation(s)
- Jun Jiao
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China.
| | - Lina Lu
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China
| | - Mengai Yin
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China
| | - Tong Wang
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China
| | - Zhijie Wang
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China
| | - Yu Zhu
- The Third Central Hospital of Tianjin, The Central Hospital of Tianjin University, Tianjin, 300170, PR China
| | - Jie Zhao
- Tianjin Hospital, Tianjin University, Tianjin, 300211, PR China
| | - Xiangrui Wang
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China
| | - Bingxin Hu
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China
| | - Fuju Dai
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China
| | - Lan Xue
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China
| | - Jiaxuan Qiao
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China
| | - Yue Liu
- Nankai University Affiliated Tianjin People's Hospital, Tianjin, 300192, PR China.
| | - Qiang Chen
- The Key Laboratory of Bioactive Materials Ministry of Education, College of Life Science, Nankai University, Weijin Road No.94, Tianjin, 300071, PR China.
| |
Collapse
|
2
|
Zhang Z, Wang X, Huang C, Wang M, Cui W, Hao L, Yang R, Wang HH, Zhang X. Size-dependent interactions between calciprotein particles and vascular endothelium. Mater Today Bio 2025; 31:101599. [PMID: 40070870 PMCID: PMC11894339 DOI: 10.1016/j.mtbio.2025.101599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
The underlying mechanisms governing the interactions between nanoparticles and vascular endothelial barrier remain largely unexplored, which is crucial for the optimal design of nanoparticles for clinical applications. In this study, the size-dependent interactions between calciprotein particles (CPPs) and endothelial cells (ECs) were investigated using a rat model of chronic kidney disease (CKD) induced by 5/6 nephrectomy. Two primary types of CPP1 were studied: small-sized CPP1 (S-CPP1, <50 nm) and larger CPP1 (L-CPP1, <100 nm), detected three and five weeks post-surgery, respectively. By adjusting the amounts of Ca2+, HPO4 2- and H2PO4 - ions in Dulbecco's Modified Eagle Medium supplemented with 10 % (V/V) fetal bovine serum and 1 % (V/V) Pen-Strep solution, S-CPP1 (<50 nm) with an elliptical shape, L-CPP1 (50-100 nm), and secondary CPPs (CPP2, >100 nm) with a needle-like crystalline structure, resembling endogenous CPPs, were synthesized. The results showed that S-CPP1 significantly increased endothelial permeability at concentrations of 445 μg/mL and 890 μg/mL, thereby disrupting the integrity of the endothelial barrier formed by a confluent monolayer of ECs. Immunofluorescence analysis revealed that L-CPP1 was internalized by ECs via endocytosis, while S-CPP1 disrupted VE-cadherin junctions, leading to irregular cell morphology and widened intercellular gaps. These structural changes likely contribute to medial calcification as CPPs accumulate within the circulatory system. In conclusion, the findings underscore that the interaction between CPPs and the vascular endothelium is strongly size-dependent, with significant implications for vascular system health and the design of nanoparticle-based therapies.
Collapse
Affiliation(s)
- Zeping Zhang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning 110016, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xinyue Wang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning 110016, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Caihao Huang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning 110016, China
- School of Materials Science and Engineering, Dalian University of Technology, Dalian, Liaoning 116024, China
| | - Meixia Wang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Wei Cui
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning 110016, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Liang Hao
- School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110026, China
| | - Rui Yang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning 110016, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Hong-hui Wang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Xing Zhang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning 110016, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
3
|
Toader C, Radoi MP, Covlea CA, Covache-Busuioc RA, Ilie MM, Glavan LA, Corlatescu AD, Costin HP, Gica MD, Dobrin N. Cerebral Aneurysm: Filling the Gap Between Pathophysiology and Nanocarriers. Int J Mol Sci 2024; 25:11874. [PMID: 39595942 PMCID: PMC11593836 DOI: 10.3390/ijms252211874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Intracranial aneurysms, characterized by abnormal dilations of cerebral arteries, pose significant health risks due to their potential to rupture, leading to subarachnoid hemorrhage with high mortality and morbidity rates. This paper aim is to explore the innovative application of nanoparticles in treating intracranial aneurysms, offering a promising avenue for enhancing current therapeutic strategies. We took into consideration the pathophysiology of cerebral aneurysms, focusing on the role of hemodynamic stress, endothelial dysfunction, and inflammation in their development and progression. By comparing cerebral aneurysms with other types, such as aortic aneurysms, we identify pathophysiological similarities and differences that could guide the adaptation of treatment approaches. The review highlights the potential of nanoparticles to improve drug delivery, targeting, and efficacy while minimizing side effects. We discuss various nanocarriers, including liposomes and polymeric nanoparticles, and their roles in overcoming biological barriers and enhancing therapeutic outcomes. Additionally, we discuss the potential of specific compounds, such as Edaravone and Tanshinone IIA, when used in conjunction with nanocarriers, to provide neuroprotective and anti-inflammatory benefits. By extrapolating insights from studies on aortic aneurysms, new research directions and therapeutic strategies for cerebral aneurysms are proposed. This interdisciplinary approach underscores the potential of nanoparticles to positively influence the management of intracranial aneurysms, paving the way for personalized treatment options that could significantly improve patient outcomes.
Collapse
Affiliation(s)
- Corneliu Toader
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Mugurel Petrinel Radoi
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Christian-Adelin Covlea
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | - Razvan-Adrian Covache-Busuioc
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | - Milena Monica Ilie
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | - Luca-Andrei Glavan
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | - Antonio-Daniel Corlatescu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | - Horia-Petre Costin
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | - Maria-Daria Gica
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.T.); (C.-A.C.); (R.-A.C.-B.); (M.M.I.); (L.-A.G.); (A.-D.C.); (H.-P.C.); (M.-D.G.)
| | | |
Collapse
|
4
|
Cong X, Zhang Z, Li H, Yang YG, Zhang Y, Sun T. Nanocarriers for targeted drug delivery in the vascular system: focus on endothelium. J Nanobiotechnology 2024; 22:620. [PMID: 39396002 PMCID: PMC11470712 DOI: 10.1186/s12951-024-02892-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/01/2024] [Indexed: 10/14/2024] Open
Abstract
Endothelial cells (ECs) are pivotal in maintaining vascular health, regulating hemodynamics, and modulating inflammatory responses. Nanocarriers hold transformative potential for precise drug delivery within the vascular system, particularly targeting ECs for therapeutic purposes. However, the complex interactions between vascular ECs and nanocarriers present significant challenges for the development and clinical translation of nanotherapeutics. This review assesses recent advancements and key strategies in employing nanocarriers for drug delivery to vascular ECs. It suggested that through precise physicochemical design and surface modifications, nanocarriers can enhance targeting specificity and improve drug internalization efficiency in ECs. Additionally, we elaborated on the applications of nanocarriers specifically designed for targeting ECs in the treatment of cardiovascular diseases, cancer metastasis, and inflammatory disorders. Despite these advancements, safety concerns, the complexity of in vivo processes, and the challenge of achieving subcellular drug delivery remain significant obstacles to the effective targeting of ECs with nanocarriers. A comprehensive understanding of endothelial cell biology and its interaction with nanocarriers is crucial for realizing the full potential of targeted drug delivery systems.
Collapse
Affiliation(s)
- Xiuxiu Cong
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China
| | - Zebin Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China
| | - He Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China
- International Center of Future Science, Jilin University, Changchun, 130015, Jilin, China
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100143, China
| | - Yuning Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China.
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, 130061, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, 130062, Jilin, China.
- International Center of Future Science, Jilin University, Changchun, 130015, Jilin, China.
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, 130012, Jilin, China.
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100143, China.
| |
Collapse
|
5
|
Zu HL, Zhuang PP, Peng Y, Peng C, Peng C, Zhu ZJ, Yao Y, Yue J, Wang QS, Zhou WH, Wang HY. Dual-Drug Nanomedicine Assembly with Synergistic Anti-Aneurysmal Effects via Inflammation Suppression and Extracellular Matrix Stabilization. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402141. [PMID: 38953313 DOI: 10.1002/smll.202402141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/03/2024] [Indexed: 07/04/2024]
Abstract
Abdominal aortic aneurysm (AAA) represents a critical cardiovascular condition characterized by localized dilation of the abdominal aorta, carrying a significant risk of rupture and mortality. Current treatment options are limited, necessitating novel therapeutic approaches. This study investigates the potential of a pioneering nanodrug delivery system, RAP@PFB, in mitigating AAA progression. RAP@PFB integrates pentagalloyl glucose (PGG) and rapamycin (RAP) within a metal-organic-framework (MOF) structure through a facile assembly process, ensuring remarkable drug loading capacity and colloidal stability. The synergistic effects of PGG, a polyphenolic antioxidant, and RAP, an mTOR inhibitor, collectively regulate key players in AAA pathogenesis, such as macrophages and smooth muscle cells (SMCs). In macrophages, RAP@PFB efficiently scavenges various free radicals, suppresses inflammation, and promotes M1-to-M2 phenotype repolarization. In SMCs, it inhibits apoptosis and calcification, thereby stabilizing the extracellular matrix and reducing the risk of AAA rupture. Administered intravenously, RAP@PFB exhibits effective accumulation at the AAA site, demonstrating robust efficacy in reducing AAA progression through multiple mechanisms. Moreover, RAP@PFB demonstrates favorable biosafety profiles, supporting its potential translation into clinical applications for AAA therapy.
Collapse
Affiliation(s)
- Hong Lin Zu
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Pei Pei Zhuang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Ying Peng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Chao Peng
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Cheng Peng
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Zi Jia Zhu
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Ye Yao
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Jie Yue
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Qing Shan Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Wen Hu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Hai Yang Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| |
Collapse
|
6
|
Zamora ME, Essien EO, Bhamidipati K, Murthy A, Liu J, Kim H, Patel MN, Nong J, Wang Z, Espy C, Chaudhry FN, Ferguson LT, Tiwari S, Hood ED, Marcos-Contreras OA, Omo-Lamai S, Shuvaeva T, Arguiri E, Wu J, Rauova L, Poncz M, Basil MC, Cantu E, Planer JD, Spiller K, Zepp J, Muzykantov VR, Myerson JW, Brenner JS. Marginated Neutrophils in the Lungs Effectively Compete for Nanoparticles Targeted to the Endothelium, Serving as a Part of the Reticuloendothelial System. ACS NANO 2024; 18:22275-22297. [PMID: 39105696 PMCID: PMC11935960 DOI: 10.1021/acsnano.4c06286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
Nanomedicine has long pursued the goal of targeted delivery to specific organs and cell types but has yet to achieve this goal with the vast majority of targets. One rare example of success in this pursuit has been the 25+ years of studies targeting the lung endothelium using nanoparticles conjugated to antibodies against endothelial surface molecules. However, here we show that such "endothelial-targeted" nanocarriers also effectively target the lungs' numerous marginated neutrophils, which reside in the pulmonary capillaries and patrol for pathogens. We show that marginated neutrophils' uptake of many of these "endothelial-targeted" nanocarriers is on par with endothelial uptake. This generalizes across diverse nanomaterials and targeting moieties and was even found with physicochemical lung tropism (i.e., without targeting moieties). Further, we observed this in ex vivo human lungs and in vivo healthy mice, with an increase in marginated neutrophil uptake of nanoparticles caused by local or distant inflammation. These findings have implications for nanomedicine development for lung diseases. These data also suggest that marginated neutrophils, especially in the lungs, should be considered a major part of the reticuloendothelial system (RES), with a special role in clearing nanoparticles that adhere to the lumenal surfaces of blood vessels.
Collapse
Affiliation(s)
- Marco E Zamora
- Drexel University School of Biomedical Engineering, Philadelphia, Pennsylvania 19104, United States
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Eno-Obong Essien
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Kartik Bhamidipati
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Aditi Murthy
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Jing Liu
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Hyunjun Kim
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Manthan N Patel
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jia Nong
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Zhicheng Wang
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Carolann Espy
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Fatima N Chaudhry
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Laura T Ferguson
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Sachchidanand Tiwari
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Elizabeth D Hood
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Oscar A Marcos-Contreras
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Serena Omo-Lamai
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Tea Shuvaeva
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Evguenia Arguiri
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jichuan Wu
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Lubica Rauova
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Mortimer Poncz
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Maria C Basil
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Edward Cantu
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Joseph D Planer
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| | - Kara Spiller
- Drexel University School of Biomedical Engineering, Philadelphia, Pennsylvania 19104, United States
| | - Jarod Zepp
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Vladimir R Muzykantov
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jacob W Myerson
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jacob S Brenner
- Perelman School of Medicine Department of System Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
- Perelman School of Medicine Department of Pulmonary, Allergy, and Critical Care, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
7
|
Wang J, Zhang H, Wan W, Yang H, Zhao J. Advances in nanotechnological approaches for the detection of early markers associated with severe cardiac ailments. Nanomedicine (Lond) 2024; 19:1487-1506. [PMID: 39121377 PMCID: PMC11318751 DOI: 10.1080/17435889.2024.2364581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/31/2024] [Indexed: 08/11/2024] Open
Abstract
Mortality from cardiovascular disease (CVD) accounts for over 30% of all deaths globally, necessitating reliable diagnostic tools. Prompt identification and precise diagnosis are critical for effective personalized treatment. Nanotechnology offers promising applications in diagnostics, biosensing and drug delivery for prevalent cardiovascular diseases. Its integration into cardiovascular care enhances diagnostic accuracy, enabling early intervention and tailored treatment plans. By leveraging nanoscale innovations, healthcare professionals can address the complexities of CVD progression and customize interventions based on individual patient needs. Ongoing advancements in nanotechnology continue to shape the landscape of cardiovascular medicine, offering potential for improved patient outcomes and reduced mortality rates from these pervasive diseases.
Collapse
Affiliation(s)
- Jie Wang
- Department of Cardiac Care Unit, Yantaishan Hospital, Yantai, Shandong, 264003, China
| | - Haifeng Zhang
- Department of Cardiology, Yantai Yeda Hospital, Yantai, Shangdong, 264006, China
| | - Weiping Wan
- Department of Ultrasound, Yantaishan Hospital, Yantai, Shandong, 264003, China
| | - Haijiao Yang
- Department of Cardiac Care Unit, Yantaishan Hospital, Yantai, Shandong, 264003, China
| | - Jing Zhao
- Department of Critical Care Medicine, Yantaishan Hospital, Yantai, Shandong, 264003, China
| |
Collapse
|
8
|
Omo-Lamai S, Nong J, Savalia K, Kelley BJ, Wu J, Esteves-Reyes S, Chase LS, Muzykantov VR, Marcos-Contreras OA, Dollé JP, Smith DH, Brenner JS. Targeting of nanoparticles to the cerebral vasculature after traumatic brain injury. PLoS One 2024; 19:e0297451. [PMID: 38857220 PMCID: PMC11164327 DOI: 10.1371/journal.pone.0297451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 01/04/2024] [Indexed: 06/12/2024] Open
Abstract
Traumatic brain injury has faced numerous challenges in drug development, primarily due to the difficulty of effectively delivering drugs to the brain. However, there is a potential solution in targeted drug delivery methods involving antibody-drug conjugates or nanocarriers conjugated with targeting antibodies. Following a TBI, the blood-brain barrier (BBB) becomes permeable, which can last for years and allow the leakage of harmful plasma proteins. Consequently, an appealing approach for TBI treatment involves using drug delivery systems that utilize targeting antibodies and nanocarriers to help restore BBB integrity. In our investigation of this strategy, we examined the efficacy of free antibodies and nanocarriers targeting a specific endothelial surface marker called vascular cell adhesion molecule-1 (VCAM-1), which is known to be upregulated during inflammation. In a mouse model of TBI utilizing central fluid percussion injury, free VCAM-1 antibody did not demonstrate superior targeting when comparing sham vs. TBI brain. However, the administration of VCAM-1-targeted nanocarriers (liposomes) exhibited a 10-fold higher targeting specificity in TBI brain than in sham control. Flow cytometry and confocal microscopy analysis confirmed that VCAM-1 liposomes were primarily taken up by brain endothelial cells post-TBI. Consequently, VCAM-1 liposomes represent a promising platform for the targeted delivery of therapeutics to the brain following traumatic brain injury.
Collapse
Affiliation(s)
- Serena Omo-Lamai
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jia Nong
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Krupa Savalia
- Departments of Neurology & Neurological Surgery, University of California—Davis, Sacramento, California, United States of America
| | - Brian J. Kelley
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jichuan Wu
- Department of Medicine, Division of Pulmonary Allergy and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Sahily Esteves-Reyes
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Liam S. Chase
- Department of Medicine, Division of Pulmonary Allergy and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Vladimir R. Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Oscar A. Marcos-Contreras
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jean-Pierre Dollé
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Douglas H. Smith
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jacob S. Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Medicine, Division of Pulmonary Allergy and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
9
|
Jerez HE, Simioni YR, Ghosal K, Morilla MJ, Romero EL. Cholesterol nanoarchaeosomes for alendronate targeted delivery as an anti-endothelial dysfunction agent. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2024; 15:517-534. [PMID: 38774586 PMCID: PMC11106671 DOI: 10.3762/bjnano.15.46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/16/2024] [Indexed: 05/24/2024]
Abstract
Sodium alendronate (ALN) is a very hydrosoluble and poorly permeable molecule used as an antiresorptive agent and with vascular anticalcifying capacity. Loaded into targeted nanovesicles, its anti-inflammatory activity may be amplified towards extra-osseous and noncalcified target cells, such as severely irritated vascular endothelium. Here cytotoxicity, mitochondrial membrane potential, ATP content, and membrane fluidity of human endothelial venous cells (HUVECs) were determined after endocytosis of ALN-loaded nanoarchaeosomes (nanoARC-Chol(ALN), made of polar lipids from Halorubrum tebenquichense: cholesterol 7:3 w/w, 166 ± 5 nm, 0.16 ± 0.02 PDI, -40.8 ± 5.4 mV potential, 84.7 ± 21 µg/mg ALN/total lipids, TL). The effect of nanoARC-Chol(ALN) was further assessed on severely inflamed HUVECs. To that aim, HUVECs were grown on a porous barrier on top of a basal compartment seeded either with macrophages or human foam cells. One lighter and one more pronounced inflammatory context was modelled by adding lipopolysaccharide (LPS) to the apical or the apical and basal compartments. The endocytosis of nanoARC-Chol(ALN), was observed to partly reduce the endothelial-mesenchymal transition of HUVECs. Besides, while 10 mg/mL dexamethasone, 7.6 mM free ALN and ALN-loaded liposomes failed, 50 μg/mL TL + 2.5 μg/mL ALN (i.e., nanoARC-Chol(ALN)) reduced the IL-6 and IL-8 levels by, respectively, 75% and 65% in the mild and by, respectively, 60% and 40% in the pronounced inflammation model. This is the first report showing that the endocytosis of nanoARC-Chol(ALN) by HUVECs magnifies the anti-inflammatory activity of ALN even under conditions of intense irritation, not only surpassing that of free ALN but also that of dexamethasone.
Collapse
Affiliation(s)
- Horacio Emanuel Jerez
- Nanomedicine Research and Development Centre (NARD), Science and Technology Department, National University of Quilmes, Roque Sáenz Peña 352, Bernal, Buenos Aires, Argentina
| | - Yamila Roxana Simioni
- Nanomedicine Research and Development Centre (NARD), Science and Technology Department, National University of Quilmes, Roque Sáenz Peña 352, Bernal, Buenos Aires, Argentina
| | - Kajal Ghosal
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja Subodh Chandra Mallick Rd., Jadavpur, Kolkata 700032, West Bengal, India
| | - Maria Jose Morilla
- Nanomedicine Research and Development Centre (NARD), Science and Technology Department, National University of Quilmes, Roque Sáenz Peña 352, Bernal, Buenos Aires, Argentina
| | - Eder Lilia Romero
- Nanomedicine Research and Development Centre (NARD), Science and Technology Department, National University of Quilmes, Roque Sáenz Peña 352, Bernal, Buenos Aires, Argentina
| |
Collapse
|
10
|
Nong J, Glassman PM, Shuvaev VV, Reyes-Esteves S, Descamps HC, Kiseleva RY, Papp TE, Alameh MG, Tam YK, Mui BL, Omo-Lamai S, Zamora ME, Shuvaeva T, Arguiri E, Gong X, Brysgel TV, Tan AW, Woolfork AG, Weljie A, Thaiss CA, Myerson JW, Weissman D, Kasner SE, Parhiz H, Muzykantov VR, Brenner JS, Marcos-Contreras OA. Targeting lipid nanoparticles to the blood-brain barrier to ameliorate acute ischemic stroke. Mol Ther 2024; 32:1344-1358. [PMID: 38454606 PMCID: PMC11081939 DOI: 10.1016/j.ymthe.2024.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 01/28/2024] [Accepted: 03/05/2024] [Indexed: 03/09/2024] Open
Abstract
Effective delivery of mRNA or small molecule drugs to the brain is a significant challenge in developing treatment for acute ischemic stroke (AIS). To address the problem, we have developed targeted nanomedicine to increase drug concentrations in endothelial cells of the blood-brain barrier (BBB) of the injured brain. Inflammation during ischemic stroke causes continuous neuronal death and an increase in the infarct volume. To enable targeted delivery to the inflamed BBB, we conjugated lipid nanocarriers (NCs) with antibodies that bind cell adhesion molecules expressed at the BBB. In the transient middle cerebral artery occlusion mouse model, NCs targeted to vascular cellular adhesion molecule-1 (VCAM) achieved the highest level of brain delivery, nearly two orders of magnitude higher than untargeted ones. VCAM-targeted lipid nanoparticles with luciferase-encoding mRNA and Cre-recombinase showed selective expression in the ischemic brain. Anti-inflammatory drugs administered intravenously after ischemic stroke reduced cerebral infarct volume by 62% (interleukin-10 mRNA) or 35% (dexamethasone) only when they were encapsulated in VCAM-targeted NCs. Thus, VCAM-targeted lipid NCs represent a new platform for strongly concentrating drugs within the compromised BBB of penumbra, thereby ameliorating AIS.
Collapse
Affiliation(s)
- Jia Nong
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Patrick M Glassman
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pharmaceutical Sciences, School of Pharmacy, Temple University, Philadelphia, PA, USA
| | - Vladimir V Shuvaev
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sahily Reyes-Esteves
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Helene C Descamps
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Raisa Y Kiseleva
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tyler E Papp
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad-Gabriel Alameh
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ying K Tam
- Acuitas Therapeutics, Vancouver, British Columbia V6T 1Z3, Canada
| | - Barbara L Mui
- Acuitas Therapeutics, Vancouver, British Columbia V6T 1Z3, Canada
| | - Serena Omo-Lamai
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Marco E Zamora
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tea Shuvaeva
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Evguenia Arguiri
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xijing Gong
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Taylor V Brysgel
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ai Wen Tan
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ashley G Woolfork
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aalim Weljie
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christoph A Thaiss
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacob W Myerson
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Drew Weissman
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Scott E Kasner
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hamideh Parhiz
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Pulmonary Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Oscar A Marcos-Contreras
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Zamora ME, Omo-Lamai S, Patel MN, Wu J, Arguiri E, Muzykantov VR, Myerson JW, Marcos-Contreras OA, Brenner JS. Combination of Physicochemical Tropism and Affinity Moiety Targeting of Lipid Nanoparticles Enhances Organ Targeting. NANO LETTERS 2024. [PMID: 38598417 DOI: 10.1021/acs.nanolett.3c05031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Two camps have emerged for targeting nanoparticles to specific organs and cell types: affinity moiety targeting and physicochemical tropism. Here we directly compare and combine both using intravenous (IV) lipid nanoparticles (LNPs) designed to target the lungs. We utilized PECAM antibodies as affinity moieties and cationic lipids for physicochemical tropism. These methods yield nearly identical lung uptake, but aPECAM LNPs show higher endothelial specificity. LNPs combining these targeting methods had >2-fold higher lung uptake than either method alone and markedly enhanced epithelial uptake. To determine if lung uptake is because the lungs are the first organ downstream of IV injection, we compared IV vs intra-arterial (IA) injection into the carotid artery, finding that IA combined-targeting LNPs achieve 35% of the injected dose per gram (%ID/g) in the first-pass organ, the brain, among the highest reported. Thus, combining the affinity moiety and physicochemical strategies provides benefits that neither targeting method achieves alone.
Collapse
Affiliation(s)
- Marco E Zamora
- Drexel University, School of Biomedical Engineering, Philadelphia, Pennsylvania 19104, United States
- University of Pennsylvania, School of Systems Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Serena Omo-Lamai
- University of Pennsylvania, Department of Bioengineering, Philadelphia, Pennsylvania 19104, United States
| | - Manthan N Patel
- University of Pennsylvania, School of Systems Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jichuan Wu
- University of Pennsylvania, School of Systems Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Evguenia Arguiri
- University of Pennsylvania, School of Systems Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Vladmir R Muzykantov
- University of Pennsylvania, School of Systems Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jacob W Myerson
- University of Pennsylvania, School of Systems Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Oscar A Marcos-Contreras
- University of Pennsylvania, School of Systems Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
| | - Jacob S Brenner
- University of Pennsylvania, School of Systems Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania 19104, United States
- University of Pennsylvania, Department of Bioengineering, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
12
|
Li X, Zou J, He Z, Sun Y, Song X, He W. The interaction between particles and vascular endothelium in blood flow. Adv Drug Deliv Rev 2024; 207:115216. [PMID: 38387770 DOI: 10.1016/j.addr.2024.115216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 01/25/2024] [Accepted: 02/14/2024] [Indexed: 02/24/2024]
Abstract
Particle-based drug delivery systems have shown promising application potential to treat human diseases; however, an incomplete understanding of their interactions with vascular endothelium in blood flow prevents their inclusion into mainstream clinical applications. The flow performance of nano/micro-sized particles in the blood are disturbed by many external/internal factors, including blood constituents, particle properties, and endothelium bioactivities, affecting the fate of particles in vivo and therapeutic effects for diseases. This review highlights how the blood constituents, hemodynamic environment and particle properties influence the interactions and particle activities in vivo. Moreover, we briefly summarized the structure and functions of endothelium and simulated devices for studying particle performance under blood flow conditions. Finally, based on particle-endothelium interactions, we propose future opportunities for novel therapeutic strategies and provide solutions to challenges in particle delivery systems for accelerating their clinical translation. This review helps provoke an increasing in-depth understanding of particle-endothelium interactions and inspires more strategies that may benefit the development of particle medicine.
Collapse
Affiliation(s)
- Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Jiahui Zou
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Zhongshan He
- Department of Critical Care Medicine and Department of Biotherapy, Frontiers Science Center for Disease-related Molecular Network, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, PR China
| | - Yanhua Sun
- Shandong Provincial Key Laboratory of Microparticles Drug Delivery Technology, Qilu Pharmaceutical Co., LtD., Jinan 250000, PR China
| | - Xiangrong Song
- Department of Critical Care Medicine and Department of Biotherapy, Frontiers Science Center for Disease-related Molecular Network, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, PR China.
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China.
| |
Collapse
|
13
|
Du YQ, Yuan B, Ye YX, Zhou FL, Liu H, Huang JJ, Wei YF. Plumbagin Regulates Snail to Inhibit Hepatocellular Carcinoma Epithelial-Mesenchymal Transition in vivo and in vitro. J Hepatocell Carcinoma 2024; 11:565-580. [PMID: 38525157 PMCID: PMC10960549 DOI: 10.2147/jhc.s452924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/01/2024] [Indexed: 03/26/2024] Open
Abstract
Background/Aims Plumbagin (PL) has been shown to effe ctively inhibit autophagy, suppressing invasion and migration of hepatocellular carcinoma (HCC) cells. However, the specific mechanism remains unclear. This study aimed to investigate the effect of PL on tumor growth factor (TGF)-β-induced epithelial-mesenchymal transition (EMT) in HCC. Methods Huh-7 cells were cultured, and in vivo models of EMT and HCC-associated lung metastasis were developed through tail vein and in situ injections of tumor cells. In vivo imaging and hematoxylin and eosin staining were used to evaluate HCC modeling and lung metastasis. After PL intervention, the expression levels of Snail, vimentin, E-cadherin, and N-cadherin in the liver were evaluated through immunohistochemistry and Western blot. An in vitro TGF-β-induced cell EMT model was used to detect Snail, vimentin, E-cadherin, and N-cadherin mRNA levels through a polymerase chain reaction. Their protein levels were detected by immunofluorescence staining and Western blot. Results In vivo experiments demonstrated that PL significantly reduced the expression of Snail, vimentin, and N-cadherin, while increasing the expression of E-cadherin at the protein levels, effectively inhibiting HCC and lung metastasis. In vitro experiments confirmed that PL up-regulated epithelial cell markers, down-regulated mesenchymal cell markers, and inhibited EMT levels in HCC cells. Conclusion PL inhibits Snail expression, up-regulates E-cadherin expression, and down-regulates N-cadherin and vimentin expression, preventing EMT in HCC cells and reducing lung metastasis.
Collapse
Affiliation(s)
- Yuan-Qin Du
- Graduate School, Guangxi University of Traditional Chinese Medicine, Nanning, 530200, People’s Republic of China
| | - Bin Yuan
- Graduate School, Guangxi University of Traditional Chinese Medicine, Nanning, 530200, People’s Republic of China
| | - Yi-Xian Ye
- Graduate School, Guangxi University of Traditional Chinese Medicine, Nanning, 530200, People’s Republic of China
| | - Feng-ling Zhou
- Graduate School, Guangxi University of Traditional Chinese Medicine, Nanning, 530200, People’s Republic of China
| | - Hong Liu
- Graduate School, Guangxi University of Traditional Chinese Medicine, Nanning, 530200, People’s Republic of China
| | - Jing-Jing Huang
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, 530024, People’s Republic of China
| | - Yan-Fei Wei
- Department of Physiology, Guangxi University of Traditional Chinese Medicine, Nanning, 530200, People’s Republic of China
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Nanning, 530200, People’s Republic of China
| |
Collapse
|
14
|
Shariati L, Esmaeili Y, Rahimmanesh I, Babolmorad S, Ziaei G, Hasan A, Boshtam M, Makvandi P. Advances in nanobased platforms for cardiovascular diseases: Early diagnosis, imaging, treatment, and tissue engineering. ENVIRONMENTAL RESEARCH 2023; 238:116933. [PMID: 37652218 DOI: 10.1016/j.envres.2023.116933] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 09/02/2023]
Abstract
Cardiovascular diseases (CVDs) present a significant threat to health, with traditional therapeutics based treatment being hindered by inefficiencies, limited biological effects, and resistance to conventional drug. Addressing these challenges requires advanced approaches for early disease diagnosis and therapy. Nanotechnology and nanomedicine have emerged as promising avenues for personalized CVD diagnosis and treatment through theranostic agents. Nanoparticles serve as nanodevices or nanocarriers, efficiently transporting drugs to injury sites. These nanocarriers offer the potential for precise drug and gene delivery, overcoming issues like bioavailability and solubility. By attaching specific target molecules to nanoparticle surfaces, controlled drug release to targeted areas becomes feasible. In the field of cardiology, nanoplatforms have gained popularity due to their attributes, such as passive or active targeting of cardiac tissues, enhanced sensitivity and specificity, and easy penetration into heart and artery tissues due to their small size. However, concerns persist about the immunogenicity and cytotoxicity of nanomaterials, necessitating careful consideration. Nanoparticles also hold promise for CVD diagnosis and imaging, enabling straightforward diagnostic procedures and real-time tracking during therapy. Nanotechnology has revolutionized cardiovascular imaging, yielding multimodal and multifunctional vehicles that outperform traditional methods. The paper provides an overview of nanomaterial delivery routes, targeting techniques, and recent advances in treating, diagnosing, and engineering tissues for CVDs. It also discusses the future potential of nanomaterials in CVDs, including theranostics, aiming to enhance cardiovascular treatment in clinical practice. Ultimately, refining nanocarriers and delivery methods has the potential to enhance treatment effectiveness, minimize side effects, and improve patients' well-being and outcomes.
Collapse
Affiliation(s)
- Laleh Shariati
- Department of Biomaterials, Nanotechnology, and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Yasaman Esmaeili
- Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ilnaz Rahimmanesh
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shahrzad Babolmorad
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ghazal Ziaei
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha, 2713, Qatar; Biomedical Research Center, Qatar University, Doha, 2713, Qatar
| | - Maryam Boshtam
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, China; School of Engineering, Institute for Bioengineering, The University of Edinburgh, Edinburgh, EH9 3JL, UK.
| |
Collapse
|
15
|
Yang Y, Zoulikha M, Xiao Q, Huang F, Jiang Q, Li X, Wu Z, He W. Pulmonary endothelium-targeted nanoassembly of indomethacin and superoxide dismutase relieves lung inflammation. Acta Pharm Sin B 2023; 13:4607-4620. [PMID: 37969734 PMCID: PMC10638505 DOI: 10.1016/j.apsb.2023.05.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 11/17/2023] Open
Abstract
Lung inflammation is an essential inducer of various diseases and is closely related to pulmonary-endothelium dysfunction. Herein, we propose a pulmonary endothelium-targeted codelivery system of anti-inflammatory indomethacin (IND) and antioxidant superoxide dismutase (SOD) by assembling the biopharmaceutical SOD onto the "vector" of rod-like pure IND crystals, followed by coating with anti-ICAM-1 antibody (Ab) for targeting endothelial cells. The codelivery system has a 237 nm diameter in length and extremely high drug loading of 39% IND and 2.3% SOD. Pharmacokinetics and biodistribution studies demonstrate the extended blood circulation and the strong pulmonary accumulation of the system after intravenous injection in the lipopolysaccharide (LPS)-induced inflammatory murine model. Particularly, the system allows a robust capacity to target pulmonary endothelium mostly due to the rod-shape and Ab coating effect. In vitro, the preparation shows the synergistic anti-inflammatory and antioxidant effects in LPS-activated endothelial cells. In vivo, the preparation exhibits superior pharmacodynamic efficacy revealed by significantly downregulating the inflammatory/oxidative stress markers, such as TNF-α, IL-6, COX-2, and reactive oxygen species (ROS), in the lungs. In conclusion, the codelivery system based on rod-like pure crystals could well target the pulmonary endothelium and effectively alleviate lung inflammation. The study offers a promising approach to combat pulmonary endothelium-associated diseases.
Collapse
Affiliation(s)
- Yi Yang
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Makhloufi Zoulikha
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Qingqing Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
- Department of Traditional Chinese Medicine Processing and Preparation, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Feifei Huang
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Qi Jiang
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Zhenfeng Wu
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| |
Collapse
|
16
|
Tang J, Liu Y, Li M, Wang X, Du A, Gu N, Yang F. Sphingosine-1-Phosphate Receptor Targeted PLGA Nanobubbles for Inflammatory Vascular Endothelial Cell Catching. Adv Healthc Mater 2023; 12:e2301407. [PMID: 37591196 DOI: 10.1002/adhm.202301407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/06/2023] [Indexed: 08/19/2023]
Abstract
Vascular inflammation is an early manifestation and common pathophysiological basis of numerous cardiovascular and cerebrovascular diseases. However, effective surveillance methods are lacking. In this study, sulfur hexafluoride (SF6 )-loaded polylactic acid-co-glycolic acid (PLGA) nanobubbles (NBs) with a surface assembly of cyclodextrin (CD) and sphingosine-1-phosphate (S1P) (S1P@CD-PLGA NBs) are designed. The characterization results show that S1P@CD-PLGA NBs with diameters of ≈200 nm have good stability, biosafety, and ultrasound imaging-enhancement effects. When interacting with inflammatory vascular endothelial cells, S1P molecules encapsulated in cyclodextrin cavities exhibit a rapid, excellent, and stable targeting effect owing to their specific interaction with the highly expressed S1P receptor 1 (S1PR1) on the inflammatory vascular endothelial cells. Particularly, the S1P-S1PR1 interaction further activates the downstream signaling pathway of S1PR1 to reduce the expression of tumor necrosis factor-α (TNF-α) to protect endothelial cells. Furthermore, mouse models of carotid endothelial injuries and mesenteric thrombosis demonstrate that S1P@CD-PLGA NBs have excellent capabilities for in vivo targeting imaging. In summary, this study proposes a new strategy of using S1P to target inflammatory vascular endothelial cells while reducing the expression of TNF-α, which has the potential to be utilized in the targeted surveillance and treatment of vascular inflammatory diseases.
Collapse
Affiliation(s)
- Jian Tang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yang Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Mingxi Li
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, China
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, 210009, China
| | - Xiao Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Anning Du
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Ning Gu
- Medical School, Nanjing University, Nanjing, 210093, P. R. China
| | - Fang Yang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, China
| |
Collapse
|
17
|
Ejma-Multański A, Wajda A, Paradowska-Gorycka A. Cell Cultures as a Versatile Tool in the Research and Treatment of Autoimmune Connective Tissue Diseases. Cells 2023; 12:2489. [PMID: 37887333 PMCID: PMC10605903 DOI: 10.3390/cells12202489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023] Open
Abstract
Cell cultures are an important part of the research and treatment of autoimmune connective tissue diseases. By culturing the various cell types involved in ACTDs, researchers are able to broaden the knowledge about these diseases that, in the near future, may lead to finding cures. Fibroblast cultures and chondrocyte cultures allow scientists to study the behavior, physiology and intracellular interactions of these cells. This helps in understanding the underlying mechanisms of ACTDs, including inflammation, immune dysregulation and tissue damage. Through the analysis of gene expression patterns, surface proteins and cytokine profiles in peripheral blood mononuclear cell cultures and endothelial cell cultures researchers can identify potential biomarkers that can help in diagnosing, monitoring disease activity and predicting patient's response to treatment. Moreover, cell culturing of mesenchymal stem cells and skin modelling in ACTD research and treatment help to evaluate the effects of potential drugs or therapeutics on specific cell types relevant to the disease. Culturing cells in 3D allows us to assess safety, efficacy and the mechanisms of action, thereby aiding in the screening of potential drug candidates and the development of novel therapies. Nowadays, personalized medicine is increasingly mentioned as a future way of dealing with complex diseases such as ACTD. By culturing cells from individual patients and studying patient-specific cells, researchers can gain insights into the unique characteristics of the patient's disease, identify personalized treatment targets, and develop tailored therapeutic strategies for better outcomes. Cell culturing can help in the evaluation of the effects of these therapies on patient-specific cell populations, as well as in predicting overall treatment response. By analyzing changes in response or behavior of patient-derived cells to a treatment, researchers can assess the response effectiveness to specific therapies, thus enabling more informed treatment decisions. This literature review was created as a form of guidance for researchers and clinicians, and it was written with the use of the NCBI database.
Collapse
Affiliation(s)
- Adam Ejma-Multański
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (A.W.); (A.P.-G.)
| | | | | |
Collapse
|
18
|
Yuan M, Han Z, Liang Y, Sun Y, He B, Chen W, Li F. mRNA nanodelivery systems: targeting strategies and administration routes. Biomater Res 2023; 27:90. [PMID: 37740246 PMCID: PMC10517595 DOI: 10.1186/s40824-023-00425-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/26/2023] [Indexed: 09/24/2023] Open
Abstract
With the great success of coronavirus disease (COVID-19) messenger ribonucleic acid (mRNA) vaccines, mRNA therapeutics have gained significant momentum for the prevention and treatment of various refractory diseases. To function efficiently in vivo and overcome clinical limitations, mRNA demands safe and stable vectors and a reasonable administration route, bypassing multiple biological barriers and achieving organ-specific targeted delivery of mRNA. Nanoparticle (NP)-based delivery systems representing leading vector approaches ensure the successful intracellular delivery of mRNA to the target organ. In this review, chemical modifications of mRNA and various types of advanced mRNA NPs, including lipid NPs and polymers are summarized. The importance of passive targeting, especially endogenous targeting, and active targeting in mRNA nano-delivery is emphasized, and different cellular endocytic mechanisms are discussed. Most importantly, based on the above content and the physiological structure characteristics of various organs in vivo, the design strategies of mRNA NPs targeting different organs and cells are classified and discussed. Furthermore, the influence of administration routes on targeting design is highlighted. Finally, an outlook on the remaining challenges and future development toward mRNA targeted therapies and precision medicine is provided.
Collapse
Affiliation(s)
- Mujie Yuan
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Zeyu Han
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yan Liang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266073, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266073, China
| | - Bin He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Wantao Chen
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Fan Li
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
19
|
He H, Han Q, Wang S, Long M, Zhang M, Li Y, Zhang Y, Gu N. Design of a Multifunctional Nanozyme for Resolving the Proinflammatory Plaque Microenvironment and Attenuating Atherosclerosis. ACS NANO 2023; 17:14555-14571. [PMID: 37350440 DOI: 10.1021/acsnano.3c01420] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2023]
Abstract
Persistent inflammation within atherosclerotic plaques is a crucial factor contributing to plaque vulnerability and rupture. It has become increasingly evident that the proinflammatory microenvironment of the plaque, characterized by heightened monocyte recruitment, oxidative stress, and impaired clearance of apoptotic cells, plays a significant role in perpetuating inflammation and impeding its resolution. Consequently, targeting and eliminating these proinflammatory features within the plaque microenvironment have emerged as a promising therapeutic approach to restore inflammation resolution and mitigate the progression of atherosclerosis. While recent advancements in nanotherapeutics have demonstrated promising results in targeting individual proinflammatory characteristics, the development of an effective therapeutic strategy capable of simultaneously addressing multiple proinflammatory features remains a challenge. In this study, we developed a multifunctional nanozyme based on Prussian blue, termed PBNZ@PP-Man, to simultaneously target and eliminate various proinflammatory factors within the plaque microenvironment. Through systematic investigations, we have elucidated the antiatherosclerotic mechanisms of PBNZ@PP-Man. Our results demonstrate that PBNZ@PP-Man possesses the ability to accumulate within atherosclerotic plaques and effectively eliminate multiple proinflammatory factors, leading to inflammation resolution. Specifically, PBNZ@PP-Man suppresses monocyte recruitment, scavenges reactive oxygen species, and enhances efferocytosis. Notably, PBNZ@PP-Man exhibits a much stronger efficacy to resolve the proinflammatory plaque microenvironment and attenuate atherosclerosis in comparison to the approach that merely eliminates one single risky factor in the plaque. It significantly enhances the inflammation resolution capabilities of macrophages and attenuates atherosclerosis. These results collectively underscore the importance of modulating the proinflammatory plaque microenvironment as a complementary strategy for resolving inflammation in atherosclerosis.
Collapse
Affiliation(s)
- Hongliang He
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210009, People's Republic of China
| | - Qinggong Han
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210009, People's Republic of China
| | - Shi Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210009, People's Republic of China
| | - Mengmeng Long
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210009, People's Republic of China
| | - Miao Zhang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210009, People's Republic of China
| | - Yan Li
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210009, People's Republic of China
| | - Yu Zhang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210009, People's Republic of China
| | - Ning Gu
- School of Medicine, Nanjing University, Nanjing 210093, People's Republic of China
| |
Collapse
|
20
|
Nong J, Glassman PM, Reyes-Esteves S, Descamps HC, Shuvaev VV, Kiseleva RY, Papp TE, Alameh MG, Tam YK, Mui BL, Omo-Lamai S, Zamora ME, Shuvaeva T, Arguiri E, Thaiss CA, Myerson JW, Weissman D, Kasner SE, Parhiz H, Muzykantov VR, Brenner JS, Marcos-Contreras OA. Targeting lipid nanoparticles to the blood brain barrier to ameliorate acute ischemic stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544645. [PMID: 37398465 PMCID: PMC10312645 DOI: 10.1101/2023.06.12.544645] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
After more than 100 failed drug trials for acute ischemic stroke (AIS), one of the most commonly cited reasons for the failure has been that drugs achieve very low concentrations in the at-risk penumbra. To address this problem, here we employ nanotechnology to significantly enhance drug concentration in the penumbra's blood-brain barrier (BBB), whose increased permeability in AIS has long been hypothesized to kill neurons by exposing them to toxic plasma proteins. To devise drug-loaded nanocarriers targeted to the BBB, we conjugated them with antibodies that bind to various cell adhesion molecules on the BBB endothelium. In the transient middle cerebral artery occlusion (tMCAO) mouse model, nanocarriers targeted with VCAM antibodies achieved the highest level of brain delivery, nearly 2 orders of magnitude higher than untargeted ones. VCAM-targeted lipid nanoparticles loaded with either a small molecule drug (dexamethasone) or mRNA (encoding IL-10) reduced cerebral infarct volume by 35% or 73%, respectively, and both significantly lowered mortality rates. In contrast, the drugs delivered without the nanocarriers had no effect on AIS outcomes. Thus, VCAM-targeted lipid nanoparticles represent a new platform for strongly concentrating drugs within the compromised BBB of penumbra, thereby ameliorating AIS. Graphical abstract Acute ischemic stroke induces upregulation of VCAM. We specifically targeted upregulated VCAM in the injured region of the brain with drug- or mRNA-loaded targeted nanocarriers. Nanocarriers targeted with VCAM antibodies achieved the highest brain delivery, nearly orders of magnitude higher than untargeted ones. VCAM-targeted nanocarriers loaded with dexamethasone and mRNA encoding IL-10 reduced infarct volume by 35% and 73%, respectively, and improved survival rates.
Collapse
|
21
|
Zhao X, Han J, Zhou L, Zhao J, Huang M, Wang Y, Kou J, Kou Y, Jin J. High mobility group box 1 derived mainly from platelet microparticles exacerbates microvascular obstruction in no reflow. Thromb Res 2023; 222:49-62. [PMID: 36566704 DOI: 10.1016/j.thromres.2022.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/07/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
INTRODUCTION No reflow manifests coronary microvascular injury caused by continuous severe myocardial ischemia and reperfusion. Microvascular obstruction (MVO) has emerged as one fundamental mechanism of no reflow. However, the underlying pathophysiology remains incompletely defined. Herein, we explore the contribution of high mobility group box 1 (HMGB1), derived mainly from platelet microparticles exacerbating MVO in no reflow. MATERIALS AND METHODS 44 STEMI patients undergoing successful primary percutaneous coronary intervention (PCI) were included in our study. Plasma HMGB1 levels in both the peripheral artery (PA) and infarct-related coronary artery (IRA) were measured by ELISA. Flow cytometry and confocal microscopy assessed the level of HMGB1+ platelet derived microparticles (PMPs) and platelet activation. Flow cytometry and western blot evaluated the procoagulant activity (PCA) and the release of inflammatory factors of human microvascular endothelial cells (HCEMCs). RESULTS HMGB1 levels were significantly higher in the IRA in no-reflow patients. The levels of HMGB1+ PMPs were considerably higher in the IRA of patients with no reflow and were strongly associated with platelet activation. Moreover, our results show that HMGB1 interacts with human microvascular endothelial cells primarily through TLR4, inducing HCMEC proinflammatory, procoagulant phenotype, and monocyte recruitment, accelerating microvascular obstruction and facilitating the development of no reflow. CONCLUSION Our results illustrate a novel mechanism by which HMGB1, derived mainly from PMPs, plays a crucial role in the pathogenesis of no-reflow, revealing a novel therapeutic target.
Collapse
Affiliation(s)
- Xinyi Zhao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Ministry of Education, Heilongjiang Province, Harbin, China
| | - Jianbin Han
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Ministry of Education, Heilongjiang Province, Harbin, China
| | - Lijin Zhou
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinjin Zhao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Ministry of Education, Heilongjiang Province, Harbin, China
| | - Meijiao Huang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Ministry of Education, Heilongjiang Province, Harbin, China
| | - Yueqing Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Ministry of Education, Heilongjiang Province, Harbin, China
| | - Junjie Kou
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Ministry of Education, Heilongjiang Province, Harbin, China.
| | - Yan Kou
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Key Laboratory of Myocardial Ischemia, Ministry of Education, Heilongjiang Province, Harbin, China.
| | - Jiaqi Jin
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Heilongjiang Province, Harbin, China; Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
22
|
Zheng W, He R, Liang X, Roudi S, Bost J, Coly P, van Niel G, Andaloussi SEL. Cell-specific targeting of extracellular vesicles through engineering the glycocalyx. J Extracell Vesicles 2022; 11:e12290. [PMID: 36463392 PMCID: PMC9719568 DOI: 10.1002/jev2.12290] [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] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/16/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022] Open
Abstract
Extracellular vesicles (EVs) are promising carriers for the delivery of a variety of chemical and biological drugs. However, their efficacy is limited by the lack of cellular specificity. Available methods to improve the tissue specificity of EVs predominantly rely on surface display of proteins and peptides, largely overlooking the dense glycocalyx that constitutes the outermost layer of EVs. In the present study, we report a reconfigurable glycoengineering strategy that can endogenously display glycans of interest on EV surface. Briefly, EV producer cells are genetically engineered to co-express a glycosylation domain (GD) inserted into the large extracellular loop of CD63 (a well-studied EV scaffold protein) and fucosyltransferase VII (FUT7) or IX (FUT9), so that the engineered EVs display the glycan of interest. Through this strategy, we showcase surface display of two types of glycan ligands, sialyl Lewis X (sLeX) and Lewis X, on EVs and achieve high specificity towards activated endothelial cells and dendritic cells, respectively. Moreover, the endothelial cell-targeting properties of sLeX-EVs were combined with the intrinsic therapeutic effects of mesenchymal stem cells (MSCs), leading to enhanced attenuation of endothelial damage. In summary, this study presents a reconfigurable glycoengineering strategy to produce EVs with strong cellular specificity and highlights the glycocalyx as an exploitable trait for engineering EVs.
Collapse
Affiliation(s)
- Wenyi Zheng
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetHuddingeSweden
- Centre for Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalHuddingeSweden
| | - Rui He
- Centre for Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalHuddingeSweden
- Experimental Cancer Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetHuddingeSweden
| | - Xiuming Liang
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetHuddingeSweden
- Centre for Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalHuddingeSweden
| | - Samantha Roudi
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetHuddingeSweden
- Centre for Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalHuddingeSweden
| | - Jeremy Bost
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetHuddingeSweden
- Centre for Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalHuddingeSweden
| | - Pierre‐Michael Coly
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266ParisFrance
- GHU Paris Psychiatrie et NeurosciencesHôpital Sainte AnneParisFrance
| | - Guillaume van Niel
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266ParisFrance
- GHU Paris Psychiatrie et NeurosciencesHôpital Sainte AnneParisFrance
| | - Samir E. L. Andaloussi
- Biomolecular Medicine, Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetHuddingeSweden
- Centre for Allogeneic Stem Cell Transplantation (CAST)Karolinska University HospitalHuddingeSweden
- EVOX Therapeutics LimitedOxfordUK
| |
Collapse
|
23
|
Jin R, Chan AKY, Wu J, Lee TMC. Relationships between Inflammation and Age-Related Neurocognitive Changes. Int J Mol Sci 2022; 23:12573. [PMID: 36293430 PMCID: PMC9604276 DOI: 10.3390/ijms232012573] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 11/18/2022] Open
Abstract
The relationship between inflammation and age-related neurocognitive changes is significant, which may relate to the age-related immune dysfunctions characterized by the senescence of immune cells and elevated inflammatory markers in the peripheral circulation and the central nervous system. In this review, we discuss the potential mechanisms, including the development of vascular inflammation, neuroinflammation, organelle dysfunctions, abnormal cholesterol metabolism, and glymphatic dysfunctions as well as the role that the key molecules play in the immune-cognition interplay. We propose potential therapeutic pharmacological and behavioral strategies for ameliorating age-related neurocognitive changes associated with inflammation. Further research to decipher the multidimensional roles of chronic inflammation in normal and pathological aging processes will help unfold the pathophysiological mechanisms underpinning neurocognitive disorders. The insight gained will lay the path for developing cost-effective preventative measures and the buffering or delaying of age-related neurocognitive decline.
Collapse
Affiliation(s)
- Run Jin
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong 999077, China
- Laboratory of Neuropsychology and Human Neuroscience, The University of Hong Kong, Hong Kong 999077, China
| | - Aidan Kai Yeung Chan
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong 999077, China
- Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Jingsong Wu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350108, China
| | - Tatia Mei Chun Lee
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong 999077, China
- Laboratory of Neuropsychology and Human Neuroscience, The University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
24
|
Zhang Z, Dalan R, Hu Z, Wang JW, Chew NW, Poh KK, Tan RS, Soong TW, Dai Y, Ye L, Chen X. Reactive Oxygen Species Scavenging Nanomedicine for the Treatment of Ischemic Heart Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202169. [PMID: 35470476 DOI: 10.1002/adma.202202169] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/08/2022] [Indexed: 06/14/2023]
Abstract
Ischemic heart disease (IHD) is the leading cause of disability and mortality worldwide. Reactive oxygen species (ROS) have been shown to play key roles in the progression of diabetes, hypertension, and hypercholesterolemia, which are independent risk factors that lead to atherosclerosis and the development of IHD. Engineered biomaterial-based nanomedicines are under extensive investigation and exploration, serving as smart and multifunctional nanocarriers for synergistic therapeutic effect. Capitalizing on cell/molecule-targeting drug delivery, nanomedicines present enhanced specificity and safety with favorable pharmacokinetics and pharmacodynamics. Herein, the roles of ROS in both IHD and its risk factors are discussed, highlighting cardiovascular medications that have antioxidant properties, and summarizing the advantages, properties, and recent achievements of nanomedicines that have ROS scavenging capacity for the treatment of diabetes, hypertension, hypercholesterolemia, atherosclerosis, ischemia/reperfusion, and myocardial infarction. Finally, the current challenges of nanomedicines for ROS-scavenging treatment of IHD and possible future directions are discussed from a clinical perspective.
Collapse
Affiliation(s)
- Zhan Zhang
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China
| | - Rinkoo Dalan
- Department of Endocrinology, Tan Tock Seng Hospital, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 408433, Singapore
| | - Zhenyu Hu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Jiong-Wei Wang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Diagnostic Radiology and Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Nicholas Ws Chew
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, 119074, Singapore
| | - Kian-Keong Poh
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, 119074, Singapore
| | - Ru-San Tan
- Department of Cardiology, National Heart Centre Singapore, Singapore, 119609, Singapore
| | - Tuck Wah Soong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yunlu Dai
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macao, Taipa, Macau SAR, 999078, China
| | - Lei Ye
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology and Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Chemical and Biomolecular Engineering and Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| |
Collapse
|
25
|
Venditto VJ, Sockolosky J, Nguyen J. Translational Drug Delivery: Time to be Frank for Future Success. Adv Drug Deliv Rev 2022; 189:114521. [PMID: 36030019 DOI: 10.1016/j.addr.2022.114521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | - Jonathan Sockolosky
- Department of Antibody Engineering, Genentech, Inc., South San Francisco, CA, USA
| | - Juliane Nguyen
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina, Eshelman School of Pharmacy, Chapel Hill, NC, USA
| |
Collapse
|