1
|
Udriște AS, Burdușel AC, Niculescu AG, Rădulescu M, Balaure PC, Grumezescu AM. Organic Nanoparticles in Progressing Cardiovascular Disease Treatment and Diagnosis. Polymers (Basel) 2024; 16:1421. [PMID: 38794614 PMCID: PMC11125450 DOI: 10.3390/polym16101421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/26/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024] Open
Abstract
Cardiovascular diseases (CVDs), the world's most prominent cause of mortality, continue to be challenging conditions for patients, physicians, and researchers alike. CVDs comprise a wide range of illnesses affecting the heart, blood vessels, and the blood that flows through and between them. Advances in nanomedicine, a discipline focused on improving patient outcomes through revolutionary treatments, imaging agents, and ex vivo diagnostics, have created enthusiasm for overcoming limitations in CVDs' therapeutic and diagnostic landscapes. Nanomedicine can be involved in clinical purposes for CVD through the augmentation of cardiac or heart-related biomaterials, which can be functionally, mechanically, immunologically, and electrically improved by incorporating nanomaterials; vasculature applications, which involve systemically injected nanotherapeutics and imaging nanodiagnostics, nano-enabled biomaterials, or tissue-nanoengineered solutions; and enhancement of sensitivity and/or specificity of ex vivo diagnostic devices for patient samples. Therefore, this review discusses the latest studies based on applying organic nanoparticles in cardiovascular illness, including drug-conjugated polymers, lipid nanoparticles, and micelles. Following the revised information, it can be concluded that organic nanoparticles may be the most appropriate type of treatment for cardiovascular diseases due to their biocompatibility and capacity to integrate various drugs.
Collapse
Affiliation(s)
- Alexandru Scafa Udriște
- Department 4 Cardio-Thoracic Pathology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Alexandra Cristina Burdușel
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (A.C.B.); (A.-G.N.); (A.M.G.)
| | - Adelina-Gabriela Niculescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (A.C.B.); (A.-G.N.); (A.M.G.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Marius Rădulescu
- Department of Inorganic Chemistry, Physical Chemistry and Electrochemistry, National University of Science and Technology Politehnica Bucharest, 1-7 Polizu St., 011061 Bucharest, Romania;
| | - Paul Cătălin Balaure
- Department of Organic Chemistry, National University of Science and Technology Politehnica Bucharest, 1-7 Polizu St., 011061 Bucharest, Romania
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (A.C.B.); (A.-G.N.); (A.M.G.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
| |
Collapse
|
2
|
Ullah A, Ullah M, Lim SI. Recent advancements in nanotechnology based drug delivery for the management of cardiovascular disease. Curr Probl Cardiol 2024; 49:102396. [PMID: 38266693 DOI: 10.1016/j.cpcardiol.2024.102396] [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: 01/06/2024] [Accepted: 01/14/2024] [Indexed: 01/26/2024]
Abstract
Cardiovascular diseases (CVDs) constitute a predominant cause of both global mortality and morbidity. To address the challenges in the early diagnosis and management of CVDs, there is growing interest in the field of nanotechnology and nanomaterials to develop innovative diagnostic and therapeutic approaches. This review focuses on the recent advancements in nanotechnology-based diagnostic techniques, including cardiac immunoassays (CIA), cardiac circulating biomarkers, cardiac exosomal biomarkers, and molecular Imaging (MOI). Moreover, the article delves into the exciting developments in nanoparticles (NPs), biomimetic NPs, nanofibers, nanogels, and nanopatchs for cardiovascular applications. And discuss how these nanoscale technologies can improve the precision, sensitivity, and speed of CVD diagnosis and management. While highlighting their vast potential, we also address the limitations and challenges that must be overcome to harness these innovations successfully. Furthermore, this review focuses on the emerging opportunities for personalized and effective cardiovascular care through the integration of nanotechnology, ultimately aiming to reduce the global burden of CVDs.
Collapse
Affiliation(s)
- Aziz Ullah
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Engineering Bldg#1, Rm1108, Busan 48513, Republic of Korea
| | - Muneeb Ullah
- College of Pharmacy, Pusan National University, Busandaehak-ro 63 beon-gil 2, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Sung In Lim
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Engineering Bldg#1, Rm1108, Busan 48513, Republic of Korea.
| |
Collapse
|
3
|
He J, Gao Y, Yang C, Guo Y, Liu L, Lu S, He H. Navigating the landscape: Prospects and hurdles in targeting vascular smooth muscle cells for atherosclerosis diagnosis and therapy. J Control Release 2024; 366:261-281. [PMID: 38161032 DOI: 10.1016/j.jconrel.2023.12.047] [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: 09/21/2023] [Revised: 12/02/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
Vascular smooth muscle cells (VSMCs) have emerged as pivotal contributors throughout all phases of atherosclerotic plaque development, effectively dispelling prior underestimations of their prevalence and significance. Recent lineage tracing studies have unveiled the clonal nature and remarkable adaptability inherent to VSMCs, thereby illuminating their intricate and multifaceted roles in the context of atherosclerosis. This comprehensive review provides an in-depth exploration of the intricate mechanisms and distinctive characteristics that define VSMCs across various physiological processes, firmly underscoring their paramount importance in shaping the course of atherosclerosis. Furthermore, this review offers a thorough examination of the significant strides made over the past two decades in advancing imaging techniques and therapeutic strategies with a precise focus on targeting VSMCs within atherosclerotic plaques, notably spotlighting meticulously engineered nanoparticles as a promising avenue. We envision the potential of VSMC-targeted nanoparticles, thoughtfully loaded with medications or combination therapies, to effectively mitigate pro-atherogenic VSMC processes. These advancements are poised to contribute significantly to the pivotal objective of modulating VSMC phenotypes and enhancing plaque stability. Moreover, our paper also delves into recent breakthroughs in VSMC-targeted imaging technologies, showcasing their remarkable precision in locating microcalcifications, dynamically monitoring plaque fibrous cap integrity, and assessing the therapeutic efficacy of medical interventions. Lastly, we conscientiously explore the opportunities and challenges inherent in this innovative approach, providing a holistic perspective on the potential of VSMC-targeted strategies in the evolving landscape of atherosclerosis research and treatment.
Collapse
Affiliation(s)
- Jianhua He
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China.
| | - Yu Gao
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Can Yang
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Yujie Guo
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Lisha Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| | - Shan Lu
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China.
| | - 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.
| |
Collapse
|
4
|
Feng W, Teng Y, Zhong Q, Zhang Y, Zhang J, Zhao P, Chen G, Wang C, Liang XJ, Ou C. Biomimetic Grapefruit-Derived Extracellular Vesicles for Safe and Targeted Delivery of Sodium Thiosulfate against Vascular Calcification. ACS NANO 2023; 17:24773-24789. [PMID: 38055864 PMCID: PMC10753875 DOI: 10.1021/acsnano.3c05261] [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: 06/11/2023] [Revised: 11/24/2023] [Accepted: 11/29/2023] [Indexed: 12/08/2023]
Abstract
As the prevalence of vascular calcification (VC), a strong contributor to cardiovascular morbidity and mortality, continues to increase, the need for pharmacologic therapies becomes urgent. Sodium thiosulfate (STS) is a clinically approved drug for therapy against VC; however, its efficacy is hampered by poor bioavailability and severe adverse effects. Plant-derived extracellular vesicles have provided options for VC treatment since they can be used as biomimetic drug carriers with higher biosafety and targeting abilities than artificial carriers. Inspired by natural grapefruit-derived extracellular vesicles (EVs), we fabricated a biomimetic nanocarrier comprising EVs loaded with STS and further modified with hydroxyapatite crystal binding peptide (ESTP) for VC-targeted delivery of STS. In vitro, the ESTP nanodrug exhibited excellent cellular uptake capacity by calcified vascular smooth muscle cells (VSMCs) and subsequently inhibited VSMCs calcification. In the VC mice model, the ESTP nanodrug showed preferentially the highest accumulation in the calcified arteries compared to other treatment groups. Mechanistically, the ESTP nanodrug significantly prevented VC via driving M2 macrophage polarization, reducing inflammation, and suppressing bone-vascular axis as demonstrated by inhibiting osteogenic phenotype trans-differentiation of VSMCs while enhancing bone quality. In addition, the ESTP nanodrug did not induce hemolysis or cause any damage to other organs. These results suggest that the ESTP nanodrug can prove to be a promising agent against VC without the concern of systemic toxicity.
Collapse
Affiliation(s)
- Weijing Feng
- The
Tenth Affiliated Hospital of Southern Medical University (Dongguan
People’s Hospital), Southern Medical University or The First
School of Clinical Medicine, Southern Medical
University, Dongguan 523018, China
- Department
of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong
Provincial Key Laboratory of Cardiac Function and Microcirculation,
Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yintong Teng
- The
Tenth Affiliated Hospital of Southern Medical University (Dongguan
People’s Hospital), Southern Medical University or The First
School of Clinical Medicine, Southern Medical
University, Dongguan 523018, China
| | - Qingping Zhong
- The
Tenth Affiliated Hospital of Southern Medical University (Dongguan
People’s Hospital), Southern Medical University or The First
School of Clinical Medicine, Southern Medical
University, Dongguan 523018, China
| | - Yangmei Zhang
- The
Tenth Affiliated Hospital of Southern Medical University (Dongguan
People’s Hospital), Southern Medical University or The First
School of Clinical Medicine, Southern Medical
University, Dongguan 523018, China
| | - Jianwu Zhang
- Department
of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong
Provincial Key Laboratory of Cardiac Function and Microcirculation,
Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Peng Zhao
- NMPA
Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong
Provincial Key Laboratory of New Drug Screening, Guangdong Provincial
Key Laboratory of Cardiac Function and Microcirculation, School of
Pharmaceutical Sciences, Southern Medical
University, Guangzhou 510515, China
| | - Guoqing Chen
- Cardiology
Department of Panyu Central Hospital and Cardiovascular Disease Institute
of Panyu District, Guangzhou 511400, China
| | - Chunming Wang
- Institute
of Chinese Medical Sciences & State Key Laboratory of Quality
Research in Chinese Medicine, University
of Macau, Macau 00000, SAR, China
| | - Xing-Jie Liang
- Chinese Academy
of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key
Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Caiwen Ou
- The
Tenth Affiliated Hospital of Southern Medical University (Dongguan
People’s Hospital), Southern Medical University or The First
School of Clinical Medicine, Southern Medical
University, Dongguan 523018, China
| |
Collapse
|
5
|
Chin DD, Patel N, Lee W, Kanaya S, Cook J, Chung EJ. Long-term, in vivo therapeutic effects of a single dose of miR-145 micelles for atherosclerosis. Bioact Mater 2023; 27:327-336. [PMID: 37122900 PMCID: PMC10140752 DOI: 10.1016/j.bioactmat.2023.04.001] [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: 12/20/2022] [Revised: 03/31/2023] [Accepted: 04/01/2023] [Indexed: 05/02/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that is characterized by the build-up of lipid-rich plaques in the arterial walls. The standard treatment for patients with atherosclerosis is statin therapy aimed to lower serum lipid levels. Despite its widespread use, many patients taking statins continue to experience acute events. Thus, to develop improved and alternative therapies, we previously reported on microRNA-145 (miR-145 micelles) and its ability to inhibit atherosclerosis by targeting vascular smooth muscle cells (VSMCs). Importantly, one dose of miR-145 micelles significantly abrogated disease progression when evaluated two weeks post-administration. Thus, in this study, to evaluate how long the sustained effects of miR-145 micelles can be maintained and towards identifying a dosing regimen that is practical for patients with chronic disease, the therapeutic effects of a single dose of miR-145 micelles were evaluated for up to two months in vivo. After one and two months post-treatment, miR-145 micelles were found to reduce plaque size and overall lesion area compared to all other controls including statins without causing adverse effects. Furthermore, a single dose of miR-145 micelle treatment inhibited VSMC transdifferentiation into pathogenic macrophage-like and osteogenic cells in plaques. Together, our data shows the long-term efficacy and sustained effects of miR-145 micelles that is amenable using a dosing frequency relevant to chronic disease patients.
Collapse
Affiliation(s)
- Deborah D. Chin
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Neil Patel
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Woori Lee
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Sonali Kanaya
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Jackson Cook
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, United States
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, 90089, United States
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, United States
| |
Collapse
|
6
|
Smith BR, Edelman ER. Nanomedicines for cardiovascular disease. NATURE CARDIOVASCULAR RESEARCH 2023; 2:351-367. [PMID: 39195953 DOI: 10.1038/s44161-023-00232-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 01/25/2023] [Indexed: 08/29/2024]
Abstract
The leading cause of death in the world, cardiovascular disease (CVD), remains a formidable condition for researchers, clinicians and patients alike. CVD comprises a broad collection of diseases spanning the heart, the vasculature and the blood that runs through and interconnects them. Limitations in CVD therapeutic and diagnostic landscapes have generated excitement for advances in nanomedicine, a field focused on improving patient outcomes through transformative therapies, imaging agents and ex vivo diagnostics. CVD nanomedicines are fundamentally shaped by their intended clinical application, including (1) cardiac or heart-related biomaterials, which can be functionally (for example, mechanically, immunologically, electrically) improved by incorporating nanomaterials; (2) the vasculature, involving systemically injected nanotherapeutics and imaging nanodiagnostics, nano-enabled biomaterials or tissue-nanoengineered solutions; and (3) improving the sensitivity and/or specificity of ex vivo diagnostic devices for patient samples. While immunotherapy has developed into a key pillar of oncology in the past dozen years, CVD immunotherapy and immunoimaging are recently emergent and likely to factor substantially in CVD management in the coming decade. The nanomaterials in CVD-related clinical trials and many promising preclinical strategies indicate that nanomedicine is on the cusp of greatly impacting patients with CVD. Here we review these recent advances, highlighting key clinical opportunities in the rapidly emerging field of CVD nanomedicine.
Collapse
Affiliation(s)
- Bryan Ronain Smith
- Department of Biomedical Engineering and Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA.
| | - Elazer R Edelman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Cardiovascular Division, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Lin CC, Chiu LH, Chang WH, Lin CAJ, Chen RM, Ho YS, Zuo CS, Changou A, Cheng YF, Lai WFT. A Non-Invasive Method for Monitoring Osteogenesis and Osseointegration Using Near-Infrared Fluorescent Imaging: A Model of Maxilla Implantation in Rats. Int J Mol Sci 2023; 24:ijms24055032. [PMID: 36902462 PMCID: PMC10003657 DOI: 10.3390/ijms24055032] [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: 12/22/2022] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
Currently, computed tomography and conventional X-ray radiography usually generate a micro-artifact around metal implants. This metal artifact frequently causes false positive or negative diagnoses of bone maturation or pathological peri-implantitis around implants. In an attempt to repair the artifacts, a highly specific nanoprobe, an osteogenic biomarker, and nano-Au-Pamidronate were designed to monitor the osteogenesis. In total, 12 Sprague Dawley rats were included in the study and could be chategorized in 3 groups: 4 rats in the X-ray and CT group, 4 rats in the NIRF group, and 4 rats in the sham group. A titanium alloy screw was implanted in the anterior hard palate. The X-ray, CT, and NIRF images were taken 28 days after implantation. The X-ray showed that the tissue surrounded the implant tightly; however, a gap of metal artifacts was noted around the interface between dental implants and palatal bone. Compared to the CT image, a fluorescence image was noted around the implant site in the NIRF group. Furthermore, the histological implant-bone tissue also exhibited a significant NIRF signal. In conclusion, this novel NIRF molecular imaging system precisely identifies the image loss caused by metal artifacts and can be applied to monitoring bone maturation around orthopedic implants. In addition, by observing the new bone formation, a new principle and timetable for an implant osseointegrated with bone can be established and a new type of implant fixture or surface treatment can be evaluated using this system.
Collapse
Affiliation(s)
- Chien-Chou Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Li-Hsuan Chiu
- McLean Imaging Center, McLean Hospital and Harvard Medical School, Belmont, MA 02478, USA
| | - Walter H. Chang
- Department of Biomedical Engineering, Chung Yuan Christian University, Taoyuan 320, Taiwan
| | - Cheng-An J. Lin
- Department of Biomedical Engineering, Chung Yuan Christian University, Taoyuan 320, Taiwan
| | - Ruei-Ming Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yuan-Soon Ho
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Chun S. Zuo
- McLean Imaging Center, McLean Hospital and Harvard Medical School, Belmont, MA 02478, USA
| | - Austin Changou
- Ph.D. Program for Translational Medicine, College of Medicine and Technology, Taipei Medical University, Taipei 110, Taiwan
- Core Facility Center, Office of Research and Development, Taipei Medical University, Taipei 110, Taiwan
| | - Yue-Fa Cheng
- College of Basic Medicine, North China University of Science and Technology, Tangshan 066008, China
| | - Wen-Fu T. Lai
- McLean Imaging Center, McLean Hospital and Harvard Medical School, Belmont, MA 02478, USA
- Institute of Graduate Clinical Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Research and Department of Dentistry, Taipei Medical University-Shuang-Ho Hospital, New Taipei City 235, Taiwan
- Correspondence:
| |
Collapse
|
8
|
Zhang WB, Qi YF, Xiao ZX, Chen H, Liu SH, Li ZZ, Zeng ZF, Wu HF. CircHIPK3 Regulates Vascular Smooth Muscle Cell Calcification Via the miR-106a-5p/MFN2 Axis. J Cardiovasc Transl Res 2022; 15:1315-1326. [PMID: 35467292 DOI: 10.1007/s12265-022-10247-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/24/2022] [Indexed: 02/06/2023]
Abstract
Atherosclerosis is the most common arterial disease and is closely related to vascular calcification. CircHIPK3 has been implicated in atherosclerosis development, but the possible downstream regulatory mechanisms remain unclear. The levels of circHIPK3, miR-106a and MFN2 in tissues and blood samples of patients with atherosclerosis were detected by RT-qPCR. The levels of circHIPK3, miR-106a and MFN2 were detected by RT-qPCR and the expression levels of MFN2, osteogenic and cartilage differentiation marker proteins were detected by western blot in vitro. ALP staining, Alizarin Red staining, and calcium content detection evaluated the degree of osteogenic differentiation of cells. Alcian blue staining detected the level of cell cartilage differentiation. Luciferase detected the targeting relationship between circHIPK3 and miR-106a-5p, as well as miR-106a-5p and MFN2. CircHIPK3 and MFN2 were low expressed and miR-106a-5p was highly expressed in tissues and blood samples of patients with atherosclerosis, as well as vascular smooth muscle cell (VSMC) with osteogenic and cartilage differentiation. Overexpression of circHIPK3 reduced the cell mineralization and calcium content. Overexpression of circHIPK3 inhibited osteogenic differentiation by decreasing ALP activity, RUNX2, and OPG expression, and increasing SM22α and SMA level. What's more, overexpression of circHIPK3 decreased the chondrogenic differentiation by inhibiting the protein level of SOX9, aggrecan, and collagen II. CircHIPK3 targeted miR-106a-5p and miR-106a-5p targeted MFN2. MiR-106a-5p overexpression or MFN2 depletion repressed the effect of circHIPK3 overexpression on VSMC calcification. CircHIPK3 regulated osteogenic and cartilage differentiation of VSMC via miR-106a-5p/MFN2 axis, indicating a target for treating vascular calcification.
Collapse
Affiliation(s)
- Wen-Bo Zhang
- Department of Vascular Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan Province, People's Republic of China.
| | - You-Fei Qi
- Department of Vascular Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan Province, People's Republic of China
| | - Zhan-Xiang Xiao
- Department of Vascular Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan Province, People's Republic of China
| | - Hao Chen
- Department of Vascular Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan Province, People's Republic of China
| | - Sa-Hua Liu
- Department of Vascular Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan Province, People's Republic of China
| | - Zhen-Zhen Li
- Department of Vascular Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan Province, People's Republic of China
| | - Zhao-Fan Zeng
- Department of Vascular Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan Province, People's Republic of China
| | - Hong-Fei Wu
- Department of Vascular Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No.19, Xiuhua Road, Haikou, 570311, Hainan Province, People's Republic of China
| |
Collapse
|
9
|
Yang J, Wang M, Yang J, Chu Z, Chen X, Wu X, Peng X. Calcifying nanoparticles initiate the calcification process of mesenchymal stem cells in vitro through the activation of the TGF-β1/Smad signaling pathway and promote the decay of echinococcosis. Open Life Sci 2022. [DOI: 10.1515/biol-2022-0503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Abstract
The role of the calcifying nanoparticles (CNPs) in the calcification process of the outer cyst wall in hepatic cystic echinococcosis (HCE) remains unknown. CNPs were isolated from the tissues of the patients with HCE. Western blotting, alkaline phosphatase staining, and alizarin staining were performed to detect the cellular calcium ion deposition induced by the CNPs. CCK-8 and flow cytometry assays were conducted to determine the effect of CNPs on the apoptosis of mesenchymal stem cells (MSCs). Western blot experiments were performed to examine the expression levels of apoptosis-related factors and TGF-β1/Smad signaling pathway constituents. Treatment with CNPs induced the differentiation of MSCs. Calcium-related proteins, including OPN, BMP-2, and RUNX2, were upregulated after the CNP treatment. Similarly, CNP exposure increased the cellular calcium ion deposition in MSCs. In addition, the expression of Bax and Caspase-8 was elevated by the CNPs in MSCs. Treatment with CNPs promoted MSC apoptosis and inhibited the MSC growth. The TGF-β1/Smad signaling pathway was also activated after the CNP treatment. This study indicated that CNPs may play a critical role in initiating calcification of the outer cyst wall of HCE and promote the decay of echinococcosis, providing a new strategy for the treatment of hepatic echinococcosis.
Collapse
Affiliation(s)
- Jian Yang
- Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , 430030 , China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Medical College, Shihezi University , No. 107, North Second Road , Shihezi , 832008, Xinjiang , China
| | - Meiyan Wang
- Department of Nursing, Shihezi University School of Medicine , Shihezi , 832000, Xinjiang , China
| | - Jing Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Medical College, Shihezi University , No. 107, North Second Road , Shihezi , 832008, Xinjiang , China
| | - Zhiqiang Chu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Medical College, Shihezi University , No. 107, North Second Road , Shihezi , 832008, Xinjiang , China
| | - Xueling Chen
- Department of Immunology, Shihezi University School of Medicine , Shihezi , 832000, Xinjiang , China
| | - Xiangwei Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Medical College, Shihezi University , No. 107, North Second Road , Shihezi , 832008, Xinjiang , China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine , Shihezi , Xinjiang, 832000 , China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, School of Medicine, Shihezi University , Shihezi , 832000, Xinjiang , China
| | - Xinyu Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Medical College, Shihezi University , No. 107, North Second Road , Shihezi , 832008, Xinjiang , China
| |
Collapse
|
10
|
Patel N, Chin DD, Magee GA, Chung EJ. Therapeutic Response of miR-145 Micelles on Patient-Derived Vascular Smooth Muscle Cells. Front Digit Health 2022; 4:836579. [PMID: 35783597 PMCID: PMC9240309 DOI: 10.3389/fdgth.2022.836579] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/20/2022] [Indexed: 11/23/2022] Open
Abstract
During atherosclerosis, vascular smooth muscle cells (VSMCs) undergo a phenotypic transition from a healthy contractile state into pathological phenotypes including a proliferative and migratory, synthetic phenotype and osteochondrogenic-like phenotype that exacerbate plaques. Thus, inhibiting the transition of healthy, quiescent VSMCs to atherogenic cell types has the potential to mitigate atherosclerosis. To that end, previously, we reported that delivery of microRNA-145 (miR-145, a potent gatekeeper of the contractile VSMC phenotype) using nanoparticle micelles limited atherosclerotic plaque growth in murine models of atherosclerosis. Building on this preclinical data and toward clinical application, in this study, we tested the therapeutic viability of miR-145 micelles on patient-derived VSMCs and evaluated their effects based on disease severity. We collected vascular tissues from 11 patients with healthy, moderate, or severe stages of atherosclerosis that were discarded following vascular surgery or organ transplant, and isolated VSMCs from these tissues. We found that with increasing disease severity, patient-derived VSMCs had decreasing levels of contractile markers (miR-145, ACTA2, MYH11) and increasing levels of synthetic markers (KLF4, KLF5, and ELK1). Treatment with miR-145 micelles showed that an increase in disease severity correlated with a more robust response to therapy in VSMCs. Notably, miR-145 micelle therapy rescued contractile marker expression to baseline contractile levels in VSMCs derived from the most severely diseased tissues. As such, we demonstrate the use of miR-145 micelles across different stages of atherosclerosis disease and present further evidence of the translatability of miR-145 micelle treatment for atherosclerosis.
Collapse
Affiliation(s)
- Neil Patel
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States
| | - Deborah D. Chin
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States
| | - Gregory A. Magee
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, United States
- Department of Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- *Correspondence: Eun Ji Chung
| |
Collapse
|
11
|
Patel N, Chin DD, Chung EJ. Exosomes in Atherosclerosis, a Double-Edged Sword: Their Role in Disease Pathogenesis and Their Potential as Novel Therapeutics. AAPS JOURNAL 2021; 23:95. [PMID: 34312734 DOI: 10.1208/s12248-021-00621-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/29/2021] [Indexed: 12/23/2022]
Abstract
Cardiovascular disease (CAD) due to atherosclerosis is a major cause of death worldwide. The development of atherosclerosis involves intercellular communication facilitated by exosomes secreted from vascular endothelial cells (VECs), vascular smooth muscle cells (VSMCs), immune cells, and platelets. In this review, we summarize the current understanding of exosome biogenesis and uptake, and discuss atherogenic and atheroprotective functions of exosomes secreted from these cell types. In addition, we examine the potential of enhancing the therapeutic and targeting ability of exosomes exhibiting atheroprotective function by drug loading and surface modification with targeting ligands. We conclude with current challenges associated with exosome engineering for therapeutic use.
Collapse
Affiliation(s)
- Neil Patel
- Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, DRB 140, California, Los Angeles, 90089, USA
| | - Deborah D Chin
- Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, DRB 140, California, Los Angeles, 90089, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, DRB 140, California, Los Angeles, 90089, USA. .,Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Keck School of Medicine, University of Southern California, California, Los Angeles, 90033, USA. .,Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, California, Los Angeles, 90089, USA. .,Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, California, Los Angeles, 90033, USA.
| |
Collapse
|
12
|
He H, Hong K, Liu L, Schwendeman A. Artificial high-density lipoprotein-mimicking nanotherapeutics for the treatment of cardiovascular diseases. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1737. [PMID: 34263549 DOI: 10.1002/wnan.1737] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 01/08/2023]
Abstract
Despite the ability of current efficacious low-density lipoprotein-cholesterol-lowering therapies to reduce total cardiovascular disease (CVD) risks, CVD still poses major risks for morbidity and mortality to the general population. Because of the pleiotropic endothelial protective effects of high-density lipoproteins (HDL), the direct infusion of reconstituted HDL (rHDL) products, including MDCO-216, CER001, and CSL112, have been tested in clinical trials to determine whether direct infusion of rHDL can reduce coronary events in CVD patients. In addition to these rHDL products, in the past two decades, there has been an increased focused on designing artificial HDL-mimicking nanotherapeutics to produce complementary therapeutic strategies for CVD patients beyond lowering of atherogenic lipoproteins. Although recent reviews have comprehensively discussed the developments of artificial HDL-mimicking nanoparticles as therapeutics for CVD, there has been little assessment of "plain" or "drug-free" HDL-mimicking nanoparticles as therapeutics alone. In this review, we will summarize the clinical outcomes of rHDL products, examine recent advances in other types of artificial HDL-mimicking nanotherapeutics, including polymeric nanoparticles, cyclodextrins, micelles, metal nanoparticles, and so on; and potential new approaches for future CVD interventions. Moreover, success stories, lessons, and interpretations of the utility and functionality of these HDL-mimicking nanotherapeutics will be an integral part of this article. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Cardiovascular Disease.
Collapse
Affiliation(s)
- Hongliang He
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, USA.,State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering and Collaborative, Innovation Center of Suzhou Nano Science and Technology, Southeast University, Nanjing, China
| | - Kristen Hong
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, USA
| | - Lisha Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, USA.,Jiangsu Province Engineering Research Center for R&D and Evaluation of Intelligent Drugs and Key Functional Excipients, China Pharmaceutical University, Nanjing, China.,Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
13
|
Cheng M, Liu Q, Liu W, Yuan F, Feng J, Jin Y, Tu L. Engineering micelles for the treatment and diagnosis of atherosclerosis. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102473] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
14
|
Chen J, Zhang X, Millican R, Sherwood J, Martin S, Jo H, Yoon YS, Brott BC, Jun HW. Recent advances in nanomaterials for therapy and diagnosis for atherosclerosis. Adv Drug Deliv Rev 2021; 170:142-199. [PMID: 33428994 PMCID: PMC7981266 DOI: 10.1016/j.addr.2021.01.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/02/2021] [Accepted: 01/03/2021] [Indexed: 12/18/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease driven by lipid accumulation in arteries, leading to narrowing and thrombosis. It affects the heart, brain, and peripheral vessels and is the leading cause of mortality in the United States. Researchers have strived to design nanomaterials of various functions, ranging from non-invasive imaging contrast agents, targeted therapeutic delivery systems to multifunctional nanoagents able to target, diagnose, and treat atherosclerosis. Therefore, this review aims to summarize recent progress (2017-now) in the development of nanomaterials and their applications to improve atherosclerosis diagnosis and therapy during the preclinical and clinical stages of the disease.
Collapse
Affiliation(s)
- Jun Chen
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Xixi Zhang
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | | | | | - Sean Martin
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States; Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Young-Sup Yoon
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Brigitta C Brott
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ho-Wook Jun
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States.
| |
Collapse
|
15
|
Wang J, Chin D, Poon C, Mancino V, Pham J, Li H, Ho PY, Hallows KR, Chung EJ. Oral delivery of metformin by chitosan nanoparticles for polycystic kidney disease. J Control Release 2021; 329:1198-1209. [PMID: 33127449 PMCID: PMC7904655 DOI: 10.1016/j.jconrel.2020.10.047] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 12/15/2022]
Abstract
Nanoparticle drug delivery has many advantages over small molecule therapeutics, including reducing off-target side effects and increasing drug potency. However, many nanoparticles are administered parenterally, which is challenging for chronic diseases such as polycystic kidney disease (PKD), the most common hereditary disease worldwide in which patients need continuous treatment over decades. To address this clinical need, we present the development of nanoparticles synthesized from chitosan, a widely available polymer chosen for its ability to improve oral bioavailability. Specifically, we optimized the synthesis parameters of chitosan nanoparticles and demonstrate mucoadhesion and permeation across an intestinal barrier model in vitro. Furthermore, when administered orally to mice, ex vivo imaging of rhodamine-loaded chitosan nanoparticles showed significantly higher accumulation in the intestines compared to the free model drug, as well as 1.3 times higher serum area under the curve (AUC), demonstrating controlled release and improved serum delivery over 24 h. To test its utility for chronic diseases such as PKD, we loaded the candidate PKD drug, metformin, into chitosan nanoparticles, and upon oral administration to a PKD murine model (Pkd1fl/fl;Pax8-rtTA;Tet-O cre), a lower cyst burden was observed compared to free metformin, and was well tolerated upon repeated dosages. Blood urea nitrogen (BUN) and creatinine levels were similar to untreated mice, demonstrating kidney and biocompatibility health. Our study builds upon previous chitosan-based drug delivery approaches, and demonstrates a novel, oral nanoformulation for PKD.
Collapse
Affiliation(s)
- Jonathan Wang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Deborah Chin
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Christopher Poon
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Valeria Mancino
- Department of Medicine, Division of Nephrology and Hypertension, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jessica Pham
- Department of Medicine, Division of Nephrology and Hypertension, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Hui Li
- Department of Medicine, Division of Nephrology and Hypertension, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Pei-Yin Ho
- Department of Medicine, Division of Nephrology and Hypertension, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kenneth R Hallows
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA; Department of Medicine, Division of Nephrology and Hypertension, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA; Department of Medicine, Division of Nephrology and Hypertension, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA; Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA; Bridge Institute, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
16
|
Fender AC, Dobrev D. Contemporary plaque imaging for risk stratification of coronary artery disease: Are we getting there? IJC HEART & VASCULATURE 2020; 31:100678. [PMID: 33294585 PMCID: PMC7695964 DOI: 10.1016/j.ijcha.2020.100678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/06/2020] [Accepted: 11/08/2020] [Indexed: 11/03/2022]
Affiliation(s)
- Anke C. Fender
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
17
|
Trac N, Chen LY, Zhang A, Liao CP, Poon C, Wang J, Ando Y, Joo J, Garri C, Shen K, Kani K, Gross ME, Chung EJ. CCR2-targeted micelles for anti-cancer peptide delivery and immune stimulation. J Control Release 2020; 329:614-623. [PMID: 33011241 DOI: 10.1016/j.jconrel.2020.09.054] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/18/2020] [Accepted: 09/29/2020] [Indexed: 02/06/2023]
Abstract
Signaling between the CC chemokine receptor 2 (CCR2) with its ligand, monocyte chemoattractant protein-1 (MCP-1) promotes cancer progression by directly stimulating tumor cell proliferation and downregulating the expression of apoptotic proteins. Additionally, the MCP-1/CCR2 signaling axis drives the migration of circulating monocytes into the tumor microenvironment, where they mature into tumor-associated macrophages (TAMs) that promote disease progression through induction of angiogenesis, tissue remodeling, and suppression of the cytotoxic T lymphocyte (CTL) response. In order to simultaneously disrupt MCP-1/CCR2 signaling and target CCR2-expressing cancer cells for drug delivery, KLAK-MCP-1 micelles consisting of a CCR2-targeting peptide sequence (MCP-1 peptide) and the apoptotic KLAKLAK peptide were synthesized. In vitro, KLAK-MCP-1 micelles were observed to bind and induce cytotoxicity to cancer cells through interaction with CCR2. In vivo, KLAK-MCP-1 micelles inhibited tumor growth (34 ± 11%) in a subcutaneous B16F10 murine melanoma model despite minimal tumor accumulation upon intravenous injection. Tumors treated with KLAK-MCP1 demonstrated reduced intratumor CCR2 expression and altered infiltration of TAMs and CTLs as evidenced by immunohistochemical and flow cytometric analysis. These studies highlight the potential application of CCR2-targeted nanotherapeutic micelles in cancer treatment.
Collapse
Affiliation(s)
- Noah Trac
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, United States
| | - Leng-Ying Chen
- Lawrence J. Ellison Institute for Transformative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Ailin Zhang
- Lawrence J. Ellison Institute for Transformative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Chun-Peng Liao
- Lawrence J. Ellison Institute for Transformative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Christopher Poon
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, United States
| | - Jonathan Wang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, United States
| | - Yuta Ando
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, United States
| | - Johan Joo
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, United States
| | - Carolina Garri
- Lawrence J. Ellison Institute for Transformative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Keyue Shen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, United States; Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, United States
| | - Kian Kani
- Lawrence J. Ellison Institute for Transformative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States; Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, United States
| | - Mitchell E Gross
- Lawrence J. Ellison Institute for Transformative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States; Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, United States
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, United States; Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, United States; Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States; Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States; Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, United States; Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States.
| |
Collapse
|
18
|
Tripathy N, Wang J, Tung M, Conway C, Chung EJ. Transdermal Delivery of Kidney-Targeting Nanoparticles Using Dissolvable Microneedles. Cell Mol Bioeng 2020; 13:475-486. [PMID: 33184578 PMCID: PMC7596160 DOI: 10.1007/s12195-020-00622-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/20/2020] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Chronic kidney disease (CKD) affects approximately 13% of the world's population and will lead to dialysis or kidney transplantation. Unfortunately, clinically available drugs for CKD show limited efficacy and toxic extrarenal side effects. Hence, there is a need to develop targeted delivery systems with enhanced kidney specificity that can also be combined with a patient-compliant administration route for such patients that need extended treatment. Towards this goal, kidney-targeted nanoparticles administered through transdermal microneedles (KNP/MN) is explored in this study. METHODS A KNP/MN patch was developed by incorporating folate-conjugated micelle nanoparticles into polyvinyl alcohol MN patches. Rhodamine B (RhB) was encapsulated into KNP as a model drug and evaluated for biocompatibility and binding with human renal epithelial cells. For MN, skin penetration efficiency was assessed using a Parafilm model, and penetration was imaged via scanning electron microscopy. In vivo, KNP/MN patches were applied on the backs of C57BL/6 wild type mice and biodistribution, organ morphology, and kidney function assessed. RESULTS KNP showed high biocompatibility and folate-dependent binding in vitro, validating KNP's targeting to folate receptors in vitro. Upon transdermal administration in vivo, KNP/MN patches dissolved within 30 min. At varying time points up to 48 h post-KNP/MN administration, higher accumulation of KNP was found in kidneys compared with MN that consisted of the non-targeting, control-NP. Histological evaluation demonstrated no signs of tissue damage, and kidney function markers, serum blood urea nitrogen and urine creatinine, were found to be within normal ranges, indicating preservation of kidney health. CONCLUSIONS Our studies show potential of KNP/MN patches as a non-invasive, self-administrable platform to direct therapies to the kidneys.
Collapse
Affiliation(s)
- Nirmalya Tripathy
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA USA
| | - Jonathan Wang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA USA
| | - Madelynn Tung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA USA
| | - Claire Conway
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA USA
- Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA USA
- Department of Medicine, Division of Nephrology and Hypertension, University of Southern California, Los Angeles, CA USA
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, University of Southern California, Los Angeles, CA USA
| |
Collapse
|
19
|
Huang Y, Jiang K, Zhang X, Chung EJ. The effect of size, charge, and peptide ligand length on kidney targeting by small, organic nanoparticles. Bioeng Transl Med 2020; 5:e10173. [PMID: 33005739 PMCID: PMC7510478 DOI: 10.1002/btm2.10173] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/19/2020] [Accepted: 07/19/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease (CKD) affects 15% of the US adult population. However, most clinically available drugs for CKD show low bioavailability to the kidneys and non-specific uptake by other organs which results in adverse side effects. Hence, a targeted, drug delivery strategy to enhance kidney drug delivery is highly desired. Recently, our group developed small, organic nanoparticles called peptide amphiphile micelles (PAM) functionalized with the zwitterionic peptide ligand, (KKEEE)3K, that passage through the glomerular filtration barrier for kidney accumulation. Despite high bioavailability to the kidneys, these micelles also accumulated in the liver to a similar extent. To further optimize the physicochemical properties and develop design rules for kidney-targeting micelles, we synthesized a library of PAMs of varying size, charge, and peptide repeats. Specifically, variations of the original (KKEEE)3K peptide including (KKEEE)2K, (KKEEE)K, (EEKKK)3E, (EEKKK)2E, (EEKKK)E, KKKKK, and EEEEE were functionalized onto nanoparticles, and peptide surface density and PEG linker molecular weight were altered. After characterization with transmission electron microscopy (TEM) and dynamic light scattering (DLS), nanoparticles were intravenously administered into wildtype mice, and biodistribution was assessed through ex vivo imaging. All micelles localized to the kidneys, but nanoparticles that are positively-charged, close to the renal filtration size cut-off, and consisted of additional zwitterionic peptide sequences generally showed higher renal accumulation. Upon immunohistochemistry, micelles were confirmed to bind to the multiligand receptor, megalin, and histological analyses showed no tissue damage. Our study provides insight into the design of micelle carriers for kidney targeting and their potential for future therapeutic application.
Collapse
Affiliation(s)
- Yi Huang
- Department of Biomedical EngineeringUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Kairui Jiang
- Department of Biomedical EngineeringUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Xuting Zhang
- Department of Biomedical EngineeringUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Eun Ji Chung
- Department of Biomedical EngineeringUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Chemical Engineering and Materials ScienceUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Medicine, Division of Nephrology and HypertensionUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Surgery, Division of Vascular Surgery and Endovascular TherapyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
20
|
Wang J, Tripathy N, Chung EJ. Targeting and therapeutic peptide-based strategies for polycystic kidney disease. Adv Drug Deliv Rev 2020; 161-162:176-189. [PMID: 32866560 PMCID: PMC7736157 DOI: 10.1016/j.addr.2020.08.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/15/2020] [Accepted: 08/27/2020] [Indexed: 12/17/2022]
Abstract
Polycystic kidney disease (PKD) is characterized by progressive cyst growth and is a leading cause of renal failure worldwide. Currently, there are limited therapeutic options available to PKD patients, and only one drug, tolvaptan, has been FDA-approved to slow cyst progression. Similar to other small molecule drugs, however, tolvaptan is costly, only moderately effective, and causes adverse events leading to high patient dropout rates. Peptides may mitigate many drawbacks of small molecule drugs, as they can be highly tissue-specific, biocompatible, and economically scaled-up. Peptides can function as targeting ligands that direct therapies to diseased renal tissue, or be potent as therapeutic agents themselves. This review discusses various aberrant signaling pathways in PKD and renal receptors that can be potential targets of peptide-mediated strategies. Additionally, peptides utilized in other kidney applications, but may prove useful in the context of PKD, are highlighted. Insights into novel peptide-based solutions that have potential to improve clinical management of PKD are provided.
Collapse
Affiliation(s)
- Jonathan Wang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Nirmalya Tripathy
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA; Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA; Department of Medicine, Division of Nephrology and Hypertension, University of Southern California, Los Angeles, CA, USA; Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
21
|
Chin DD, Poon C, Trac N, Wang J, Cook J, Joo J, Jiang Z, Maria NSS, Jacobs RE, Chung EJ. Collagenase-Cleavable Peptide Amphiphile Micelles as a Novel Theranostic Strategy in Atherosclerosis. ADVANCED THERAPEUTICS 2020; 3:1900196. [PMID: 34295964 PMCID: PMC8294202 DOI: 10.1002/adtp.201900196] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Indexed: 11/10/2022]
Abstract
Atherosclerosis is an inflammatory disease characterized by plaques that can cause sudden myocardial infarction upon rupture. Such rupture-prone plaques have thin fibrous caps due to collagenase degradation, and a noninvasive diagnostic tool and targeted therapy that can identify and treat vulnerable plaques and may inhibit the onset of acute cardiac events. Toward this goal, monocyte-binding, collagenase-inhibiting, and gadolinium-modified peptide amphiphile micelles (MCG PAMs) are developed. Monocyte chemoattractant protein-1 (MCP-1) binds to C-C chemokine receptor-2 expressed on pathological cell types present within plaques. Through the peptide binding motif of MCP-1, MCG PAMs bind to monocytes and vascular smooth muscle cells in vitro. Moreover, using magnetic resonance imaging, MCG PAMs show enhanced targeting and successful detection of plaques in diseased mice in vivo and act as contrast agents for molecular imaging. Through the collagenase-cleaving peptide sequence of collagen [VPMS-MRGG], MCG PAMs can compete for collagenases that degrade the fibrous cap of plaques, providing therapy. MCG PAM-treated mice show increased fibrous cap thickness by 61% and 113% histologically compared to nontargeting micelle- or PBS-treated mice (p = 0.0075 and 0.001, respectively). Overall, this novel multimodal nanoparticle offers new theranostic opportunities for noninvasive diagnosis and treatment of atherosclerotic plaques.
Collapse
Affiliation(s)
- Deborah D Chin
- Department of Biomedical Engineering, University of Southern California, Los Angeles USC 90089 CA, USA
| | - Christopher Poon
- Department of Biomedical Engineering, University of Southern California, Los Angeles USC 90089 CA, USA
| | - Noah Trac
- Department of Biomedical Engineering, University of Southern California, Los Angeles USC 90089 CA, USA
| | - Jonathan Wang
- Department of Biomedical Engineering, University of Southern California, Los Angeles USC 90089 CA, USA
| | - Jackson Cook
- Department of Biomedical Engineering, University of Southern California, Los Angeles USC 90089 CA, USA
| | - Johan Joo
- Department of Biomedical Engineering, University of Southern California, Los Angeles USC 90089 CA, USA
| | - Zhangjingyi Jiang
- Department of Biomedical Engineering, University of Southern California, Los Angeles USC 90089 CA, USA
| | - Naomi Sulit Sta Maria
- Department of Physiology and Neuroscience, Zilkha Neurogenetic, Institute and Keck School of Medicine, University of Southern California, Los Angeles 90033 CA, USA
| | - Russell E Jacobs
- Department of Physiology and Neuroscience, Zilkha Neurogenetic, Institute and Keck School of Medicine, University of Southern California, Los Angeles 90033 CA, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles USC 90089 CA, USA
| |
Collapse
|