1
|
Long J, Liang X, Ao Z, Tang X, Li C, Yan K, Yu X, Wan Y, Li Y, Li C, Zhou M. Stimulus-responsive drug delivery nanoplatforms for inflammatory bowel disease therapy. Acta Biomater 2024; 188:27-47. [PMID: 39265673 DOI: 10.1016/j.actbio.2024.09.007] [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: 05/22/2024] [Revised: 08/26/2024] [Accepted: 09/06/2024] [Indexed: 09/14/2024]
Abstract
Inflammatory bowel disease (IBD) manifests as inflammation in the colon, rectum, and ileum, presenting a global health concern with increasing prevalence. Therefore, effective anti-inflammatory therapy stands as a promising strategy for the prevention and management of IBD. However, conventional nano drug delivery systems (NDDSs) for IBD face many challenges in targeting the intestine, such as physiological and pathological barriers, genetic variants, disease severity, and nutritional status, which often result in nonspecific tissue distribution and uncontrolled drug release. To address these limitations, stimulus-responsive NDDSs have received considerable attention in recent years due to their advantages in providing controlled release and enhanced targeting. This review provides an overview of the pathophysiological mechanisms underlying IBD and summarizes recent advancements in microenvironmental stimulus-responsive nanocarriers for IBD therapy. These carriers utilize physicochemical stimuli such as pH, reactive oxygen species, enzymes, and redox substances to deliver drugs for IBD treatment. Additionally, pivotal challenges in the future development and clinical translation of stimulus-responsive NDDSs are emphasized. By offering insights into the development and optimization of stimulus-responsive drug delivery nanoplatforms, this review aims to facilitate their application in treating IBD. STATEMENT OF SIGNIFICANCE: This review highlights recent advancements in stimulus-responsive nano drug delivery systems (NDDSs) for the treatment of inflammatory bowel disease (IBD). These innovative nanoplatforms respond to specific environmental triggers, such as pH reactive oxygen species, enzymes, and redox substances, to release drugs directly at the inflammation site. By summarizing the latest research, our work underscores the potential of these technologies to improve drug targeting and efficacy, offering new directions for IBD therapy. This review is significant as it provides a comprehensive overview for researchers and clinicians, facilitating the development of more effective treatments for IBD and other chronic inflammatory diseases.
Collapse
Affiliation(s)
- Jiang Long
- Department of Cardiology, Xuyong County People's Hospital, Luzhou, Sichuan 646000, China
| | - Xiaoya Liang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zuojin Ao
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiao Tang
- College of Integrated Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chuang Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Kexin Yan
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xin Yu
- Chinese Pharmacy Laboratory, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ying Wan
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yao Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; Science and Technology Department, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Meiling Zhou
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
2
|
Patel Y, Manturthi S, Tiwari S, Gahunia E, Courtemanche A, Gandelman M, Côté M, Gadde S. Development of Pro-resolving and Pro-efferocytic Nanoparticles for Atherosclerosis Therapy. ACS Pharmacol Transl Sci 2024; 7:3086-3095. [PMID: 39416959 PMCID: PMC11475319 DOI: 10.1021/acsptsci.4c00292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 10/19/2024]
Abstract
Atherosclerosis is a major contributor to cardiovascular diseases with a high global prevalence. It is characterized by the formation of lipid-laden plaques in the arteries, which eventually lead to plaque rupture and thrombosis. While the current lipid-lowering therapies are generally effective in lowering the risk of cardiovascular events, they do not address the underlying causes of disease. Defective resolution of inflammation and impaired efferocytosis are the main driving forces of atherosclerosis. Macrophages recognize cells for clearance by the expression of "eat me" and "do not eat me" signals, including the CD47-SIRPα axis. However, the "do not eat me" signal CD47 is overexpressed in atherosclerotic plaques, leading to compromised efferocytosis and secondary necrosis. In this context, prophagocytic antibodies have been explored to stimulate the clearance of apoptotic cells, but they are nonspecific and impact healthy tissues. In macrophages, downstream of signal regulatory protein α, lie protein tyrosine phosphatases, SHP 1/2, which can serve as effective targets for selectively phagocytosing apoptotic cells. While increasing the efferocytosis targets the end stages of lesion development, the underlying issue of inflammation still persists. Simultaneously increasing efferocytosis and reducing inflammation can be effective therapeutic strategies for managing atherosclerosis. For instance, IL-10 is a key anti-inflammatory mediator that enhances efferocytosis via phosphoSTAT3 (pSTAT3) activation. In this study, we developed a combination nanotherapy by encapsulating an SHP-1 inhibitor (NSC 87877) and IL-10 in a single nanoparticle platform [(S + IL)-NPs] to enhance efferocytosis and inflammation resolution. Our studies suggest that (S + IL)-NPs successfully encapsulated both agents, entered the macrophages, and delivered the agents into intracellular compartments. Additionally, (S + IL)-NPs decreased inflammation by suppressing pro-inflammatory markers and enhancing anti-inflammatory mediators. They also exhibited the potential for improved phagocytic activity via pSTAT3 activation. Our nanomedicine-mediated upregulation of the anti-inflammatory and efferocytic responses in macrophages shows promise for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yukta Patel
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research
Institute, Ottawa, Ontario K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Shireesha Manturthi
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research
Institute, Ottawa, Ontario K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Saras Tiwari
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research
Institute, Ottawa, Ontario K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Esha Gahunia
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research
Institute, Ottawa, Ontario K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Amandine Courtemanche
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research
Institute, Ottawa, Ontario K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Michelle Gandelman
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research
Institute, Ottawa, Ontario K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Marceline Côté
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Department
of Biochemistry Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Suresh Gadde
- Department
of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Kidney
Research Centre, Ottawa Hospital Research
Institute, Ottawa, Ontario K1H 8M5, Canada
- Ottawa
Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Centre
for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
- Ottawa−Carleton
Institute for Biomedical Engineering (OCIBME), Ottawa K1N 6N5, Canada
| |
Collapse
|
3
|
Imraish A, Thiab TA, Zihlif M, Al-Hunaiti A. Anti-Inflammatory and Antioxidant Potential of Green Synthesized Iron Zinc Oxide (Fe0.25-ZnO) Nanoparticles of the Elaeagnus angustifolia. Chem Biodivers 2024; 21:e202401060. [PMID: 38949232 DOI: 10.1002/cbdv.202401060] [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: 04/26/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/02/2024]
Abstract
Research interest in examining Elaeagnus angustifolia's potential as a source of anti-inflammatory and antioxidant agents has grown as a result of the plant's endorsement as a rich source of bioactive chemicals with promising anti-inflammatory and antioxidant activity. In this study, zinc oxide (Fe0.25-ZnO) bimetallic nanoparticles (E.ang-Fe0.25-ZnO NPs) were synthesized using an aqueous extract of Elaeagnus angustifolia. Synthesized Fe0.25-ZnO nanoparticles were characterized by FTIR and XRD. The anti-inflammatory and antioxidant activities were investigated in LPS-stimulated RAW 264.7 macrophages using RT-PCR and ELISA techniques for antioxidant- and inflammation-related genes. The concentration of 39.6 μg/ml of E.ang-Fe0.25-ZnO NPs demonstrated a significant anti-inflammatory activity by suppressing the mRNA levels of TNF-α and IL-6 by 88.3 %±1.9 and 93.6 %±0.1, respectively, compared to LPS-stimulated cells. This was confirmed by the significant reduction of TNF-α and IL-6 secretion levels from 95.2 and 495.6 pg/ml in LPS-stimulated cells to 5.6 and 26.5 pg/ml in E.ang-Fe0.25-ZnO treated group. In addition, E.ang-Fe0.25-ZnO NPs nanoparticles treatment significantly enhanced the expression of antioxidant-related genes, SOD and CAT. Together, our results proved that phyto-mediated Fe0.25-ZnO nanoparticles using Elaeagnus angustifolia have great potential in biomedical applications such as anti-inflammatory and antioxidant.
Collapse
Affiliation(s)
- Amer Imraish
- Department of Biological Sciences, School of Science, The University of Jordan, Queen Rania Al-Abdullah Street, Amman, 11942, Jordan
| | - Tuqa Abu Thiab
- Department of Biological Sciences, School of Science, The University of Jordan, Queen Rania Al-Abdullah Street, Amman, 11942, Jordan
| | - Malek Zihlif
- Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman, Jordan
| | - Afnan Al-Hunaiti
- Department of Chemistry, School of Science, The University of Jordan, Queen Rania Al-Abdullah Street, Amman, 11942, Jordan
| |
Collapse
|
4
|
He Z, Chen W, Hu K, Luo Y, Zeng W, He X, Li T, Ouyang J, Li Y, Xie L, Zhang Y, Xu Q, Yang S, Guo M, Zou W, Li Y, Huang L, Chen L, Zhang X, Saiding Q, Wang R, Zhang MR, Kong N, Xie T, Song X, Tao W. Resolvin D1 delivery to lesional macrophages using antioxidative black phosphorus nanosheets for atherosclerosis treatment. NATURE NANOTECHNOLOGY 2024; 19:1386-1398. [PMID: 38898135 DOI: 10.1038/s41565-024-01687-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 04/29/2024] [Indexed: 06/21/2024]
Abstract
The buildup of plaques in atherosclerosis leads to cardiovascular events, with chronic unresolved inflammation and overproduction of reactive oxygen species (ROS) being major drivers of plaque progression. Nanotherapeutics that can resolve inflammation and scavenge ROS have the potential to treat atherosclerosis. Here we demonstrate the potential of black phosphorus nanosheets (BPNSs) as a therapeutic agent for the treatment of atherosclerosis. BPNSs can effectively scavenge a broad spectrum of ROS and suppress atherosclerosis-associated pro-inflammatory cytokine production in lesional macrophages. We also demonstrate ROS-responsive, targeted-peptide-modified BPNS-based carriers for the delivery of resolvin D1 (an inflammation-resolving lipid mediator) to lesional macrophages, which further boosts the anti-atherosclerotic efficacy. The targeted nanotherapeutics not only reduce plaque areas but also substantially improve plaque stability in high-fat-diet-fed apolipoprotein E-deficient mice. This study presents a therapeutic strategy against atherosclerosis, and highlights the potential of BPNS-based therapeutics to treat other inflammatory diseases.
Collapse
Affiliation(s)
- Zhongshan He
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Kuan Hu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yaoyao Luo
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wanqin Zeng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University, Chengdu, China
| | - Xi He
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Tingting Li
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jiang Ouyang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lin Xie
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Yiding Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Qin Xu
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shuping Yang
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Mengran Guo
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zou
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yanfei Li
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lingjing Huang
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University, Chengdu, China
| | - Xingcai Zhang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tian Xie
- College of Pharmacy, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Xiangrong Song
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Park JE, Kim DH. Advanced Immunomodulatory Biomaterials for Therapeutic Applications. Adv Healthc Mater 2024:e2304496. [PMID: 38716543 DOI: 10.1002/adhm.202304496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 04/15/2024] [Indexed: 05/22/2024]
Abstract
The multifaceted biological defense system modulating complex immune responses against pathogens and foreign materials plays a critical role in tissue homeostasis and disease progression. Recently developed biomaterials that can specifically regulate immune responses, nanoparticles, graphene, and functional hydrogels have contributed to the advancement of tissue engineering as well as disease treatment. The interaction between innate and adaptive immunity, collectively determining immune responses, can be regulated by mechanobiological recognition and adaptation of immune cells to the extracellular microenvironment. Therefore, applying immunomodulation to tissue regeneration and cancer therapy involves manipulating the properties of biomaterials by tailoring their composition in the context of the immune system. This review provides a comprehensive overview of how the physicochemical attributes of biomaterials determine immune responses, focusing on the physical properties that influence innate and adaptive immunity. This review also underscores the critical aspect of biomaterial-based immune engineering for the development of novel therapeutics and emphasizes the importance of understanding the biomaterials-mediated immunological mechanisms and their role in modulating the immune system.
Collapse
Affiliation(s)
- Ji-Eun Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
- Department of Integrative Energy Engineering, College of Engineering, Korea University, Seoul, 02841, Republic of Korea
- Biomedical Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| |
Collapse
|
6
|
Beach M, Nayanathara U, Gao Y, Zhang C, Xiong Y, Wang Y, Such GK. Polymeric Nanoparticles for Drug Delivery. Chem Rev 2024; 124:5505-5616. [PMID: 38626459 PMCID: PMC11086401 DOI: 10.1021/acs.chemrev.3c00705] [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] [Indexed: 04/18/2024]
Abstract
The recent emergence of nanomedicine has revolutionized the therapeutic landscape and necessitated the creation of more sophisticated drug delivery systems. Polymeric nanoparticles sit at the forefront of numerous promising drug delivery designs, due to their unmatched control over physiochemical properties such as size, shape, architecture, charge, and surface functionality. Furthermore, polymeric nanoparticles have the ability to navigate various biological barriers to precisely target specific sites within the body, encapsulate a diverse range of therapeutic cargo and efficiently release this cargo in response to internal and external stimuli. However, despite these remarkable advantages, the presence of polymeric nanoparticles in wider clinical application is minimal. This review will provide a comprehensive understanding of polymeric nanoparticles as drug delivery vehicles. The biological barriers affecting drug delivery will be outlined first, followed by a comprehensive description of the various nanoparticle designs and preparation methods, beginning with the polymers on which they are based. The review will meticulously explore the current performance of polymeric nanoparticles against a myriad of diseases including cancer, viral and bacterial infections, before finally evaluating the advantages and crucial challenges that will determine their wider clinical potential in the decades to come.
Collapse
Affiliation(s)
- Maximilian
A. Beach
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Umeka Nayanathara
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yanting Gao
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Changhe Zhang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yijun Xiong
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yufu Wang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Georgina K. Such
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
7
|
Oyelaja O, Najneen T, Alamy H, Horn WL, Niño Medina JA, Duarte LE, Yaqobi A, Farooqi P, Mohammadi R, I Kh Almadhoun MK, Mia Khail B, Saeed A. Applications of Nanotechnology in the Field of Cardiology. Cureus 2024; 16:e58059. [PMID: 38738046 PMCID: PMC11088442 DOI: 10.7759/cureus.58059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2024] [Indexed: 05/14/2024] Open
Abstract
Cardiovascular diseases (CVDs) are a leading cause of death globally, demanding innovative therapeutic strategies. Nanoformulations, including nanoparticles, address challenges in drug delivery, stem cell therapy, imaging, and gene delivery. Nanoparticles enhance drug solubility, bioavailability, and targeted delivery, with gas microbubbles, liposomal preparations, and paramagnetic nanoparticles showing potential in treating atherosclerosis and reducing systemic side effects. In stem cell therapy, nanoparticles improve cell culture, utilizing three-dimensional nanofiber scaffolds and enhancing cardiomyocyte growth. Gold nanoparticles and poly(lactic-co-glycolic acid) (PLGA)-derived microparticles promote stem cell survival. Stem cell imaging utilizes direct labeling with nanoparticles for magnetic resonance imaging (MRI), while optical tracking employs dye-conjugated nanoparticles. In gene delivery, polymeric nanoparticles like polyethylenimine (PEI) and dendrimers, graphene-based carriers, and chitosan nanoparticles offer alternatives to virus-mediated gene transfer. The potential of magnetic nanoparticles in gene therapy is explored, particularly in hepatocellular carcinoma. Overall, nanoparticles have transformative potential in cardiovascular disease management, with ongoing research poised to enhance clinical outcomes.
Collapse
Affiliation(s)
- Oluwaseyi Oyelaja
- Medicine and Surgery, New York City Health and Hospitals Corporation (NYCHHC), New York, USA
| | - Tazkia Najneen
- Paediatrics, Dhaka Medical College and Hospital, Dhaka, BGD
| | - Haroon Alamy
- Internal Medicine, Armed Forces Science Academy, Kabul, AFG
| | - Wendys L Horn
- Health Sciences, University of Carabobo, Valencia, VEN
| | - Jose A Niño Medina
- Health Sciences, University of Carabobo, Valencia, VEN
- Law and Political Sciences, University of Carabobo, Valencia, VEN
| | | | - Adila Yaqobi
- Obstetrics and Gynaecology, Malalai Maternity Hospital, Kabul, AFG
| | - Palwasha Farooqi
- Internal Medicine, Kabul University of Medical Sciences, Kabul, AFG
| | | | | | | | - Abed Saeed
- Cardiovascular Medicine, Ali Abad Teaching Hospital, Kabul, AFG
| |
Collapse
|
8
|
Moisa SM, Burlacu A, Butnariu LI, Vasile CM, Brinza C, Spoiala EL, Maștaleru A, Leon MM, Rosu ST, Vatasescu R, Cinteză EE. Nanotechnology Innovations in Pediatric Cardiology and Cardiovascular Medicine: A Comprehensive Review. Biomedicines 2024; 12:185. [PMID: 38255290 PMCID: PMC10813221 DOI: 10.3390/biomedicines12010185] [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: 11/29/2023] [Revised: 01/09/2024] [Accepted: 01/13/2024] [Indexed: 01/24/2024] Open
Abstract
(1) Background: Nanomedicine, incorporating various nanoparticles and nanomaterials, offers significant potential in medical practice. Its clinical adoption, however, faces challenges like safety concerns, regulatory hurdles, and biocompatibility issues. Despite these, recent advancements have led to the approval of many nanotechnology-based products, including those for pediatric use. (2) Methods: Our approach included reviewing clinical, preclinical, and animal studies, as well as literature reviews from the past two decades and ongoing trials. (3) Results: Nanotechnology has introduced innovative solutions in cardiovascular care, particularly in managing myocardial ischemia. Key developments include drug-eluting stents, nitric oxide-releasing coatings, and the use of magnetic nanoparticles in cardiomyocyte transplantation. These advancements are pivotal for early detection and treatment. In cardiovascular imaging, nanotechnology enables noninvasive assessments. In pediatric cardiology, it holds promise in assisting the development of biological conduits, synthetic valves, and bioartificial grafts for congenital heart defects, and offers new treatments for conditions like dilated cardiomyopathy and pulmonary hypertension. (4) Conclusions: Nanomedicine presents groundbreaking solutions for cardiovascular diseases in both adults and children. It has the potential to transform cardiac care, from enhancing myocardial ischemia treatment and imaging techniques to addressing congenital heart issues. Further research and guideline development are crucial for optimizing its clinical application and revolutionizing patient care.
Collapse
Affiliation(s)
- Stefana Maria Moisa
- Department of Pediatrics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (E.L.S.)
- “Sfanta Maria” Clinical Emergency Hospital for Children, 700309 Iasi, Romania (S.T.R.)
| | - Alexandru Burlacu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Institute of Cardiovascular Diseases “Prof. Dr. George I.M. Georgescu”, 700503 Iasi, Romania
| | - Lacramioara Ionela Butnariu
- “Sfanta Maria” Clinical Emergency Hospital for Children, 700309 Iasi, Romania (S.T.R.)
- Department of Medical Genetics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Corina Maria Vasile
- Pediatric and Adult Congenital Cardiology Department, Centre Hospitalier Universitaire de Bordeaux, 33000 Bordeaux, France;
| | - Crischentian Brinza
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Institute of Cardiovascular Diseases “Prof. Dr. George I.M. Georgescu”, 700503 Iasi, Romania
| | - Elena Lia Spoiala
- Department of Pediatrics, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania (E.L.S.)
| | - Alexandra Maștaleru
- Department of Medical Specialties I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Maria Magdalena Leon
- Department of Medical Specialties I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Solange Tamara Rosu
- “Sfanta Maria” Clinical Emergency Hospital for Children, 700309 Iasi, Romania (S.T.R.)
- Department of Nursing, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Radu Vatasescu
- Cardio-Thoracic Department, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Clinical Emergency Hospital, 050098 Bucharest, Romania
| | - Eliza Elena Cinteză
- Department of Pediatrics, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Pediatric Cardiology, “Marie Curie” Emergency Children’s Hospital, 041451 Bucharest, Romania
| |
Collapse
|
9
|
Macvanin MT, Gluvic ZM, Klisic AN, Manojlovic MS, Suri JS, Rizzo M, Isenovic ER. The Link between miRNAs and PCKS9 in Atherosclerosis. Curr Med Chem 2024; 31:6926-6956. [PMID: 37990898 DOI: 10.2174/0109298673262124231102042914] [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: 05/12/2023] [Revised: 06/30/2023] [Accepted: 09/11/2023] [Indexed: 11/23/2023]
Abstract
Cardiovascular disease (CDV) represents the major cause of death globally. Atherosclerosis, as the primary cause of CVD, is a chronic immune-inflammatory disorder with complex multifactorial pathophysiology encompassing oxidative stress, enhanced immune-inflammatory cascade, endothelial dysfunction, and thrombosis. An initiating event in atherosclerosis is the subendothelial accumulation of low-density lipoprotein (LDL), followed by the localization of macrophages to fatty deposits on blood vessel walls, forming lipid-laden macrophages (foam cells) that secrete compounds involved in plaque formation. Given the fact that foam cells are one of the key culprits that underlie the pathophysiology of atherosclerosis, special attention has been paid to the investigation of the efficient therapeutic approach to overcome the dysregulation of metabolism of cholesterol in macrophages, decrease the foam cell formation and/or to force its degradation. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a secretory serine proteinase that has emerged as a significant regulator of the lipid metabolism pathway. PCSK9 activation leads to the degradation of LDL receptors (LDLRs), increasing LDL cholesterol (LDL-C) levels in the circulation. PCSK9 pathway dysregulation has been identified as one of the mechanisms involved in atherosclerosis. In addition, microRNAs (miRNAs) are investigated as important epigenetic factors in the pathophysiology of atherosclerosis and dysregulation of lipid metabolism. This review article summarizes the recent findings connecting the role of PCSK9 in atherosclerosis and the involvement of various miRNAs in regulating the expression of PCSK9-related genes. We also discuss PCSK9 pathway-targeting therapeutic interventions based on PCSK9 inhibition, and miRNA levels manipulation by therapeutic agents.
Collapse
Affiliation(s)
- Mirjana T Macvanin
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Zoran M Gluvic
- Department of Endocrinology and Diabetes, School of Medicine, University Clinical-Hospital Centre Zemun-Belgrade, Clinic of Internal Medicine, University of Belgrade, Belgrade, Serbia
| | - Aleksandra N Klisic
- Faculty of Medicine, Center for Laboratory Diagnostic, Primary Health Care Center, University of Montenegro, Podgorica, Montenegro
| | - Mia S Manojlovic
- Faculty of Medicine Novi Sad, University of Novi Sad, Novi Sad, Serbia
- Clinic for Endocrinology, Diabetes and Metabolic Disorders, Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Jasjit S Suri
- Stroke Monitoring and Diagnostic Division, Athero- Point™, Roseville, CA95661, USA
| | - Manfredi Rizzo
- Department of Health Promotion, School of Medicine, Mother and Child Care and Medical Specialties (Promise), University of Palermo, Palermo, Italy
| | - Esma R Isenovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
10
|
Mohammad-Rafiei F, Khojini JY, Ghazvinian F, Alimardan S, Norioun H, Tahershamsi Z, Tajbakhsh A, Gheibihayat SM. Cell membrane biomimetic nanoparticles in drug delivery. Biotechnol Appl Biochem 2023; 70:1843-1859. [PMID: 37387120 DOI: 10.1002/bab.2487] [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: 09/02/2022] [Accepted: 06/05/2023] [Indexed: 07/01/2023]
Abstract
Despite the efficiency of nanoparticle (NP) therapy, in vivo investigations have shown that it does not perform as well as in vitro. In this case, NP confronts many defensive hurdles once they enter the body. The delivery of NP to sick tissue is inhibited by these immune-mediated clearance mechanisms. Hence, using a cell membrane to hide NP for active distribution offers up a new path for focused treatment. These NPs are better able to reach the disease's target location, leading to enhanced therapeutic efficacy. In this emerging class of drug delivery vehicles, the inherent relation between the NPs and the biological components obtained from the human body was utilized, which mimic the properties and activities of native cells. This new technology has shown the viability of using biomimicry to evade immune system-provided biological barriers, with an emphasis on restricting clearance from the body before reaching its intended target. Furthermore, by providing signaling cues and transplanted biological components that favorably change the intrinsic immune response at the disease site, the NPs would be capable interacting with immune cells regarding the biomimetic method. Thus, we aimed to provide a current landscape and future trends of biomimetic NPs in drug delivery.
Collapse
Affiliation(s)
- Fatemeh Mohammad-Rafiei
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Javad Yaghmoorian Khojini
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Ghazvinian
- Department of Life science and biotechnology, Faculty of Natural Sciences, University of Shahid Beheshti, Tehran, Iran
| | - Sajad Alimardan
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamid Norioun
- Medical Genetics Department, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Zahra Tahershamsi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Munich, Germany
| |
Collapse
|
11
|
Matos AI, Peres C, Carreira B, Moura LIF, Acúrcio RC, Vogel T, Wegener E, Ribeiro F, Afonso MB, Santos FMF, Martínez‐Barriocanal Á, Arango D, Viana AS, Góis PMP, Silva LC, Rodrigues CMP, Graca L, Jordan R, Satchi‐Fainaro R, Florindo HF. Polyoxazoline-Based Nanovaccine Synergizes with Tumor-Associated Macrophage Targeting and Anti-PD-1 Immunotherapy against Solid Tumors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300299. [PMID: 37434063 PMCID: PMC10477894 DOI: 10.1002/advs.202300299] [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: 01/13/2023] [Revised: 06/22/2023] [Indexed: 07/13/2023]
Abstract
Immune checkpoint blockade reaches remarkable clinical responses. However, even in the most favorable cases, half of these patients do not benefit from these therapies in the long term. It is hypothesized that the activation of host immunity by co-delivering peptide antigens, adjuvants, and regulators of the transforming growth factor (TGF)-β expression using a polyoxazoline (POx)-poly(lactic-co-glycolic) acid (PLGA) nanovaccine, while modulating the tumor-associated macrophages (TAM) function within the tumor microenvironment (TME) and blocking the anti-programmed cell death protein 1 (PD-1) can constitute an alternative approach for cancer immunotherapy. POx-Mannose (Man) nanovaccines generate antigen-specific T-cell responses that control tumor growth to a higher extent than poly(ethylene glycol) (PEG)-Man nanovaccines. This anti-tumor effect induced by the POx-Man nanovaccines is mediated by a CD8+ -T cell-dependent mechanism, in contrast to the PEG-Man nanovaccines. POx-Man nanovaccine combines with pexidartinib, a modulator of the TAM function, restricts the MC38 tumor growth, and synergizes with PD-1 blockade, controlling MC38 and CT26 tumor growth and survival. This data is further validated in the highly aggressive and poorly immunogenic B16F10 melanoma mouse model. Therefore, the synergistic anti-tumor effect induced by the combination of nanovaccines with the inhibition of both TAM- and PD-1-inducing immunosuppression, holds great potential for improving immunotherapy outcomes in solid cancer patients.
Collapse
Affiliation(s)
- Ana I. Matos
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Lisbon Academic Medical CenterUniversidade de LisboaLisbon1649‐028Portugal
| | - Carina Peres
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Lisbon Academic Medical CenterUniversidade de LisboaLisbon1649‐028Portugal
| | - Barbara Carreira
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Liane I. F. Moura
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Rita C. Acúrcio
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Theresa Vogel
- Department of Chemistry, Faculty of Chemistry and Food Chemistry, School of ScienceTechnische Universität Dresden01062DresdenGermany
| | - Erik Wegener
- Department of Chemistry, Faculty of Chemistry and Food Chemistry, School of ScienceTechnische Universität Dresden01062DresdenGermany
| | - Filipa Ribeiro
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Lisbon Academic Medical CenterUniversidade de LisboaLisbon1649‐028Portugal
| | - Marta B. Afonso
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Fábio M. F. Santos
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Águeda Martínez‐Barriocanal
- Group of Biomedical Research in Digestive Tract TumorsCIBBIM‐NanomedicineVall d'Hebron Research Institute (VHIR)Universitat Autònoma de Barcelona (UAB)Barcelona08035Spain
- Group of Molecular OncologyLleida Biomedical Research Institute (IRBLleida)Lleida25198Spain
| | - Diego Arango
- Group of Biomedical Research in Digestive Tract TumorsCIBBIM‐NanomedicineVall d'Hebron Research Institute (VHIR)Universitat Autònoma de Barcelona (UAB)Barcelona08035Spain
- Group of Molecular OncologyLleida Biomedical Research Institute (IRBLleida)Lleida25198Spain
| | - Ana S. Viana
- Centro de Química EstruturalDepartamento de Química e BioquímicaInstitute of Molecular SciencesFaculty of SciencesUniversidade de LisboaLisbon1749‐016Portugal
| | - Pedro M. P. Góis
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Liana C. Silva
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Cecília M. P. Rodrigues
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| | - Luis Graca
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Lisbon Academic Medical CenterUniversidade de LisboaLisbon1649‐028Portugal
| | - Rainer Jordan
- Department of Chemistry, Faculty of Chemistry and Food Chemistry, School of ScienceTechnische Universität Dresden01062DresdenGermany
| | - Ronit Satchi‐Fainaro
- Department of Physiology and PharmacologyFaculty of MedicineSagol School of NeuroscienceTel Aviv UniversityTel Aviv69978Israel
| | - Helena F. Florindo
- Grouf of BioNanoSciences ‐ Drug Delivery and Immunoengineering, Research Institute for Medicines (iMed.ULisboa), Department of Pharmacy, Pharmacology and Health TechnologiesFaculty of PharmacyUniversidade de LisboaLisbon1649‐003Portugal
| |
Collapse
|
12
|
Novorolsky RJ, Kasheke GDS, Hakim A, Foldvari M, Dorighello GG, Sekler I, Vuligonda V, Sanders ME, Renden RB, Wilson JJ, Robertson GS. Preserving and enhancing mitochondrial function after stroke to protect and repair the neurovascular unit: novel opportunities for nanoparticle-based drug delivery. Front Cell Neurosci 2023; 17:1226630. [PMID: 37484823 PMCID: PMC10360135 DOI: 10.3389/fncel.2023.1226630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/22/2023] [Indexed: 07/25/2023] Open
Abstract
The neurovascular unit (NVU) is composed of vascular cells, glia, and neurons that form the basic component of the blood brain barrier. This intricate structure rapidly adjusts cerebral blood flow to match the metabolic needs of brain activity. However, the NVU is exquisitely sensitive to damage and displays limited repair after a stroke. To effectively treat stroke, it is therefore considered crucial to both protect and repair the NVU. Mitochondrial calcium (Ca2+) uptake supports NVU function by buffering Ca2+ and stimulating energy production. However, excessive mitochondrial Ca2+ uptake causes toxic mitochondrial Ca2+ overloading that triggers numerous cell death pathways which destroy the NVU. Mitochondrial damage is one of the earliest pathological events in stroke. Drugs that preserve mitochondrial integrity and function should therefore confer profound NVU protection by blocking the initiation of numerous injury events. We have shown that mitochondrial Ca2+ uptake and efflux in the brain are mediated by the mitochondrial Ca2+ uniporter complex (MCUcx) and sodium/Ca2+/lithium exchanger (NCLX), respectively. Moreover, our recent pharmacological studies have demonstrated that MCUcx inhibition and NCLX activation suppress ischemic and excitotoxic neuronal cell death by blocking mitochondrial Ca2+ overloading. These findings suggest that combining MCUcx inhibition with NCLX activation should markedly protect the NVU. In terms of promoting NVU repair, nuclear hormone receptor activation is a promising approach. Retinoid X receptor (RXR) and thyroid hormone receptor (TR) agonists activate complementary transcriptional programs that stimulate mitochondrial biogenesis, suppress inflammation, and enhance the production of new vascular cells, glia, and neurons. RXR and TR agonism should thus further improve the clinical benefits of MCUcx inhibition and NCLX activation by increasing NVU repair. However, drugs that either inhibit the MCUcx, or stimulate the NCLX, or activate the RXR or TR, suffer from adverse effects caused by undesired actions on healthy tissues. To overcome this problem, we describe the use of nanoparticle drug formulations that preferentially target metabolically compromised and damaged NVUs after an ischemic or hemorrhagic stroke. These nanoparticle-based approaches have the potential to improve clinical safety and efficacy by maximizing drug delivery to diseased NVUs and minimizing drug exposure in healthy brain and peripheral tissues.
Collapse
Affiliation(s)
- Robyn J. Novorolsky
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Brain Repair Centre, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Gracious D. S. Kasheke
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Brain Repair Centre, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Antoine Hakim
- School of Pharmacy, Faculty of Science, University of Waterloo, Waterloo, ON, Canada
| | - Marianna Foldvari
- School of Pharmacy, Faculty of Science, University of Waterloo, Waterloo, ON, Canada
| | - Gabriel G. Dorighello
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Brain Repair Centre, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Israel Sekler
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben Gurion University, Beersheva, Israel
| | | | | | - Robert B. Renden
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, United States
| | - Justin J. Wilson
- Department of Chemistry and Chemical Biology, College of Arts and Sciences, Cornell University, Ithaca, NY, United States
| | - George S. Robertson
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Brain Repair Centre, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Department of Psychiatry, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
13
|
Wang X, Xia Z, Wang H, Wang D, Sun T, Hossain E, Pang X, Liu Y. Cell-membrane-coated nanoparticles for the fight against pathogenic bacteria, toxins, and inflammatory cytokines associated with sepsis. Theranostics 2023; 13:3224-3244. [PMID: 37351162 PMCID: PMC10283065 DOI: 10.7150/thno.81520] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 05/05/2023] [Indexed: 06/24/2023] Open
Abstract
Sepsis is the main cause of death in patients suffering from serious illness. Yet, there is still no specific treatment for sepsis, and management relies on infection control. Cell membrane-coated nanoparticles (MNPs) are a new class of biomimetic nanoparticles based on covering the surface of synthetic nanoparticles (NPs) with natural cell membranes. They retain the physicochemical properties of synthetic nanomaterials and inherit the specific properties of cellular membranes, showing excellent biological compatibility, enhanced biointerfacing capabilities, capacity to hold cellular functions and characteristics, immunological escape, and longer half-life when in circulation. Additionally, they prevent the decomposition of the encapsulated drug and active targeting. Over the years, studies on MNPs have multiplied and a breakthrough has been achieved for cancer therapy. Nevertheless, the use of "bio"-related approaches is still rare for treating sepsis. Herein, we discussed current state-of-the-art on MNPs for the treatment of bacterial sepsis by combining the pathophysiology and therapeutic benefits of sepsis, i.e., pathogenic bacteria, bacteria-producing toxins, and inflammatory cytokines produced in the dysregulated inflammatory response associated with sepsis.
Collapse
Affiliation(s)
- Xiaoyi Wang
- Henan Key Laboratory of Pediatric Hematology and Oncology Medicine, Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zongping Xia
- Department of Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Huaili Wang
- Henan Key Laboratory of Pediatric Hematology and Oncology Medicine, Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Dao Wang
- Henan Key Laboratory of Pediatric Hematology and Oncology Medicine, Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Tongwen Sun
- Department of Integrated ICU, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Eamran Hossain
- Henan Key Laboratory of Pediatric Hematology and Oncology Medicine, Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xin Pang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yufeng Liu
- Henan Key Laboratory of Pediatric Hematology and Oncology Medicine, Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
14
|
Voicu G, Mocanu CA, Safciuc F, Anghelache M, Deleanu M, Cecoltan S, Pinteala M, Uritu CM, Droc I, Simionescu M, Manduteanu I, Calin M. Nanocarriers of shRNA-Runx2 directed to collagen IV as a nanotherapeutic system to target calcific aortic valve disease. Mater Today Bio 2023; 20:100620. [PMID: 37063777 PMCID: PMC10102408 DOI: 10.1016/j.mtbio.2023.100620] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023] Open
Abstract
Runx2 is a key transcription factor involved in valvular interstitial cells (VIC) osteodifferentiation, a process actively entwined with the calcific aortic valve disease (CAVD). We hypothesize that a strategy intended to silence Runx2 could be a valuable novel therapeutic option for CAVD. To this intent, we aimed at (i) developing targeted nanoparticles for efficient delivery of short hairpin (sh)RNA sequences specific for Runx2 to the aortic valve employing a relevant mouse model for CAVD and (ii) investigate their therapeutic potential in osteoblast-differentiated VIC (oVIC) cultivated into a 3D scaffold. Since collagen IV was used as a target, a peptide that binds specifically to collagen IV (Cp) was conjugated to the surface of lipopolyplexes encapsulating shRNA-Runx2 (Cp-LPP/shRunx2). The results showed that Cp-LPP/shRunx2 were (i) cytocompatible; (ii) efficiently taken up by 3D-cultured oVIC; (iii) diminished the osteodifferentiation of human VIC (cultured in a 3D hydrogel-derived from native aortic root) by reducing osteogenic molecules expression, alkaline phosphatase activity, and calcium concentration; and (iv) were recruited in aortic valve leaflets in a murine model of atherosclerosis. Taken together, these data recommend Cp-LPP/shRunx2 as a novel targeted nanotherapy to block the progression of CAVD, with a good perspective to be introduced in practical use.
Collapse
Affiliation(s)
- Geanina Voicu
- “Medical and Pharmaceutical Bionanotechnologies” Laboratory, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568, Bucharest, Romania
| | - Cristina Ana Mocanu
- “Medical and Pharmaceutical Bionanotechnologies” Laboratory, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568, Bucharest, Romania
| | - Florentina Safciuc
- “Medical and Pharmaceutical Bionanotechnologies” Laboratory, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568, Bucharest, Romania
| | - Maria Anghelache
- “Medical and Pharmaceutical Bionanotechnologies” Laboratory, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568, Bucharest, Romania
| | - Mariana Deleanu
- “Liquid and Gas Chromatography” Laboratory, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568, Bucharest, Romania
| | - Sergiu Cecoltan
- “Medical and Pharmaceutical Bionanotechnologies” Laboratory, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568, Bucharest, Romania
| | - Mariana Pinteala
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, “Petru Poni” Institute of Macromolecular Chemistry, 700487, Iasi, Romania
| | - Cristina Mariana Uritu
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, “Petru Poni” Institute of Macromolecular Chemistry, 700487, Iasi, Romania
- Advanced Centre for Research-Development in Experimental Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 700115, Iasi, Romania
| | - Ionel Droc
- Central Military Hospital “Dr. Carol Davila”, Cardiovascular Surgery Clinic, Bucharest, Romania
| | - Maya Simionescu
- “Medical and Pharmaceutical Bionanotechnologies” Laboratory, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568, Bucharest, Romania
| | - Ileana Manduteanu
- “Medical and Pharmaceutical Bionanotechnologies” Laboratory, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568, Bucharest, Romania
| | - Manuela Calin
- “Medical and Pharmaceutical Bionanotechnologies” Laboratory, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568, Bucharest, Romania
- Corresponding author. “Medical and Pharmaceutical Bionanotechnologies” Laboratory, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568, Bucharest, Romania.
| |
Collapse
|
15
|
Xu B, Ye J, Fan BS, Wang X, Zhang JY, Song S, Song Y, Jiang WB, Wang X, Yu JK. Protein-spatiotemporal partition releasing gradient porous scaffolds and anti-inflammatory and antioxidant regulation remodel tissue engineered anisotropic meniscus. Bioact Mater 2023; 20:194-207. [PMID: 35702607 PMCID: PMC9160676 DOI: 10.1016/j.bioactmat.2022.05.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 01/02/2023] Open
Abstract
Meniscus is a wedge-shaped fibrocartilaginous tissue, playing important roles in maintaining joint stability and function. Meniscus injuries are difficult to heal and frequently progress into structural breakdown, which then leads to osteoarthritis. Regeneration of heterogeneous tissue engineering meniscus (TEM) continues to be a scientific and translational challenge. The morphology, tissue architecture, mechanical strength, and functional applications of the cultivated TEMs have not been able to meet clinical needs, which may due to the negligent attention on the importance of microenvironment in vitro and in vivo. Herein, we combined the 3D (three-dimensional)-printed gradient porous scaffolds, spatiotemporal partition release of growth factors, and anti-inflammatory and anti-oxidant microenvironment regulation of Ac2-26 peptide to prepare a versatile meniscus composite scaffold with heterogeneous bionic structures, excellent biomechanical properties and anti-inflammatory and anti-oxidant effects. By observing the results of cell activity and differentiation, and biomechanics under anti-inflammatory and anti-oxidant microenvironments in vitro, we explored the effects of anti-inflammatory and anti-oxidant microenvironments on construction of regional and functional heterogeneous TEM via the growth process regulation, with a view to cultivating a high-quality of TEM from bench to bedside. A polycaprolactone meniscus scaffold with the gradient porous architecture. Spatiotemporal partition release of two growth factors to promote heterogeneous phenotypes. Anti-inflammatory and antioxidant regulation by Ac2-26 peptide. Scaffold with biomimetic morphology, biomechanics, heterogeneity of native meniscus.
Collapse
|
16
|
Gamma irradiation mediated production improvement of some myco-fabricated nanoparticles and exploring their wound healing, anti-inflammatory and acetylcholinesterase inhibitory potentials. Sci Rep 2023; 13:1629. [PMID: 36717680 PMCID: PMC9887004 DOI: 10.1038/s41598-023-28670-5] [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: 10/01/2022] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
In the current scenario, scaling up the microbial production of nanoparticles with diverse biological applications is an emerging prospect for NPs' sustainable industry. Thus, this paper was conducted to develop a suitable applicative process for the myco-fabrication of cobalt-ferrite (CoFeNPs), selenium (SeNPs), and zinc oxide (ZnONPs) nanoparticles. A strain improvement program using gamma irradiation mutagenesis was applied to improve the NPs-producing ability of the fungal strains. The achieved yields of CoFeNPs, SeNPs, and ZnONPs were intensified by a 14.47, 7.85, and 22.25-fold increase from the initial yield following gamma irradiation and isolation of stable mutant strains. The myco-fabricated CoFeNPs, SeNPs, and ZnONPs were then exploited to study their wound healing, and anti-inflammatory. In addition, the acetylcholinesterase inhibition activities of the myco-fabricated NPs were evaluated and analyzed by molecular docking. The obtained results confirmed the promising wound healing, anti-inflammatory, and acetylcholinesterase inhibition potentials of the three types of NPs. Additionally, data from analyzing the interaction of NPs with acetylcholinesterase enzyme by molecular docking were in conformation with the experimental data.
Collapse
|
17
|
Nie Q, Li C, Wang Y, Hu Y, Pu W, Zhang Q, Cai J, Lin Y, Li G, Wang C, Li L, Dou Y, Zhang J. Pathologically triggered in situ aggregation of nanoparticles for inflammation-targeting amplification and therapeutic potentiation. Acta Pharm Sin B 2023; 13:390-409. [PMID: 36815041 PMCID: PMC9939322 DOI: 10.1016/j.apsb.2022.07.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/24/2022] [Accepted: 04/28/2022] [Indexed: 12/01/2022] Open
Abstract
Uncontrolled and persistent inflammation is closely related to numerous acute and chronic diseases. However, effective targeting delivery systems remain to be developed for precision therapy of inflammatory diseases. Herein we report a novel strategy for engineering inflammation-accumulation nanoparticles via phenolic functionalization. Different phenol-functionalized nanoparticles were first developed, which can undergo in situ aggregation upon triggering by the inflammatory/oxidative microenvironment. Phenolic compound-decorated poly (lactide-co-glycolide) nanoparticles, in particular tyramine (Tyr)-coated nanoparticles, showed significantly enhanced accumulation at inflammatory sites in mouse models of colitis, acute liver injury, and acute lung injury, mainly resulting from in situ cross-linking and tissue anchoring of nanoparticles triggered by local myeloperoxidase and reactive oxygen species. By combining a cyclodextrin-derived bioactive material with Tyr decoration, a multifunctional nanotherapy (TTN) was further developed, which displayed enhanced cellular uptake, anti-inflammatory activities, and inflammatory tissue accumulation, thereby affording amplified therapeutic effects in mice with colitis or acute liver injury. Moreover, TTN can serve as a bioactive and inflammation-targeting nanoplatform for site-specifically delivering a therapeutic peptide to the inflamed colon post oral administration, leading to considerably potentiated in vivo efficacies. Preliminary studies also revealed good safety of orally delivered TTN. Consequently, Tyr-based functionalization is promising for inflammation targeting amplification and therapeutic potentiation of nanotherapies.
Collapse
Affiliation(s)
- Qiang Nie
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chenwen Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yu Wang
- Department of Radiology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing 400030, China
| | - Yi Hu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wendan Pu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qixiong Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jiajun Cai
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yongyao Lin
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Gang Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chenping Wang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lanlan Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yin Dou
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China,State Key Lab of Trauma, Burn and Combined Injury, Institute of Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China,Corresponding author. Tel.: +86 23 68771637.
| |
Collapse
|
18
|
Park M, Oh HJ, Han J, Hong SH, Park W, Song H. Liposome-mediated small RNA delivery to convert the macrophage polarity: A novel therapeutic approach to treat inflammatory uterine disease. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 30:663-676. [PMID: 36569217 PMCID: PMC9758500 DOI: 10.1016/j.omtn.2022.11.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
Macrophages are present in all tissues for maintaining tissue homeostasis, and macrophage polarization plays a vital role in alleviating inflammation. Therefore, specific delivery of polarization modulators to macrophages in situ is critical for treating inflammatory diseases. We demonstrate that a size-controlled miRNA-encapsulated macrophage-targeting liposomes (miR/MT-Lip) specifically targets macrophages to promote M1-to-M2 polarization conversion, alleviating inflammation without cytotoxicity. miR/MT-Lip, approximately 1.2 μm, showed excellent internalization through phagocytosis and/or macropinocytosis in macrophages. miR-10a/MT-Lip, but not scramble miR-Fluorescein amidite (FAM)/MT-Lip as control, effectively converted the polarization of lipopolysaccharide (LPS)-induced M1 macrophages to M2 in vitro. When miR-10a/MT-Lip was intravenously delivered to mice insulted with LPS for inflammation, the proportion of M2 macrophages was significantly increased without disturbing the population of other immune cells. Furthermore, scramble miR-FAM/MT-Lip was mainly detected in macrophages, but not other immune cells. When our miR/MT-Lip was administered to mice with Asherman's syndrome that suffer from infertility because of sterile uterine inflammation, macrophage-specific targeting of miR-10a/MT-Lip facilitated M1-to-M2 conversion for angiogenesis in the impaired uterus, resulting in restoration of healthy uterine conditions. The results indicate that our MT-Lip encapsulating small RNAs has excellent potential to treat various inflammatory disorders by fine-tuning macrophage polarization in vivo without any side effects.
Collapse
Affiliation(s)
- Mira Park
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam, Gyeonggi 13488, Republic of Korea
| | - Hyeon-Ji Oh
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi 16419, Republic of Korea
| | - Jieun Han
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi 16419, Republic of Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, Kangwon National University, Chuncheon, Kangwon 24341, Republic of Korea
| | - Wooram Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi 16419, Republic of Korea,Corresponding author Wooram Park, PhD, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi 16419, Republic of Korea.
| | - Haengseok Song
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam, Gyeonggi 13488, Republic of Korea,Corresponding author Haengseok Song, PhD, Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam, Gyeonggi 13488, Republic of Korea.
| |
Collapse
|
19
|
Ciciriello AJ, Surnar B, Medy GD, Su X, Dhar S, Dumont CM. Biomaterial-targeted precision nanoparticle delivery to the injured spinal cord. Acta Biomater 2022; 152:532-545. [PMID: 36087868 PMCID: PMC10551882 DOI: 10.1016/j.actbio.2022.08.077] [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: 05/04/2022] [Revised: 07/29/2022] [Accepted: 08/31/2022] [Indexed: 11/01/2022]
Abstract
Drug delivery requires precision in timing, location, and dosage to achieve therapeutic benefits. Challenges in addressing all three of these critical criteria result in poor temporal dexterity, widespread accumulation and off-target effects, and high doses with the potential for toxicity. To address these challenges, we have developed the BiomatErial Accumulating Carriers for On-demand Nanotherapy (BEACON) platform that utilizes an implantable biomaterial to serve as a target for systemically delivered nanoparticles (NPs). With the BEACON system, administered NPs are conjugated with a ligand that has high affinity for a receptor in the implanted biomaterial. To test BEACON, an in vivo spinal cord injury (SCI) model was used as it provides an injury model where the three identified criteria can be tested as it is a dynamic and complicated injury model with no currently approved therapies. Through our work, we have demonstrated temporal dexterity in NP administration by injecting 6 days post-SCI, decreased off-target accumulation with a significant drop in liver accumulation, and retention of our NPs in the target biomaterial. The BEACON system can be applied broadly, beyond the nervous system, to improve systemically delivered NP accumulation at an implanted biomaterial target. STATEMENT OF SIGNIFICANCE: Targeted drug delivery approaches have the potential to improve therapeutic regimens for patients on a case-by-case basis. Improved localization of a therapeutic to site of interest can result in increased efficacy and limit the need for repeat dosing. Unfortunately, targeted strategies can fall short when receptors on cells or tissues are too widespread or change over the course of disease or injury progression. The BEACON system developed herein eliminates the need to target a cell or tissue receptor by targeting an implantable biomaterial with location-controllable accumulation and sustained presentation over time. The targeting paradigm presented by BEACON is widely applicable throughout tissue engineering and regenerative medicine without the need to retool for each new application.
Collapse
Affiliation(s)
- Andrew J Ciciriello
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL 33146, United States; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW 7th Avenue, Miami, Florida 33136, United States
| | - Bapurao Surnar
- Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW 7th Avenue, Miami, Florida 33136, United States; Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States
| | - Giovanni D Medy
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL 33146, United States
| | - Xiaoyu Su
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL 33146, United States
| | - Shanta Dhar
- Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW 7th Avenue, Miami, Florida 33136, United States; Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States; Sylvester Comprehensive Cancer Center, University of Miami, 1475 NW 12th Avenue, Miami, Florida 33136, United States
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL 33146, United States; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW 7th Avenue, Miami, Florida 33136, United States; Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States.
| |
Collapse
|
20
|
Hetherington I, Totary-Jain H. Anti-atherosclerotic therapies: Milestones, challenges, and emerging innovations. Mol Ther 2022; 30:3106-3117. [PMID: 36065464 PMCID: PMC9552812 DOI: 10.1016/j.ymthe.2022.08.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/22/2022] Open
Abstract
Atherosclerosis is the main underlying pathology for many cardiovascular diseases (CVDs), which are the leading cause of death globally and represent a serious health crisis. Atherosclerosis is a chronic condition that can lead to myocardial infarction, ischemic cardiomyopathy, stroke, and peripheral arterial disease. Elevated plasma lipids, hypertension, and high glucose are the major risk factors for developing atherosclerotic plaques. To date, most pharmacological therapies aim to control these risk factors, but they do not target the plaque-causing cells themselves. In patients with acute coronary syndromes, surgical revascularization with percutaneous coronary intervention has greatly reduced mortality rates. However, stent thrombosis and neo-atherosclerosis have emerged as major safety concerns of drug eluting stents due to delayed re-endothelialization. This review summarizes the major milestones, strengths, and limitations of current anti-atherosclerotic therapies. It provides an overview of the recent discoveries and emerging game-changing technologies in the fields of nanomedicine, mRNA therapeutics, and gene editing that have the potential to revolutionize CVD clinical practice by steering it toward precision medicine.
Collapse
Affiliation(s)
- Isabella Hetherington
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., MDC08, 2170, Tampa, FL 33612, USA
| | - Hana Totary-Jain
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., MDC08, 2170, Tampa, FL 33612, USA.
| |
Collapse
|
21
|
Qin CX, Norling LV, Vecchio EA, Brennan EP, May LT, Wootten D, Godson C, Perretti M, Ritchie RH. Formylpeptide receptor 2: Nomenclature, structure, signalling and translational perspectives: IUPHAR review 35. Br J Pharmacol 2022; 179:4617-4639. [PMID: 35797341 PMCID: PMC9545948 DOI: 10.1111/bph.15919] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 05/22/2022] [Accepted: 06/09/2022] [Indexed: 12/26/2022] Open
Abstract
We discuss the fascinating pharmacology of formylpeptide receptor 2 (FPR2; often referred to as FPR2/ALX since it binds lipoxin A4 ). Initially identified as a low-affinity 'relative' of FPR1, FPR2 presents complex and diverse biology. For instance, it is activated by several classes of agonists (from peptides to proteins and lipid mediators) and displays diverse expression patterns on myeloid cells as well as epithelial cells and endothelial cells, to name a few. Over the last decade, the pharmacology of FPR2 has progressed from being considered a weak chemotactic receptor to a master-regulator of the resolution of inflammation, the second phase of the acute inflammatory response. We propose that exploitation of the biology of FPR2 offers innovative ways to rectify chronic inflammatory states and represents a viable avenue to develop novel therapies. Recent elucidation of FPR2 structure will facilitate development of the anti-inflammatory and pro-resolving drugs of next decade.
Collapse
Affiliation(s)
- Cheng Xue Qin
- Drug Discovery Biology, Monash Institute of Pharmaceutical SciencesMonash UniversityMelbourneVictoriaAustralia
| | - Lucy V. Norling
- William Harvey Research Institute, Barts and the London School of MedicineQueen Mary University of LondonLondonUK
| | - Elizabeth A. Vecchio
- Drug Discovery Biology, Monash Institute of Pharmaceutical SciencesMonash UniversityMelbourneVictoriaAustralia
| | - Eoin P. Brennan
- Diabetes Complications Research Centre, Conway Institute and School of MedicineUniversity College DublinDublinIreland
| | - Lauren T. May
- Drug Discovery Biology, Monash Institute of Pharmaceutical SciencesMonash UniversityMelbourneVictoriaAustralia
| | - Denise Wootten
- Drug Discovery Biology, Monash Institute of Pharmaceutical SciencesMonash UniversityMelbourneVictoriaAustralia
| | - Catherine Godson
- Diabetes Complications Research Centre, Conway Institute and School of MedicineUniversity College DublinDublinIreland
| | - Mauro Perretti
- William Harvey Research Institute, Barts and the London School of MedicineQueen Mary University of LondonLondonUK
| | - Rebecca H. Ritchie
- Drug Discovery Biology, Monash Institute of Pharmaceutical SciencesMonash UniversityMelbourneVictoriaAustralia
| |
Collapse
|
22
|
Li W, Jiang H, Bai C, Yu S, Pan Y, Wang C, Li H, Li M, Sheng Y, Chu F, Wang J, Chen Y, Li J, Jiang J. Ac2-26 attenuates hepatic ischemia-reperfusion injury in mice via regulating IL-22/IL-22R1/STAT3 signaling. PeerJ 2022; 10:e14086. [PMID: 36193422 PMCID: PMC9526407 DOI: 10.7717/peerj.14086] [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: 05/26/2022] [Accepted: 08/29/2022] [Indexed: 01/27/2023] Open
Abstract
Hepatic ischemia-reperfusion injury (HIRI) is one of the major sources of mortality and morbidity associated with hepatic surgery. Ac2-26, a short peptide of Annexin A1 protein, has been proved to have a protective effect against IRI. However, whether it exerts a protective effect on HIRI has not been reported. The HIRI mice model and the oxidative damage model of H2O2-induced AML12 cells were established to investigate whether Ac2-26 could alleviate HIRI by regulating the activation of IL-22/IL-22R1/STAT3 signaling. The protective effect of Ac2-26 was measured by various biochemical parameters related to liver function, apoptosis, inflammatory reaction, mitochondrial function and the expressions of IL-22, IL-22R1, p-STAT3Tyr705. We discovered that Ac2-26 reduced the Suzuki score and cell death rate, and increased the cell viability after HIRI. Moreover, we unraveled that Ac2-26 significantly decreased the number of apoptotic hepatocytes, and the expressions of cleaved-caspase-3 and Bax/Bcl-2 ratio. Furthermore, HIRI increased the contents of malondialdehyde (MDA), NADP+/NADPH ratio and reactive oxygen species (ROS), whereas Ac2-26 decreased them significantly. Additionally, Ac2-26 remarkably alleviated mitochondria dysfunction, which was represented by an increase in the adenosine triphosphate (ATP) content and mitochondrial membrane potential, a decrease in mitochondrial DNA (mtDNA) damage. Finally, we revealed that Ac2-26 pretreatment could significantly inhibit the activation of IL-22/IL22R1/STAT3 signaling. In conclusion, this work demonstrated that Ac2-26 ameliorated HIRI by reducing oxidative stress and inhibiting the mitochondrial apoptosis pathway, which might be closely related to the inhibition of the IL-22/IL22R1/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Wanzhen Li
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Hongxin Jiang
- Morphology Lab, Weifang Medical University, Weifang, Shandong, China
| | - Chen Bai
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Shuna Yu
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Yitong Pan
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Chenchen Wang
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Huiting Li
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Ming Li
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Yaxin Sheng
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Fangfang Chu
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Jie Wang
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Yuting Chen
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Jianguo Li
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Jiying Jiang
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
23
|
Li W, Gonzalez KM, Chung J, Kim M, Lu J. Surface-modified nanotherapeutics targeting atherosclerosis. Biomater Sci 2022; 10:5459-5471. [PMID: 35980230 PMCID: PMC9529904 DOI: 10.1039/d2bm00660j] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Atherosclerosis is a chronic and metabolic-related disease that is a serious threat to human health. Currently available diagnostic and therapeutic measures for atherosclerosis lack adequate efficiency which requires promising alternative approaches. Nanotechnology-based nano-delivery systems allow for new perspectives for atherosclerosis therapy. Surface-modified nanoparticles could achieve highly effective therapeutic effects by binding to specific receptors that are abnormally overexpressed in atherosclerosis, with less adverse effects on non-target tissues. The main purpose of this review is to summarize the research progress and design ideas to target atherosclerosis using a variety of ligand-modified nanoparticle systems, discuss the shortcomings of current vector design, and look at future development directions. We hope that this review will provide novel research strategies for the design and development of nanotherapeutics targeting atherosclerosis.
Collapse
Affiliation(s)
- Wenpan Li
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, USA.
| | - Karina Marie Gonzalez
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, USA.
| | - Jinha Chung
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, USA.
| | - Minhyeok Kim
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, USA.
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, USA.
- NCI-designated University of Arizona Comprehensive Cancer Center, Tucson, Arizona, 85721, USA
- BIO5 Institute, The University of Arizona, Tucson, Arizona, 85721, USA
- Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, 85721, USA
| |
Collapse
|
24
|
Hossain MI, Nanda SS, Selvan ST, Yi DK. Recent Insights into NIR-Light-Responsive Materials for Photothermal Cell Treatments. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3318. [PMID: 36234446 PMCID: PMC9565779 DOI: 10.3390/nano12193318] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 06/16/2023]
Abstract
Controlling cells using photo-responsive materials is highly indispensable in the current biomedical sector. Considering the potential side effects of nanoparticles, it has become a challenge to control cells with photo-responsive materials. Recent studies have described several methods for controlling cell behavior using nanoparticles subjected to the near-infrared (NIR) laser light operating at the wavelength of 808 nm to 980 nm and at the power densities of 0.33 to 0.72 W·cm-2. The challenge here is the preparation of biocompatible nanoparticles for both in vivo and in vitro studies and understanding cell behavior with an external light source recommended for biological application. Earlier studies have well documented many approaches and associated mechanisms for controlling cell behavior and the interaction between nanoparticles, cells, and appropriate external light sources. In this review, various nanomaterials such as metal nanomaterials and carbon-based nanomaterials are compared systematically regarding the effects of controlling cell behavior and inflammation by studying their mechanisms, route of administration, dose, and adverse effects such as toxicity and the interaction of nanoparticles with a specific wavelength of the light. Future directions should focus on stable and efficient light-responsive materials with minimal cytotoxicity.
Collapse
Affiliation(s)
- Md Imran Hossain
- Department of Chemistry, Myongji University, Yongin 17058, Korea
| | | | - Subramanian Tamil Selvan
- Alpha Biomedical Pte Ltd., 21 Biopolis Road, Nucleos North Tower #02-03, Singapore 138567, Singapore
| | - Dong Kee Yi
- Department of Chemistry, Myongji University, Yongin 17058, Korea
| |
Collapse
|
25
|
Zhang Q, Li F, Ritchie RH, Woodman OL, Zhou X, Qin CX. Novel strategies to promote resolution of inflammation to treat lower extremity artery disease. Curr Opin Pharmacol 2022; 65:102263. [DOI: 10.1016/j.coph.2022.102263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/24/2022] [Accepted: 05/31/2022] [Indexed: 12/24/2022]
|
26
|
Inhibitory role of Annexin A1 in pathological bone resorption and therapeutic implications in periprosthetic osteolysis. Nat Commun 2022; 13:3919. [PMID: 35798730 PMCID: PMC9262976 DOI: 10.1038/s41467-022-31646-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/20/2022] [Indexed: 12/26/2022] Open
Abstract
There is currently no therapy available for periprosthetic osteolysis, the most common cause of arthroplasty failure. Here, the role of AnxA1 in periprosthetic osteolysis and potential therapeutics were investigated. Reducing the expression of AnxA1 in calvarial tissue was found to be associated with increased osteolytic lesions and the osteolytic lesions induced by debris implantation were more severe in AnxA1-defecient mice than in wild-type mice. AnxA1 inhibits the differentiation of osteoclasts through suppressing NFκB signaling and promoting the PPAR-γ pathway. Administration of N-terminal-AnxA1 (Ac2-26 peptide) onto calvariae significantly reduced osteolytic lesions triggered by wear debris. These therapeutic effects were abrogated in mice that had received the PPAR-γ antagonist, suggesting that the AnxA1/PPAR-γ axis has an inhibitory role in osteolysis. The administration of Ac2–26 suppressed osteolysis induced by TNF-α and RANKL injections in mice. These findings indicate that AnxA1 is a potential therapeutic agent for the treatment of periprosthetic osteolysis. Periprosthetic osteolysis is a cause of arthroplasty failure without available therapies. Here the authors show that Annexin A1 (AnxA1) is involved in in periprosthetic osteolysis and exerts potential therapeutic effects through suppressing NFκB signaling and promoting the PPAR-γ pathway resulting in inhibition of inflammation and osteoclasts differentiation induced by wear debris.
Collapse
|
27
|
Decoding microRNA drivers in Atherosclerosis. Biosci Rep 2022; 42:231479. [PMID: 35758143 PMCID: PMC9289798 DOI: 10.1042/bsr20212355] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/17/2022] [Accepted: 06/26/2022] [Indexed: 11/17/2022] Open
Abstract
An estimated 97% of the human genome consists of non-protein-coding sequences. As our understanding of genome regulation improves, this has led to the characterization of a diverse array of non-coding RNAs (ncRNA). Among these, micro-RNAs (miRNAs) belong to the short ncRNA class (22–25 nucleotides in length), with approximately 2500 miRNA genes encoded within the human genome. From a therapeutic perspective, there is interest in exploiting miRNA as biomarkers of disease progression and response to treatments, as well as miRNA mimics/repressors as novel medicines. miRNA have emerged as an important class of RNA master regulators with important roles identified in the pathogenesis of atherosclerotic cardiovascular disease. Atherosclerosis is characterized by a chronic inflammatory build-up, driven largely by low-density lipoprotein cholesterol accumulation within the artery wall and vascular injury, including endothelial dysfunction, leukocyte recruitment and vascular remodelling. Conventional therapy focuses on lifestyle interventions, blood pressure-lowering medications, high-intensity statin therapy and antiplatelet agents. However, a significant proportion of patients remain at increased risk of cardiovascular disease. This continued cardiovascular risk is referred to as residual risk. Hence, a new drug class targeting atherosclerosis could synergise with existing therapies to optimise outcomes. Here, we review our current understanding of the role of ncRNA, with a focus on miRNA, in the development and progression of atherosclerosis, highlighting novel biological mechanisms and therapeutic avenues.
Collapse
|
28
|
Yan Z, Cheng X, Wang T, Hong X, Shao G, Fu C. Therapeutic potential for targeting Annexin A1 in fibrotic diseases. Genes Dis 2022; 9:1493-1505. [PMID: 36157506 PMCID: PMC9485289 DOI: 10.1016/j.gendis.2022.05.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 05/30/2022] [Indexed: 11/23/2022] Open
Abstract
Annexin A1, a well-known endogenous anti-inflammatory mediator, plays a critical role in a variety of pathological processes. Fibrosis is described by a failure of tissue regeneration and contributes to the development of many diseases. Accumulating evidence supports that Annexin A1 participates in the progression of tissue fibrosis. However, the fundamental mechanisms by which Annexin A1 regulates fibrosis remain elusive, and even the functions of Annexin A1 in fibrotic diseases are still paradoxical. This review focuses on the roles of Annexin A1 in the development of fibrosis of lung, liver, heart, and other tissues, with emphasis on the therapy potential of Annexin A1 in fibrosis, and presents future research interests and directions in fibrotic diseases.
Collapse
|
29
|
Pharmacological treatment with annexin A1-derived peptide protects against cisplatin-induced hearing loss. Toxicol Lett 2022; 363:27-35. [PMID: 35561849 DOI: 10.1016/j.toxlet.2022.05.001] [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: 11/30/2021] [Revised: 04/11/2022] [Accepted: 05/02/2022] [Indexed: 11/20/2022]
Abstract
Cisplatin is an antineoplastic agent widely used, and no effective treatments capable of preventing cisplatin-induced ototoxicity and neurotoxicity in humans have yet been identified. This study evaluated the effect of the anti-inflammatory annexin A1 (AnxA1)-derived peptide Ac2-26 in a cisplatin-induced ototoxicity model. Wistar rats received intraperitoneal injections of cisplatin (10mg/kg/day) for 3 days to induce hearing loss, and Ac2-26 (1mg/kg) was administered 15minutes before cisplatin administration. Control animals received an equal volume of saline. Hearing thresholds were measured by distortion product otoacoustic emissions (DPOAE) before and after treatments. Pharmacological treatment with Ac2-26 protected against cisplatin-induced hearing loss, as evidenced by DPOAE results showing similar signal-noise ratios between the control and Ac2-26-treated groups. These otoprotective effects of Ac2-26 were associated with an increased number of ganglion neurons compared with the untreated cisplatin group. Additionally, Ac2-26 treatment produced reduced immunoreactivity on cleaved caspase 3 and phosphorylated ERK levels in the ganglion neurons, compared to the untreated group, supporting the neuroprotective effects of the Ac2-26. Our results suggest that Ac2-26 has a substantial otoprotective effect in this cisplatin-induced ototoxicity model mediated by neuroprotection and the regulation of the ERK pathway.
Collapse
|
30
|
Chen W, Schilperoort M, Cao Y, Shi J, Tabas I, Tao W. Macrophage-targeted nanomedicine for the diagnosis and treatment of atherosclerosis. Nat Rev Cardiol 2022; 19:228-249. [PMID: 34759324 PMCID: PMC8580169 DOI: 10.1038/s41569-021-00629-x] [Citation(s) in RCA: 199] [Impact Index Per Article: 99.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/22/2021] [Indexed: 12/12/2022]
Abstract
Nanotechnology could improve our understanding of the pathophysiology of atherosclerosis and contribute to the development of novel diagnostic and therapeutic strategies to further reduce the risk of cardiovascular disease. Macrophages have key roles in atherosclerosis progression and, therefore, macrophage-associated pathological processes are important targets for both diagnostic imaging and novel therapies for atherosclerosis. In this Review, we highlight efforts in the past two decades to develop imaging techniques and to therapeutically manipulate macrophages in atherosclerotic plaques with the use of rationally designed nanoparticles. We review the latest progress in nanoparticle-based imaging modalities that can specifically target macrophages. Using novel molecular imaging technology, these modalities enable the identification of advanced atherosclerotic plaques and the assessment of the therapeutic efficacy of medical interventions. Additionally, we provide novel perspectives on how macrophage-targeting nanoparticles can deliver a broad range of therapeutic payloads to atherosclerotic lesions. These nanoparticles can suppress pro-atherogenic macrophage processes, leading to improved resolution of inflammation and stabilization of plaques. Finally, we propose future opportunities for novel diagnostic and therapeutic strategies and provide solutions to challenges in this area for the purpose of accelerating the clinical translation of nanomedicine for the treatment of atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Wei Chen
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maaike Schilperoort
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Yihai Cao
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Wei Tao
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
31
|
Merlin JPJ, Li X. Role of Nanotechnology and Their Perspectives in the Treatment of Kidney Diseases. Front Genet 2022; 12:817974. [PMID: 35069707 PMCID: PMC8766413 DOI: 10.3389/fgene.2021.817974] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 12/06/2021] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles (NPs) are differing in particle size, charge, shape, and compatibility of targeting ligands, which are linked to improved pharmacologic characteristics, targetability, and bioavailability. Researchers are now tasked with developing a solution for enhanced renal treatment that is free of side effects and delivers the medicine to the active spot. A growing number of nano-based medication delivery devices are being used to treat renal disorders. Kidney disease management and treatment are currently causing a substantial global burden. Renal problems are multistep processes involving the accumulation of a wide range of molecular and genetic alterations that have been related to a variety of kidney diseases. Renal filtration is a key channel for drug elimination in the kidney, as well as a burgeoning topic of nanomedicine. Although the use of nanotechnology in the treatment of renal illnesses is still in its early phases, it offers a lot of potentials. In this review, we summarized the properties of the kidney and characteristics of drug delivery systems, which affect a drug’s ability should focus on the kidney and highlight the possibilities, problems, and opportunities.
Collapse
Affiliation(s)
- J P Jose Merlin
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
32
|
Thi Kim Dung D, Umezawa M, Ohnuki K, Nigoghossian K, Okubo K, Kamimura M, Yamaguchi M, Fujii H, Soga K. The influence of Gd-DOTA ratios conjugating PLGA-PEG micelles encapsulated IR-1061 in bimodal over–1000 nm near–infrared fluorescence and magnetic resonance imaging. Biomater Sci 2022; 10:1217-1230. [DOI: 10.1039/d1bm01574e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Multimodal imaging can provide multidimensional information for understanding concealed microstructures or bioprocesses in biological objects. The combination of over–1000 nm near–infrared (OTN–NIR) fluorescence imaging and magnetic resonance (MR) imaging is...
Collapse
|
33
|
Chen L, Zhou Z, Hu C, Maitz MF, Yang L, Luo R, Wang Y. Platelet Membrane-Coated Nanocarriers Targeting Plaques to Deliver Anti-CD47 Antibody for Atherosclerotic Therapy. RESEARCH (WASHINGTON, D.C.) 2022; 2022:9845459. [PMID: 35118420 PMCID: PMC8791388 DOI: 10.34133/2022/9845459] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/24/2021] [Indexed: 12/22/2022]
Abstract
Atherosclerosis, the principle cause of cardiovascular disease (CVD) worldwide, is mainly characterized by the pathological accumulation of diseased vascular cells and apoptotic cellular debris. Atherogenesis is associated with the upregulation of CD47, a key antiphagocytic molecule that is known to render malignant cells resistant to programmed cell removal, or "efferocytosis." Here, we have developed platelet membrane-coated mesoporous silicon nanoparticles (PMSN) as a drug delivery system to target atherosclerotic plaques with the delivery of an anti-CD47 antibody. Briefly, the cell membrane coat prolonged the circulation of the particles by evading the immune recognition and provided an affinity to plaques and atherosclerotic sites. The anti-CD47 antibody then normalized the clearance of diseased vascular tissue and further ameliorated atherosclerosis by blocking CD47. In an atherosclerosis model established in ApoE-/- mice, PMSN encapsulating anti-CD47 antibody delivery significantly promoted the efferocytosis of necrotic cells in plaques. Clearing the necrotic cells greatly reduced the atherosclerotic plaque area and stabilized the plaques reducing the risk of plaque rupture and advanced thrombosis. Overall, this study demonstrated the therapeutic advantages of PMSN encapsulating anti-CD47 antibodies for atherosclerosis therapy, which holds considerable promise as a new targeted drug delivery platform for efficient therapy of atherosclerosis.
Collapse
Affiliation(s)
- Liang Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Zhongyi Zhou
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Cheng Hu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Manfred F. Maitz
- Max Bergmann Center of Biomaterials, Leibniz Institute of Polymer Research Dresden, Dresden 01069, Germany
- Key Lab. for Advanced Technologies of Materials, Ministry of Education, School of Material Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Rifang Luo
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| |
Collapse
|
34
|
Zhang R, Zhang Q, Zhu S, Liu B, Liu F, Xu Y. Mulberry leaf (Morus alba L.): A review of its potential influences in mechanisms of action on metabolic diseases. Pharmacol Res 2021; 175:106029. [PMID: 34896248 DOI: 10.1016/j.phrs.2021.106029] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/17/2021] [Accepted: 12/06/2021] [Indexed: 12/18/2022]
Abstract
The leaves of Morus alba L. (called Sangye in Chinese, ML), which belong to the genus Morus., are highly valuable edible plants in nutrients and nutraceuticals. In Asian countries including China, Japan and Korea, ML are widely used as functional foods including beverages, noodles and herbal tea because of its biological and nutritional value. Meanwhile, ML-derived products in the form of powders, extracts and capsules are widely consumed as dietary supplements for controlling blood glucose and sugar. Clinical studies showed that ML play an important role in the treatment of metabolic diseases including the diabetes, dyslipidemia, obesity, atherosclerosis and hypertension. People broadly use ML due to their nutritiousness, deliciousness, safety, and abundant active benefits. However, the systematic pharmacological mechanisms of ML on metabolic diseases have not been fully revealed. Therefore, in order to fully utilize and scale relevant products about ML, this review summarizes the up-to-date information about the ML and its constituents effecting on metabolic disease.
Collapse
Affiliation(s)
- Ruiyuan Zhang
- Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, People's Republic of China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, People's Republic of China
| | - Qian Zhang
- Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, People's Republic of China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, People's Republic of China
| | - Shun Zhu
- Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, People's Republic of China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, People's Republic of China
| | - Biyang Liu
- Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, People's Republic of China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, People's Republic of China
| | - Fang Liu
- Pharmacy College of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, People's Republic of China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, People's Republic of China.
| | - Yao Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, People's Republic of China.
| |
Collapse
|
35
|
Efferocytosis induces macrophage proliferation to help resolve tissue injury. Cell Metab 2021; 33:2445-2463.e8. [PMID: 34784501 PMCID: PMC8665147 DOI: 10.1016/j.cmet.2021.10.015] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/17/2021] [Accepted: 10/26/2021] [Indexed: 01/01/2023]
Abstract
Apoptotic cell clearance by macrophages (efferocytosis) promotes resolution signaling pathways, which can be triggered by molecules derived from the phagolysosomal degradation of apoptotic cells. We show here that nucleotides derived from the hydrolysis of apoptotic cell DNA by phagolysosomal DNase2a activate a DNA-PKcs-mTORC2/Rictor pathway that increases Myc to promote non-inflammatory macrophage proliferation. Efferocytosis-induced proliferation expands the pool of resolving macrophages in vitro and in mice, including zymosan-induced peritonitis, dexamethasone-induced thymocyte apoptosis, and atherosclerosis regression. In the dexamethasone-thymus model, hematopoietic Rictor deletion blocked efferocytosing macrophage proliferation, apoptotic cell clearance, and tissue resolution. In atherosclerosis regression, silencing macrophage Rictor or DNase2a blocked efferocyte proliferation, apoptotic cell clearance, and plaque stabilization. In view of previous work showing that other types of apoptotic cell cargo can promote resolution in individual efferocytosing macrophages, the findings here suggest that signaling-triggered apoptotic cell-derived nucleotides can amplify this benefit by increasing the number of these macrophages.
Collapse
|
36
|
Pillai SC, Borah A, Jacob EM, Kumar DS. Nanotechnological approach to delivering nutraceuticals as promising drug candidates for the treatment of atherosclerosis. Drug Deliv 2021; 28:550-568. [PMID: 33703990 PMCID: PMC7954496 DOI: 10.1080/10717544.2021.1892241] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 12/25/2022] Open
Abstract
Atherosclerosis is Caesar's sword, which poses a huge risk to the present generation. Understanding the atherosclerotic disease cycle would allow ensuring improved diagnosis, better care, and treatment. Unfortunately, a highly effective and safe way of treating atherosclerosis in the medical community remains a continuous challenge. Conventional treatments have shown considerable success, but have some adverse effects on the human body. Natural derived medications or nutraceuticals have gained immense popularity in the treatment of atherosclerosis due to their decreased side effects and toxicity-related issues. In hindsight, the contribution of nutraceuticals in imparting enhanced clinical efficacy against atherosclerosis warrants more experimental evidence. On the other hand, nanotechnology and drug delivery systems (DDS) have revolutionized the way therapeutics are performed and researchers have been constantly exploring the positive effects that DDS brings to the field of therapeutic techniques. It could be as exciting as ever to apply nano-mediated delivery of nutraceuticals as an additional strategy to target the atherosclerotic sites boasting high therapeutic efficiency of the nutraceuticals and fewer side effects.
Collapse
Affiliation(s)
- Sindhu C. Pillai
- Bio-Nano Electronics Research Centre, Graduate School of Interdisciplinary New Science, Toyo University, Saitama, Japan
| | - Ankita Borah
- Bio-Nano Electronics Research Centre, Graduate School of Interdisciplinary New Science, Toyo University, Saitama, Japan
| | - Eden Mariam Jacob
- Bio-Nano Electronics Research Centre, Graduate School of Interdisciplinary New Science, Toyo University, Saitama, Japan
| | - D. Sakthi Kumar
- Bio-Nano Electronics Research Centre, Graduate School of Interdisciplinary New Science, Toyo University, Saitama, Japan
| |
Collapse
|
37
|
Molinaro R, Yu M, Sausen G, Bichsel CA, Corbo C, Folco EJ, Lee GY, Liu Y, Tesmenitsky Y, Shvartz E, Sukhova GK, Kloss F, Croce KJ, Farokhzad OC, Shi J, Libby P. Targeted delivery of protein arginine deiminase-4 inhibitors to limit arterial intimal NETosis and preserve endothelial integrity. Cardiovasc Res 2021; 117:2652-2663. [PMID: 33751034 PMCID: PMC8783386 DOI: 10.1093/cvr/cvab074] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
AIMS Recent evidence suggests that 'vulnerable plaques', which have received intense attention as underlying mechanism of acute coronary syndromes over the decades, actually rarely rupture and cause clinical events. Superficial plaque erosion has emerged as a growing cause of residual thrombotic complications of atherosclerosis in an era of increased preventive measures including lipid lowering, antihypertensive therapy, and smoking cessation. The mechanisms of plaque erosion remain poorly understood, and we currently lack validated effective diagnostics or therapeutics for superficial erosion. Eroded plaques have a rich extracellular matrix, an intact fibrous cap, sparse lipid, and few mononuclear cells, but do harbour neutrophil extracellular traps (NETs). We recently reported that NETs amplify and propagate the endothelial damage at the site of arterial lesions that recapitulate superficial erosion in mice. We showed that genetic loss of protein arginine deiminase (PAD)-4 function inhibited NETosis and preserved endothelial integrity. The current study used systemic administration of targeted nanoparticles to deliver an agent that limits NETs formation to probe mechanisms of and demonstrate a novel therapeutic approach to plaque erosion that limits endothelial damage. METHODS AND RESULTS We developed Collagen IV-targeted nanoparticles (Col IV NP) to deliver PAD4 inhibitors selectively to regions of endothelial cell sloughing and collagen IV-rich basement membrane exposure. We assessed the binding capability of the targeting ligand in vitro and evaluated Col IV NP targeting to areas of denuded endothelium in vivo in a mouse preparation that recapitulates features of superficial erosion. Delivery of the PAD4 inhibitor GSK484 reduced NET accumulation at sites of intimal injury and preserved endothelial continuity. CONCLUSIONS NPs directed to Col IV show selective uptake and delivery of their payload to experimentally eroded regions, illustrating their translational potential. Our results further support the role of PAD4 and NETs in superficial erosion.
Collapse
Affiliation(s)
- Roberto Molinaro
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Business Development of Research, IRCCS San Raffaele Hospital, Milan, Italy
| | - Mikyung Yu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Grasiele Sausen
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Colette A Bichsel
- Department of Surgery, Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Claudia Corbo
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine and Surgery, Nanomedicine Center NANOMIB, University of Milano-Bicocca, Milano, Italy
| | - Eduardo J Folco
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Gha Young Lee
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yuan Liu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yevgenia Tesmenitsky
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Eugenia Shvartz
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Galina K Sukhova
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Frederik Kloss
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Kevin J Croce
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| |
Collapse
|
38
|
Vartak T, Godson C, Brennan E. Therapeutic potential of pro-resolving mediators in diabetic kidney disease. Adv Drug Deliv Rev 2021; 178:113965. [PMID: 34508793 DOI: 10.1016/j.addr.2021.113965] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 07/26/2021] [Accepted: 09/05/2021] [Indexed: 02/06/2023]
Abstract
Renal microvascular disease associated with diabetes [Diabetic kidney disease - DKD] is the leading cause of chronic kidney disease. In DKD, glomerular basement membrane thickening, mesangial expansion, endothelial dysfunction, podocyte cell loss and renal tubule injury contribute to progressive glomerulosclerosis and tubulointerstitial fibrosis. Chronic inflammation is recognized as a major pathogenic mechanism for DKD, with resident and circulating immune cells interacting with local kidney cell populations to provoke an inflammatory response. The onset of inflammation is driven by the release of well described proinflammatory mediators, and this is typically followed by a resolution phase. Inflammation resolution is achieved through the bioactions of endogenous specialized pro-resolving lipid mediators (SPMs). As our understanding of SPMs advances 'resolution pharmacology' based approaches using these molecules are being explored in DKD.
Collapse
Affiliation(s)
- Tanwi Vartak
- Diabetes Complications Research Centre, Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland
| | - Catherine Godson
- Diabetes Complications Research Centre, Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland
| | - Eoin Brennan
- Diabetes Complications Research Centre, Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland.
| |
Collapse
|
39
|
Reischl S, Lee JH, Miltschitzky JRE, Vieregge V, Walter RL, Twardy V, Kasajima A, Friess H, Kamaly N, Neumann PA. Ac2-26-Nanoparticles Induce Resolution of Intestinal Inflammation and Anastomotic Healing via Inhibition of NF-κB Signaling in a Model of Perioperative Colitis. Inflamm Bowel Dis 2021; 27:1379-1393. [PMID: 33512505 DOI: 10.1093/ibd/izab008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Although in most patients with inflammatory bowel diseases, conservative therapy is successful, a significant proportion of patients still require surgery once in their lifetime. Development of a safe perioperative treatment to dampen colitis activity without disturbance of anastomotic healing is an urgent and unmet medical need. Annexin A1 (ANXA1) has been shown to be effective in reducing colitis activity. Herein, a nanoparticle-based perioperative treatment approach was used for analysis of the effects of ANXA1 on the resolution of inflammation after surgery for colitis. METHODS Anxa1-knockout mice were used to delineate the effects of ANXA1 on anastomotic healing. A murine model of preoperative dextran sodium sulfate colitis was performed. Collagen-IV-targeted polymeric nanoparticles, loaded with the ANXA1 biomimetic peptide Ac2-26 (Ac2-26-NPs), were synthesized and administered perioperatively during colitis induction. The effects of the Ac2-26-NPs on postoperative recovery and anastomotic healing were evaluated using the disease activity index, histological healing scores, and weight monitoring. Ultimately, whole-genome RNA sequencing of the anastomotic tissue was performed to unravel underlying molecular mechanisms. RESULTS Anxa1-knockout exacerbated the inflammatory response in the healing anastomosis. Treatment with Ac2-26-NPs improved preoperative colitis activity (P < 0.045), postoperative healing scores (P < 0.018), and weight recovery (P < 0.015). Whole-genome RNA sequencing revealed that the suppression of proinflammatory cytokine and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling was associated with the treatment effects and a phenotypic switch toward anti-inflammatory M2 macrophages. CONCLUSIONS Proresolving therapy with Ac2-26-NPs promises to be a potent perioperative therapy because it improves colitis activity and even intestinal anastomotic healing by the suppression of proinflammatory signaling.
Collapse
Affiliation(s)
- Stefan Reischl
- Technical University of Munich, School of Medicine, Department of Surgery, Munich, Germany
| | - Jong Hyun Lee
- Technical University of Denmark, Department of Health Technology, Copenhagen, Denmark
| | | | - Vincent Vieregge
- Technical University of Munich, School of Medicine, Department of Surgery, Munich, Germany
| | - Robert Leon Walter
- Technical University of Munich, School of Medicine, Department of Surgery, Munich, Germany
| | - Vanessa Twardy
- University of Muenster, School of Medicine, Department of Surgery, Muenster, Germany
| | - Atsuko Kasajima
- Technical Technical University of Munich, School of Medicine, Institute of Pathology, Munich, Germany
| | - Helmut Friess
- Technical University of Munich, School of Medicine, Department of Surgery, Munich, Germany
| | - Nazila Kamaly
- Technical University of Denmark, Department of Health Technology, Copenhagen, Denmark.,Imperial College London, Department of Chemistry, Molecular Sciences Research Hub, London, United Kingdom
| | | |
Collapse
|
40
|
Resolving thromboinflammation. Blood 2021; 137:1444-1446. [PMID: 33734338 DOI: 10.1182/blood.2020010627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
41
|
Bender EC, Kraynak CA, Huang W, Suggs LJ. Cell-Inspired Biomaterials for Modulating Inflammation. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:279-294. [PMID: 33528306 DOI: 10.1089/ten.teb.2020.0276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Inflammation is a crucial part of wound healing and pathogen clearance. However, it can also play a role in exacerbating chronic diseases and cancer progression when not regulated properly. A subset of current innate immune engineering research is focused on how molecules such as lipids, proteins, and nucleic acids native to a healthy inflammatory response can be harnessed in the context of biomaterial design to promote healing, decrease disease severity, and prolong survival. The engineered biomaterials in this review inhibit inflammation by releasing anti-inflammatory cytokines, sequestering proinflammatory cytokines, and promoting phenotype switching of macrophages in chronic inflammatory disease models. Conversely, other biomaterials discussed here promote inflammation by mimicking pathogen invasion to inhibit tumor growth in cancer models. The form that these biomaterials take spans a spectrum from nanoparticles to large-scale hydrogels to surface coatings on medical devices. Cell-inspired molecules have been incorporated in a variety of creative ways, including loaded into or onto the surface of biomaterials or used as the biomaterials themselves.
Collapse
Affiliation(s)
- Elizabeth C Bender
- Department of Biomedical Engineering and The University of Texas at Austin, Austin, Texas, USA
| | - Chelsea A Kraynak
- Department of Biomedical Engineering and The University of Texas at Austin, Austin, Texas, USA
| | - Wenbai Huang
- Department of Biomedical Engineering and The University of Texas at Austin, Austin, Texas, USA.,Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, Texas, USA
| | - Laura J Suggs
- Department of Biomedical Engineering and The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
42
|
Atashgah RB, Ghasemi A, Raoufi M, Abdollahifar MA, Zanganeh S, Nejadnik H, Abdollahi A, Sharifi S, Lea B, Cuerva M, Akbarzadeh M, Alvarez-Lorenzo C, Ostad SN, Theus AS, LaRock DL, LaRock CN, Serpooshan V, Sarrafi R, Lee KB, Vali H, Schönherr H, Gould L, Taboada P, Mahmoudi M. Restoring Endogenous Repair Mechanisms to Heal Chronic Wounds with a Multifunctional Wound Dressing. Mol Pharm 2021; 18:3171-3180. [PMID: 34279974 DOI: 10.1021/acs.molpharmaceut.1c00400] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Current treatment of chronic wounds has been critically limited by various factors, including bacterial infection, biofilm formation, impaired angiogenesis, and prolonged inflammation. Addressing these challenges, we developed a multifunctional wound dressing-based three-pronged approach for accelerating wound healing. The multifunctional wound dressing, composed of nanofibers, functional nanoparticles, natural biopolymers, and selected protein and peptide, can target multiple endogenous repair mechanisms and represents a promising alternative to current wound healing products.
Collapse
Affiliation(s)
- Rahimeh B Atashgah
- Colloids and Polymers Physics Group, Particle Physics Department, Faculty of Physics and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain.,Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14167-53955, Iran
| | - Amir Ghasemi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 13169-43551, Iran
| | - Mohammad Raoufi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 13169-43551, Iran.,Physical Chemistry I, Department of Chemistry and Biology & Research Center of Micro and Nanochemistry and Engineering (Cμ), University of Siegen, Siegen 57076, Germany
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 19395-4719, Iran
| | - Steven Zanganeh
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, Massachusetts 02747, United States
| | - Hossein Nejadnik
- Department of Radiology, University of Pennsylvania, Philladelphia, Pennsylvania 19104, United States
| | - Alieh Abdollahi
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14167-53955, Iran
| | - Shahriar Sharifi
- Precision Health Program, Michigan State University, East Lansing, Michigan 48824, United States
| | - Baltazar Lea
- Colloids and Polymers Physics Group, Particle Physics Department, Faculty of Physics and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Miguel Cuerva
- NANOMAG Group, Technological Research Institute (IIT), Physical Chemistry Department, University of Santiago de Compostela (USC), Santiago de Compostela 15782, Spain
| | - Mehdi Akbarzadeh
- Sadra Wound, Ostomy and Osteomyelitis Specialist Center, Tehran, Iran
| | - Carmen Alvarez-Lorenzo
- R+D Pharma Group, Pharmacology, Pharmacy and Pharmaceutical Technology Department, Faculty of Pharmacy and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Seyed Nasser Ostad
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14167-53955, Iran
| | - Andrea S Theus
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, Georgia 30322, United States
| | - Doris L LaRock
- Department of Microbiology and Immunology, Emory Antibiotic Resistance Center, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Christopher N LaRock
- Department of Microbiology and Immunology, Emory Antibiotic Resistance Center, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Vahid Serpooshan
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, Georgia 30322, United States.,Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia 30309, United States.,Children's Healthcare of Atlanta, Atlanta, Georgia 30322, United States
| | | | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Hojatollah Vali
- Department of Anatomy and Cell Biology and Facility for Electron Microscopy Research, McGill University, Montreal, Quebec H3A 0C3, Canada
| | - Holger Schönherr
- Physical Chemistry I, Department of Chemistry and Biology & Research Center of Micro and Nanochemistry and Engineering (Cμ), University of Siegen, Siegen 57076, Germany
| | - Lisa Gould
- Brown University School of Medicine, Providence, Rhode Island 02903, United States.,South Shore Health System Center for Wound Healing, Weymouth, Massachusetts 02189, United States
| | - Pablo Taboada
- Colloids and Polymers Physics Group, Particle Physics Department, Faculty of Physics and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Morteza Mahmoudi
- Precision Health Program, Michigan State University, East Lansing, Michigan 48824, United States.,Department of Anesthesiology, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States.,Mary Horrigan Connors Center for Women's Health & Gender Biology, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
43
|
Qin X, He L, Fan D, Liang W, Wang Q, Fang J. Targeting the resolution pathway of inflammation using Ac2-26 peptide-loaded PEGylated lipid nanoparticles for the remission of rheumatoid arthritis. Asian J Pharm Sci 2021; 16:483-493. [PMID: 34703497 PMCID: PMC8520054 DOI: 10.1016/j.ajps.2021.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/03/2021] [Accepted: 03/05/2021] [Indexed: 12/29/2022] Open
Abstract
Rheumatoid arthritis (RA) is a common autoimmune disease characterized by joint inflammation and immune dysfunction. Although various therapeutic approaches have been utilized for the treatment of RA in clinical applications, the low responsiveness of RA patients and undesired systemic toxicity are still unresolved problems. Targeting the resolution pathway of inflammation with pro-resolving mediators would evoke the protective actions of patient for combating the inflammation. Ac2-26, a 25-amino acid peptide derived from Annexin A (a pro-resolving mediator), has shown good efficacy in the treatment of inflammatory disorders. However, the low bioavailability of Ac2-26 peptides hinders their efficacy in vivo. In this paper, we formed PEGylated lipid nanoparticles (LDNPs) by the co-assembly of l-ascorbyl palmitate (L-AP) and N-(carbonyl methoxypolyethylene glycol-2000)-1,2-distearoyl-sn‑glycero-3-phosphoethanolamine (DSPE-PEG2k) to encapsulate and deliver Ac2-26 peptides to the arthritic rats. They showed good stability and biocompatibility. After being intravenously administrated, Ac2-26 peptide-loaded PEGylated lipid nanoparticles (ADNPs) showed the prolonged in vivo circulation time and enhanced accumulation in inflamed sites. In vivo therapeutic evaluations revealed that ADNPs could attenuate synovial inflammation and improve joint pathology. Therefore, the pro-resolving therapeutic strategy using ADNPs is effective in RA treatment.
Collapse
Affiliation(s)
- Xianyan Qin
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Liming He
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Donghao Fan
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Wenlang Liang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Qin Wang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiyu Fang
- Advanced Materials Processing and Analysis Center and Department of Materials Science and Engineering, University of Central Florida, Florida 32816, United State
| |
Collapse
|
44
|
Tao W, Yurdagul A, Kong N, Li W, Wang X, Doran AC, Feng C, Wang J, Islam MA, Farokhzad OC, Tabas I, Shi J. siRNA nanoparticles targeting CaMKIIγ in lesional macrophages improve atherosclerotic plaque stability in mice. Sci Transl Med 2021; 12:12/553/eaay1063. [PMID: 32718990 DOI: 10.1126/scitranslmed.aay1063] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 02/26/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022]
Abstract
Atherosclerotic lesional macrophages express molecules that promote plaque progression, but lack of mechanisms to therapeutically target these molecules represents a major gap in translational cardiovascular research. Here, we tested the efficacy of a small interfering RNA (siRNA) nanoparticle (NP) platform targeting a plaque-destabilizing macrophage molecule-Ca2+/calmodulin-dependent protein kinase γ (CaMKIIγ). CaMKIIγ becomes activated in advanced human and mouse plaque macrophages and drives plaque necrosis by suppressing the expression of the efferocytosis receptor MerTK. When macrophage-targeted siCamk2g NPs were administered to Western diet-fed Ldlr -/- mice, the atherosclerotic lesions showed decreased CaMKIIγ and increased MerTK expression in macrophages, improved phagocytosis of apoptotic cells (efferocytosis), decreased necrotic core area, and increased fibrous cap thickness-all signs of increased plaque stability-compared with mice treated with control siRNA NPs. These findings demonstrate that atherosclerosis-promoting genes in plaque macrophages can be targeted with siRNA NPs in a preclinical model of advanced atherosclerosis.
Collapse
Affiliation(s)
- Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Arif Yurdagul
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wenliang Li
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Amanda C Doran
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Chan Feng
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Junqing Wang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mohammad Ariful Islam
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA. .,Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA.,Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
45
|
Raimondo TM, Mooney DJ. Anti-inflammatory nanoparticles significantly improve muscle function in a murine model of advanced muscular dystrophy. SCIENCE ADVANCES 2021; 7:7/26/eabh3693. [PMID: 34162554 PMCID: PMC8221619 DOI: 10.1126/sciadv.abh3693] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/10/2021] [Indexed: 05/24/2023]
Abstract
Chronic inflammation contributes to the pathogenesis of all muscular dystrophies. Inflammatory T cells damage muscle, while regulatory T cells (Tregs) promote regeneration. We hypothesized that providing anti-inflammatory cytokines in dystrophic muscle would promote proregenerative immune phenotypes and improve function. Primary T cells from dystrophic (mdx) mice responded appropriately to inflammatory or suppressive cytokines. Subsequently, interleukin-4 (IL-4)- or IL-10-conjugated gold nanoparticles (PA4, PA10) were injected into chronically injured, aged, mdx muscle. PA4 and PA10 increased T cell recruitment, with PA4 doubling CD4+/CD8- T cells versus controls. Further, 50% of CD4+/CD8- T cells were immunosuppressive Tregs following PA4, versus 20% in controls. Concomitant with Treg recruitment, muscles exhibited increased fiber area and fourfold increases in contraction force and velocity versus controls. The ability of PA4 to shift immune responses, and improve dystrophic muscle function, suggests that immunomodulatory treatment may benefit many genetically diverse muscular dystrophies, all of which share inflammatory pathology.
Collapse
Affiliation(s)
- Theresa M Raimondo
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| |
Collapse
|
46
|
Li LC, Tian Y, Xiao J, Yang Y, Wu JN, Chen Y, Zhang PH, Gao-Smith F, Wang JG, Jin SW. Dexmedetomidine promotes inflammation resolving through TGF-β1 secreted by F4/80 +Ly6G + macrophage. Int Immunopharmacol 2021; 95:107480. [PMID: 33676148 DOI: 10.1016/j.intimp.2021.107480] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 01/07/2023]
Abstract
Dexmedetomidine (DEX) is a highly selective α2-adrenoceptor agonist, which can regulate inflammatory responses. However, whether DEX interferes with the inflammation resolving remains unclear. Here, we reported the effects of DEX on zymosan-induced generalized inflammation in mice during resolution. Mice were administered intraperitoneally with DEX after the initiation of sepsis. The resolution interval (Ri), a vital resolution indice, decreased from twelve hours to eight hours after the administration of DEX. The induction of peritoneal pro-inflammatory interleukin [IL] - 1β and tumour necrosis factor-α (TNF-α) appeared to be inhibited. Of interest, the anti-inflammatory transforming growth factor-β1 (TGF-β1) but not IL-10 levels were up-regulated at twenty-four hours in the DEX group along with 1.0 mg/mice zymosan A (ZyA) treatment. The expression levels of multiple genes related to protective immune processes and clearance functions were detected and revealed the same trends. DEX markedly increased the F4/80+Ly6G+ macrophage population. Additionally, the adequate apoptotic neutrophil clearance from injury after DEX installation could be reverse by opsonization or co-instillation of TGF-β1 neutralizing antibody in vivo, promoting the inflammation-resolution programs. In conclusion, DEX post-treatment, via the increase of F4/80+Ly6G+ macrophages, provokes further secretion of TGF-β1, leading to the attenuated cytokine storm and accelerated inflammation resolving.
Collapse
Affiliation(s)
- Lin-Chao Li
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Yang Tian
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Ji Xiao
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Yi Yang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Jin-Ni Wu
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Central North Road, Wenzhou 325035, People's Republic of China
| | - Yan Chen
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Pu-Hong Zhang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China
| | - Fang Gao-Smith
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China.
| | - Jian-Guang Wang
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Central North Road, Wenzhou 325035, People's Republic of China.
| | - Sheng-Wei Jin
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, People's Republic of China.
| |
Collapse
|
47
|
Chen S, Peterson CW, Parker JA, Rice SA, Ferguson AL, Scherer NF. Data-driven reaction coordinate discovery in overdamped and non-conservative systems: application to optical matter structural isomerization. Nat Commun 2021; 12:2548. [PMID: 33953159 PMCID: PMC8099877 DOI: 10.1038/s41467-021-22794-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 03/22/2021] [Indexed: 11/09/2022] Open
Abstract
Optical matter (OM) systems consist of (nano-)particle constituents in solution that can self-organize into ordered arrays that are bound by electrodynamic interactions. They also manifest non-conservative forces, and the motions of the nano-particles are overdamped; i.e., they exhibit diffusive trajectories. We propose a data-driven approach based on principal components analysis (PCA) to determine the collective modes of non-conservative overdamped systems, such as OM structures, and harmonic linear discriminant analysis (HLDA) of time trajectories to estimate the reaction coordinate for structural transitions. We demonstrate the approach via electrodynamics-Langevin dynamics simulations of six electrodynamically-bound nanoparticles in an incident laser beam. The reaction coordinate we discover is in excellent accord with a rigorous committor analysis, and the identified mechanism for structural isomerization is in very good agreement with the experimental observations. The PCA-HLDA approach to data-driven discovery of reaction coordinates can aid in understanding and eventually controlling non-conservative and overdamped systems including optical and active matter systems.
Collapse
Affiliation(s)
- Shiqi Chen
- Department of Chemistry, University of Chicago, Chicago, IL, USA
- James Franck Institute, University of Chicago, Chicago, IL, USA
| | - Curtis W Peterson
- Department of Chemistry, University of Chicago, Chicago, IL, USA
- James Franck Institute, University of Chicago, Chicago, IL, USA
| | - John A Parker
- James Franck Institute, University of Chicago, Chicago, IL, USA
- Department of Physics, University of Chicago, Chicago, IL, USA
| | - Stuart A Rice
- Department of Chemistry, University of Chicago, Chicago, IL, USA
- James Franck Institute, University of Chicago, Chicago, IL, USA
| | - Andrew L Ferguson
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA.
| | - Norbert F Scherer
- Department of Chemistry, University of Chicago, Chicago, IL, USA.
- James Franck Institute, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
48
|
Wang Q, Qin X, Fang J, Sun X. Nanomedicines for the treatment of rheumatoid arthritis: State of art and potential therapeutic strategies. Acta Pharm Sin B 2021; 11:1158-1174. [PMID: 34094826 PMCID: PMC8144894 DOI: 10.1016/j.apsb.2021.03.013] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/11/2020] [Accepted: 01/22/2021] [Indexed: 02/06/2023] Open
Abstract
Increasing understanding of the pathogenesis of rheumatoid arthritis (RA) has remarkably promoted the development of effective therapeutic regimens of RA. Nevertheless, the inadequate response to current therapies in a proportion of patients, the systemic toxicity accompanied by long-term administration or distribution in non-targeted sites and the comprised efficacy caused by undesirable bioavailability, are still unsettled problems lying across the full remission of RA. So far, these existing limitations have inspired comprehensive academic researches on nanomedicines for RA treatment. A variety of versatile nanocarriers with controllable physicochemical properties, tailorable drug release pattern or active targeting ability were fabricated to enhance the drug delivery efficiency in RA treatment. This review aims to provide an up-to-date progress regarding to RA treatment using nanomedicines in the last 5 years and concisely discuss the potential application of several newly emerged therapeutic strategies such as inducing the antigen-specific tolerance, pro-resolving therapy or regulating the immunometabolism for RA treatments.
Collapse
Affiliation(s)
- Qin Wang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xianyan Qin
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiyu Fang
- Advanced Materials Processing and Analysis Center and Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32816, USA
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
49
|
Li Y, Chen X, Jin R, Chen L, Dang M, Cao H, Dong Y, Cai B, Bai G, Gooding JJ, Liu S, Zou D, Zhang Z, Yang C. Injectable hydrogel with MSNs/microRNA-21-5p delivery enables both immunomodification and enhanced angiogenesis for myocardial infarction therapy in pigs. SCIENCE ADVANCES 2021; 7:7/9/eabd6740. [PMID: 33627421 PMCID: PMC7904259 DOI: 10.1126/sciadv.abd6740] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 01/11/2021] [Indexed: 05/05/2023]
Abstract
Current therapeutic strategies such as angiogenic therapy and anti-inflammatory therapy for treating myocardial infarction have limited success. An effective approach may benefit from resolution of excessive inflammation combined with enhancement of angiogenesis. Here, we developed a microRNA-21-5p delivery system using functionalized mesoporous silica nanoparticles (MSNs) with additional intrinsic therapeutic effects. These nanocarriers were encapsulated into an injectable hydrogel matrix (Gel@MSN/miR-21-5p) to enable controlled on-demand microRNA-21 delivery triggered by the local acidic microenvironment. In a porcine model of myocardial infarction, we demonstrated that the released MSN complexes notably inhibited the inflammatory response by inhibiting the polarization of M1 macrophage within the infarcted myocardium, while further microRNA-21-5p delivery by MSNs to endothelial cells markedly promoted local neovascularization and rescued at-risk cardiomyocytes. The synergy of anti-inflammatory and proangiogenic effects effectively reduced infarct size in a porcine model of myocardial infarction.
Collapse
Affiliation(s)
- Yan Li
- National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Xin Chen
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an 710049, China
| | - Ronghua Jin
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an 710049, China
| | - Lu Chen
- National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Ming Dang
- School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hao Cao
- Department of Cardiac Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yun Dong
- Department of Cardiac Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Bolei Cai
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Guo Bai
- National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - J Justin Gooding
- School of Chemistry, Australian Centre for NanoMedicine and ARC Australian, Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney 2052, Australia
| | - Shiyu Liu
- Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Duohong Zou
- National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Zhiyuan Zhang
- National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Chi Yang
- National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Department of Oral Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
50
|
Wang N, Zhang Y, Liu H, Wang A, Ren T, Gou J, Zhang Y, Yin T, He H, Tang X. Toxicity Reduction and Efficacy Promotion of Doxorubicin in the Treatment of Breast Tumors Assisted by Enhanced Oral Absorption of Curcumin-Loaded Lipid-Polyester Mixed Nanoparticles. Mol Pharm 2020; 17:4533-4547. [PMID: 33201717 DOI: 10.1021/acs.molpharmaceut.0c00718] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Curcumin (CUR), a polyphenol derived from turmeric, exhibits anticancer and anti-inflammatory properties. However, it has poor water solubility, stability, and oral bioavailability. To overcome these limitations, lipid-polyester mixed nanoparticles (NPs) embedded in enteric polymer-EudragitL100-55(Eu) were formulated (CUR-NPs-Eu). NPs composed of mPEG-b-PCL have a hybrid core made up of middle chain triglyceride (MCT) and poly(ε-caprolactone) (PCL) for enhancing drug loading. The CUR-NPs with MCT content of 10% had a particle size of 121.2 ± 16.8 nm, ζ potential of -16.25 ± 1.38 mV, drug loading of 9.8%, and encapsulation efficiency of 87.4%. The transport of the CUR-NPs-Eu across Caco-2 monolayers is enhanced compared with CUR alone (1.98 ± 0.94 × 10-6 of curcumin versus 55.43 ± 6.06 × 10-6 cm/s of curcumin-loaded NPs) because of the non-disassociated nanostructure during absorption. The absolute bioavailability of CUR-NPs-Eu was 7.14%, which was drastically improved from 1.08% of the CUR suspension (CUR-Sus). Therefore, in the xenograft 4T1 tumor-bearing mice, increased drug accumulation in heart and tumor was noticed because of enhanced oral bioavailability of CUR. The chemosensitizing effect of CUR was attributed to its NF-κB reduction effect (148 ± 11.83 of DOX alone versus 104 ± 8.71 of combined therapy, ng/g tissue). The cardioprotective effect of CUR was associated with maintenance of cardiac antioxidant enzyme activity and down-regulation of NF-κB. This study provided a partial illustration of the mechanisms of chemosensitizing and cardioprotective effects of CUR utilizing the oral availability promotion effect brought by the NPs-Eu formulation. And these results further demonstrated that the capability of this NPs-Eu system in oral delivery of poorly soluble and poorly permeable drugs.
Collapse
Affiliation(s)
- Na Wang
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang 110016, China
| | | | | | - Andong Wang
- School of Pharmacy, Nantong University, Nantong 226001, China
| | - Tianyang Ren
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | | | | | | | | | | |
Collapse
|