1
|
Karunakar KK, Edwin ER, Gopalakrishnan M, Cheriyan BV, Ramaiyan V, Karthikha VS, Justin JP. Advances in nephroprotection: the therapeutic role of selenium, silver, and gold nanoparticles in renal health. Int Urol Nephrol 2025; 57:479-510. [PMID: 39312019 DOI: 10.1007/s11255-024-04212-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 09/18/2024] [Indexed: 01/29/2025]
Abstract
Renal toxicity is a disorder that causes considerable issues in healthcare systems world, highlighting the critical importance of creating alternative treatments. Metallic nanoparticles have recently emerged as promising therapeutic agents for nephroprotection because of their remarkable properties. Numerous disciplines, including medicine, biotechnology, and the food industry, are currently investigating and exploring metallic nanoparticles, such as selenium, silver, and gold, with promising outcomes. In this overview, we provide the most current findings on cutting-edge nephroprotection through metallic nanoparticles, especially selenium, silver, and gold nanoparticles. While outlining the benefits, we outline possible methods for developing metallic nanoparticles, characterization techniques, and nephroprotection therapies. Selenium nanoparticles (SeNPs) minimize oxidative stress, a primary cause of nephrotoxicity through cell regeneration which protects kidneys. Silver nanoparticles (AgNPs) have anti-inflammatory capabilities that help alleviate kidney damage and nephrotoxicity. Gold nanoparticles (AuNPs), which are biocompatible and immune-modifying, reduce inflammation and promote renal cell regeneration, indicating nephroprotective advantages. Renal protection via the use of metallic nanoparticles represents a promising new frontier in the fight against kidney disease and other renal disorders. Metallic nanoparticles of selenium, silver, and gold can protect the kidneys by lowering oxidative stress, reducing inflammation, and improving cell repair. Through their mechanisms, these nanoparticles effectively safeguard and repair kidney function, making them suitable for treating renal diseases. The potential applications of selenium, silver, and gold nanoparticles, as well as their complex modes of action and renal penetration, provide fresh hope for improving renal health and quality of life in patients with kidney disease. The current study highlights therapeutic ability, stability, nephroprotection, and toxicity profiles, as well as the importance of continuous research in this dynamic and evolving field.
Collapse
Affiliation(s)
- Karthik K Karunakar
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Elizabeth Rani Edwin
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Meenaloshini Gopalakrishnan
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Binoy Varghese Cheriyan
- Department of Pharmaceutical Chemistry, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India.
| | - Velmurugan Ramaiyan
- Department of Pharmacology, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - V S Karthikha
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Jerry Peliks Justin
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| |
Collapse
|
2
|
He S, Li X, He Y, Guo L, Dong Y, Wang L, Yang L, Li L, Huang S, Fu J, Lin Q, Zhang Z, Zhang L. High-density lipoprotein nanoparticles spontaneously target to damaged renal tubules and alleviate renal fibrosis by remodeling the fibrotic niches. Nat Commun 2025; 16:1061. [PMID: 39870661 PMCID: PMC11772610 DOI: 10.1038/s41467-025-56223-z] [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: 12/19/2023] [Accepted: 01/10/2025] [Indexed: 01/30/2025] Open
Abstract
Chronic kidney disease (CKD) ultimately causes renal fibrosis and end-stage renal disease, thus seriously threatens human health. However, current medications for CKD and fibrosis are inefficient, which is often due to poor targeting capability to renal tubule. In this study, we discover that biomimetic high-density lipoprotein (bHDL) lipid nanoparticles possess excellent targeting ability to injured tubular epithelial cells by kidney injury molecule-1(KIM-1) mediated internalization. Thus, we co-load anti-inflammatory drug triptolide (TP) and anti-fibrotic drug nintedanib (BIBF) on bHDL nanoparticles to treat CKD. Based on the targeted delivery and mutual enhancement of the efficacy of co-delivered drugs, the bHDL-based system effectively reduces kidney injury and alleviates renal fibrosis in different CKD mouse models. The mechanistic study shows that BIBF and TP synergistically remodel the fibrotic niches by decreasing inflammatory cytokines, limiting immune cell infiltration and inhibiting the activation of myofibroblasts. The bHDL vehicle also possesses high manufacturability, good safety and adequately reduces the toxicity of TP. Thus, this system is promising for the treatment of CKD and bHDL has good potential for delivering agents to damaged renal tubular epithelial cells.
Collapse
Affiliation(s)
- Shanshan He
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaoyang Li
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yuanyuan He
- College of Polymer Science and Engineering, West China School of Public Health, Med-X center of materials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Ling Guo
- National Engineering Technology Research Center for Miao Medicine, Guizhou Engineering Technology Research Center for Processing and Preparation of Traditional Chinese Medicine and Ethnic Medicine, College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, P. R. China
| | - Yunzhou Dong
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Leilei Wang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lan Yang
- College of Polymer Science and Engineering, West China School of Public Health, Med-X center of materials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Lin Li
- College of Polymer Science and Engineering, West China School of Public Health, Med-X center of materials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Shiyun Huang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jiali Fu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qing Lin
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ling Zhang
- College of Polymer Science and Engineering, West China School of Public Health, Med-X center of materials, Sichuan University, Chengdu, Sichuan, 610065, China.
| |
Collapse
|
3
|
Sabra MS, Allam EAH, El-Aal MA, Hassan NH, Mostafa AHM, Ahmed AAN. A novel pharmacological strategy using nanoparticles with glutathione and virgin coconut oil to treat gentamicin-induced acute renal failure in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:933-950. [PMID: 39093465 PMCID: PMC11787276 DOI: 10.1007/s00210-024-03303-4] [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: 05/17/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024]
Abstract
In acute renal failure (ARF), the glomerular filtration rate is reduced, and nitrogenous waste products accumulate persistently, which can last anywhere from a few hours to several days. There is hope for a reversal of the rapid loss of renal function caused by this condition. This study, with gentamicin-induced acute ARF as a prospective setting, sets out to examine the reno-protective benefits of virgin coconut oil (VCO) and GSH. Furthermore, the study evaluated the effect of medication nanoparticle compositions on several kidney function markers. The induction of ARF is achieved with the intraperitoneal injection of gentamicin. To assess renal function, rats underwent 24 h of dehydration and hunger before their deaths. The study examined various aspects, including kidney function tests, markers of oxidative stress, histology of kidney tissue, inflammatory cytokines, immunohistochemistry expression of nuclear factor-kappa B (NF-κB), and specific biomarkers for kidney tissue damage, such as kidney injury molecule-1 (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL). The results of our study indicated that the combination of VCO and GSH, using both regular and nanoparticle formulations, had a better protective impact on the kidneys compared to using either drug alone. The recovery of renal tissue and serum markers, which are symptomatic of organ damage, indicates improvement. This was also demonstrated by the reduction in tubular expression of TNF-α, IL-1β, KIM-1, and NGAL. The immunohistochemical studies showed that the combination therapy, especially with the nanoforms, greatly improved the damaged cellular changes in the kidneys, as shown by higher levels of NF-κB. The study shows that VCO and GSH, when administered individually or combined, significantly improve ARF in a gentamicin-induced rat model, highlighting potential therapeutic implications. Notably, the combined nanoparticulate formulations exhibit substantial effectiveness.
Collapse
Affiliation(s)
- Mahmoud S Sabra
- Pharmacology Department, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt.
| | - Essmat A H Allam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| | - Mohamed Abd El-Aal
- Chemistry Department, Faculty of Science, Assiut University, Assiut, 71516, Egypt
| | - Nessma H Hassan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| | - Al-Hassan Mohammed Mostafa
- Department of Pathology and Clinical Pathology, Agricultural Research Centre, Animal Health Research Institute, Assiut, 71526, Egypt
| | - Ahmed A N Ahmed
- Pharmacology Department, Faculty of Medicine, Al-Azhar University, Assiut Branch, , Assiut, 71526, Egypt
| |
Collapse
|
4
|
Zhang J, Ren X, Nie Z, You Y, Zhu Y, Chen H, Yu H, Mo GP, Su L, Peng Z, Tang MC. Dual-responsive renal injury cells targeting nanoparticles for vitamin E delivery to treat ischemia reperfusion-induced acute kidney injury. J Nanobiotechnology 2024; 22:626. [PMID: 39407248 PMCID: PMC11481814 DOI: 10.1186/s12951-024-02894-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Ischemia/reperfusion (I/R) is an important inducer of acute kidney injury (AKI), and triggers the generation of reactive oxygen species (ROS) and the expression of matrix metalloproteinase 2 (MMP2), exacerbating kidney damage. Given the immense potential of vitamin E (VitE) as a natural fat-soluble antioxidant in kidney protection, we designed the nanoparticles (NPs) that could dual respond to ROS and MMP2, aiming to accurately deliver VitE to renal injury cells. The NPs utilized Gel-SH as a sensitive receptor for MMP2 and diselenide as a sensitive receptor for ROS, while PEG2k modification enhanced biocompatibility and prevented phagocytosis mediated by the mononuclear phagocyte system. The amphiphilic Gel-SH and diselenide encapsulate the liposoluble VitE and self-assemble into the NPs with a hydrodynamic size of 69.92 nm. Both in vivo and in vitro experiments based on these NPs show good biocompatibility and the ability of target renal injury cells. In vivo kidney I/R injury models and in vitro cell hypoxia/reoxygenation models, the NPs have demonstrated effects in reducing oxidative stress and alleviating AKI. Notably, VitE can preferentially react with peroxyl radical (LOO•) than polyunsaturated fatty acid (PUFA), inhibiting the formation of carbon centered radical (L•), thereby blocking the chain reaction between PUFA and LOO• in ferroptosis. The NPs also inhibit the transition from AKI to chronic kidney disease, with few side effects. Thus, the NPs with dual-responsiveness to MMP2 and ROS for targeted delivery of VitE to renal injury cells exhibit remarkable effects in inhibiting ROS and the chain reactions of ferroptosis, making it a promising therapeutic agent against AKI caused by I/R.
Collapse
Affiliation(s)
- Jiahao Zhang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430000, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Xi Ren
- Dyson School of Design Engineering, Imperial College London, London, SW7 2BX, UK
- Institute of Materials Research, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518000, China
| | - Zhaoyang Nie
- Institute of Materials Research, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518000, China
| | - Yue You
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430000, Hubei, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Yao Zhu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430000, Hubei, China
| | - Hui Chen
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430000, Hubei, China
| | - Haichuan Yu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430000, Hubei, China
| | - Gaozhi P Mo
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430000, Hubei, China
| | - Lianjiu Su
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430000, Hubei, China.
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China.
- Department of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| | - Zhiyong Peng
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430000, Hubei, China.
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China.
- Center of Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, USA.
| | - Man-Chung Tang
- Institute of Materials Research, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518000, China.
| |
Collapse
|
5
|
Llaguno-Munive M, Vazquez-Lopez MI, Garcia-Lopez P. Solid Lipid Nanoparticles, an Alternative for the Treatment of Triple-Negative Breast Cancer. Int J Mol Sci 2024; 25:10712. [PMID: 39409041 PMCID: PMC11476567 DOI: 10.3390/ijms251910712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
Within the field of nanomedicine, which is revolutionizing cancer treatment, solid lipid nanoparticles (SLNs) have shown advantages over conventional chemotherapy when tested on cancer cells in preclinical studies. SLNs have proven to be an innovative strategy for the treatment of triple-negative breast cancer cells, providing greater efficiency than existing treatments in various studies. The encapsulation of antineoplastic drugs in SLNs has facilitated a sustained, controlled, and targeted release, which enhances therapeutic efficiency and reduces adverse effects. Moreover, the surface of SLNs can be modified to increase efficiency. For instance, the coating of these particles with polyethylene glycol (PEG) decreases their opsonization, resulting in a longer life in the circulatory system. The creation of positively charged cationic SLNs (cSLNs), achieved by the utilization of surfactants or ionic lipids with positively charged structural groups, increases their affinity for cell membranes and plasma proteins. Hyaluronic acid has been added to SLNs so that the distinct pH of tumor cells would stimulate the release of the drug and/or genetic material. The current review summarizes the recent research on SLNs, focusing on the encapsulation and transport of therapeutic agents with a cytotoxic effect on triple-negative breast cancer.
Collapse
Affiliation(s)
- Monserrat Llaguno-Munive
- Laboratorio de Física Médica, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico;
| | - Maria Ines Vazquez-Lopez
- Laboratorio de Fármaco-Oncología y Nanomedicina, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico;
| | - Patricia Garcia-Lopez
- Laboratorio de Fármaco-Oncología y Nanomedicina, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico;
| |
Collapse
|
6
|
Bui DT, Nagasaki Y. Developing poly(ethylene glycol)- b-poly(β-hydroxybutyrate)-based self-assembling prodrug for the management of cisplatin-induced acute kidney injury. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2024; 25:2382084. [PMID: 39166178 PMCID: PMC11334744 DOI: 10.1080/14686996.2024.2382084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/01/2024] [Accepted: 07/15/2024] [Indexed: 08/22/2024]
Abstract
Although β-hydroxybutyrate (BHB), one of the endogenous body ketones, possesses high bioactivities, it is rapidly consumed, metabolized, and eliminated from the body. In this study, we designed new self-assembling nanoparticles that sustainably released BHB to improve bioavailability and evaluated their efficacy in in vivo experiments using rodent animal models. Since poly(β-hydroxybutyrate) [poly(BHB)] is regarded as a polymeric prodrug that is hydrolyzed by endogenous enzymes and releases BHB in a sustained manner, our idea was to engineer hydrophobic poly(BHB) in one of the segments in the amphiphilic block copolymer, of which self-assembles in water to form nanoparticles of tens of nanometers in size (abbreviated as NanoBHB). Here, methoxy-poly(ethylene glycol) was employed as the hydrophilic segment of the block copolymer to stabilize the nanoparticles in aqueous environments, thus enabling NanoBHBs to be administrable both orally and through injection. Experimental results showed that NanoBHB has low toxicity and releases free BHB for an extended period in vitro and in vivo. Moreover, NanoBHB exhibits superior nephroprotective effects in cisplatin-induced acute kidney injury mouse models compared to low-molecular-weight (LMW) sodium BHB, suggesting the potential of NanoBHB as a sustainable release formulation to supply BHB for medicinal applications.
Collapse
Affiliation(s)
- Duc Tri Bui
- Degree Program of Pure and Applied Sciences, Graduate School of Science and Technology, University of Tsukuba, Ibaraki, Japan
| | - Yukio Nagasaki
- Department of Materials Science, Faculty of Pure and Applied Sciences, University of Tsukuba, Ibaraki, Japan
- Master’s School of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
- Center for Research in Radiation and Earth System Science (CRiES), University of Tsukuba, Ibaraki, Japan
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- High-Value Biomaterials Research and Commercialization Center (HBRCC), National Taipei University of Technology, Taipei, Taiwan
| |
Collapse
|
7
|
Prylutskyy Y, Nozdrenko D, Omelchuk O, Prylutska S, Motuziuk O, Soroсa V, Vareniuk I, Stetska V, Bogutska K, Ritter U, Piosik J. Effect of C 60 Fullerene on Muscle Injury-Induced Rhabdomyolysis and Associated Acute Renal Failure. Int J Nanomedicine 2024; 19:8043-8058. [PMID: 39130686 PMCID: PMC11316485 DOI: 10.2147/ijn.s468013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/19/2024] [Indexed: 08/13/2024] Open
Abstract
Introduction Rhabdomyolysis, as an acute stage of myopathy, causes kidney damage. It is known that this pathology is caused by the accumulation of muscle breakdown products and is associated with oxidative stress. Therefore, the present study evaluated the effect of intraperitoneal administration (dose 1 mg/kg) of water-soluble C60 fullerenes, as powerful antioxidants, on the development of rat kidney damage due to rhabdomyolysis caused by mechanical trauma of the muscle soleus of different severity (crush syndrome lasting 1 min under a pressure of 2.5, 3.5, and 4.5 kg/cm2, respectively). Methods Using tensometry, biochemical and histopathological analyses, the biomechanical parameters of muscle soleus contraction (contraction force and integrated muscle power), biochemical indicators of rat blood (concentrations of creatinine, creatine phosphokinase, urea and hydrogen peroxide, catalase and superoxide dismutase activity), glomerular filtration rate and fractional sodium excretion value, as well as pathohistological and morphometric features of muscle and kidney damages in rats on days 1, 3, 6 and 9 after the initiation of the injury were studied. Results Positive changes in biomechanical and biochemical parameters were found during the experiment by about 27-30 ± 2%, as well as a decrease in pathohistological and morphometric features of muscle and kidney damages in rats treated with water-soluble C60 fullerenes. Conclusion These findings indicate the potential application of water-soluble C60 fullerenes in the treatment of pathological conditions of the muscular system caused by rhabdomyolysis and the associated oxidative stress.
Collapse
Affiliation(s)
- Yuriy Prylutskyy
- ESC ”institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Dmytro Nozdrenko
- ESC ”institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Olexandr Omelchuk
- Faculty of Biology and Forestry, Lesya Ukrainka Volyn National University, Lutsk, Ukraine
| | - Svitlana Prylutska
- Faculty of Plant Protection, Biotechnology and Ecology, National University of Life and Environmental Science of Ukraine, Kyiv, Ukraine
| | - Olexandr Motuziuk
- ESC ”institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
- Faculty of Biology and Forestry, Lesya Ukrainka Volyn National University, Lutsk, Ukraine
| | - Vasil Soroсa
- ESC ”institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Igor Vareniuk
- ESC ”institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Viktoria Stetska
- ESC ”institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Kateryna Bogutska
- ESC ”institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Uwe Ritter
- Institute of Chemistry and Biotechnology, Technical University of Ilmenau, Ilmenau, Germany
| | - Jacek Piosik
- Intercollegiate Faculty of Biotechnology, University of Gdansk, Gdańsk, Poland
| |
Collapse
|
8
|
Vasylaki A, Ghosh P, Jaimes EA, Williams RM. Targeting the Kidneys at the Nanoscale: Nanotechnology in Nephrology. KIDNEY360 2024; 5:618-630. [PMID: 38414130 PMCID: PMC11093552 DOI: 10.34067/kid.0000000000000400] [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: 11/14/2023] [Accepted: 02/16/2024] [Indexed: 02/29/2024]
Abstract
Kidney diseases, both acute and chronic, are a substantial burden on individual and public health, and they continue to increase in frequency. Despite this and an intense focus on the study of disease mechanisms, few new therapeutic approaches have extended to the clinic. This is in part due to poor pharmacology of many, if not most, therapeutics with respect to the sites of kidney disease within the glomerulus or nephron. Considering this, within the past decade, and more pointedly over the past 2 years, there have been substantial developments in nanoparticle systems to deliver therapeutics to the sites of kidney disease. Here, we provide a broad overview of the various classes of nanomaterials that have been developed to improve therapeutic development for kidney diseases, the strategy used to provide kidney accumulation, and briefly the disease models they focused on, if any. We then focus on one specific system, polymeric mesoscale nanoparticles, which has broadly been used over 13 publications, demonstrating targeting of the tubular epithelium with 26-fold specificity compared with other organs. While there have been several nanomedicines that have advanced to the clinic in the past several decades, including mRNA-based coronavirus disease vaccines and others, none have focused on kidney diseases specifically. In total, we are confident that the rapid advancement of nanoscale-based kidney targeting and a concerted focus by clinicians, scientists, engineers, and other stakeholders will push one or more of these technologies into clinical trials over the next decade.
Collapse
Affiliation(s)
- Anastasiia Vasylaki
- Department of Biomedical Engineering, The City College of New York, New York, New York
| | - Pratyusha Ghosh
- Department of Biomedical Engineering, The City College of New York, New York, New York
| | - Edgar A. Jaimes
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Ryan M. Williams
- Department of Biomedical Engineering, The City College of New York, New York, New York
- PhD Program in Chemistry, The Graduate Center of CUNY, New York, New York
| |
Collapse
|
9
|
Ullah S, Burki S, Munir AB, Yousaf G, Shafique M. Nanocarrier-based localized and effective treatment of renal disorders: currently employed targeting strategies. Nanomedicine (Lond) 2024; 19:345-361. [PMID: 38293889 DOI: 10.2217/nnm-2023-0251] [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] [Indexed: 02/01/2024] Open
Abstract
Renal disorders pose a global health threat, with targeted drug-delivery systems emerging as a promising strategy to enhance therapy safety and efficacy. Recent efforts have harnessed targeted nanomaterials for kidney disease treatment. While some systems remain in the early stages, they show immense potential in delivering cargo to specific sites. Through animal model experimentations, it has been demonstrated to reduce systemic side effects and enhance treatment effectiveness. This review presents current strategies for kidney disorder treatment, emphasizing site-specific targeting critical to renal disease pathophysiology. Recent advancements in nano-drug delivery systems for kidney targeting are explored. Finally, toxicological aspects and prospects of the most promising kidney-targeting delivery systems are discussed in this review article.
Collapse
Affiliation(s)
- Shafi Ullah
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Superior University, Lahore, Punjab, 54000, Pakistan
| | - Samiullah Burki
- Department of Pharmacology, Jinnah Sindh Medical University, Karachi, 75510, Pakistan
| | - Abu Bakar Munir
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Superior University, Lahore, Punjab, 54000, Pakistan
| | - Ghulam Yousaf
- PAF Ruth Pfau Medical College and Hospital Faisal Base Karachi, Karachi, 75350, Pakistan
| | - Muhammad Shafique
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra, 11961, Saudi Arabia
| |
Collapse
|
10
|
Morishita M, Kida M, Motomura T, Tsukamoto R, Atari M, Higashiwaki K, Masuda K, Katsumi H, Yamamoto A. Elucidation of the Tissue Distribution and Host Immunostimulatory Activity of Exogenously Administered Probiotic-Derived Extracellular Vesicles for Immunoadjuvant. Mol Pharm 2023; 20:6104-6113. [PMID: 37931251 DOI: 10.1021/acs.molpharmaceut.3c00460] [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] [Indexed: 11/08/2023]
Abstract
Extracellular vesicles (EVs) are cell-derived nanoparticles that can be used as novel biomaterials. In the development of EVs-based therapeutic systems, it is essential to understand the in vivo fate of exogenously administered EVs and subsequent biological responses mediated by EVs. Although probiotics and microorganisms that modulate the host immune system also secrete EVs, their tissue distribution and biological reactions after administration to the host have not been sufficiently elucidated. In this study, we characterized EVs released from the probiotics Bifidobacterium longum (B-EVs) and Lactobacillus plantarum WCFS1 (L-EVs) in terms of tissue distribution and immune-activating capacity after intravenous and subcutaneous administration in mice. B-EVs and L-EVs exhibited particle sizes of approximately 100-160 nm and negative zeta potentials. These EVs contained peptidoglycan, DNA, and RNA as their cargoes. Intravenously administered B-EVs and L-EVs mainly accumulated in the liver and spleen. Furthermore, liver F4/80 and splenic CD169 macrophages took up the intravenously administered EVs. Subcutaneously administered B-EVs and L-EVs accumulated in the lymph nodes and were mainly located in the B-lymphocyte zone, indicating that exogenously administered probiotic-derived EVs showed a similar biodistribution, irrespective of the EVs-secreting cell type. Evaluation of EVs-mediated immune reactions demonstrated that intravenously administered EVs showed little activation potency. In contrast, subcutaneously administered B-EVs strongly increased the expression of inflammatory cytokine (TNF-α) and co-stimulatory molecules (CD40 and CD80) than L-EVs. These findings indicate that the subcutaneous administration of B-EVs is a useful strategy for the development of novel EVs-based immunotherapies.
Collapse
Affiliation(s)
- Masaki Morishita
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Masakatsu Kida
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Tomomi Motomura
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Rihito Tsukamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Mizuho Atari
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Kazuya Higashiwaki
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Kisa Masuda
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Hidemasa Katsumi
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| | - Akira Yamamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University, Misasagi, Yamashina-Ku, Kyoto 607-8414, Japan
| |
Collapse
|
11
|
Li R, Mao J, Zheng P, Wang R, Yang Z, Qian S. Improving the biocompatibility and antibacterial efficacy of silver nanoparticles functionalized with (LLRR) 3 antimicrobial peptide. World J Microbiol Biotechnol 2023; 40:1. [PMID: 37923918 DOI: 10.1007/s11274-023-03792-0] [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: 07/28/2023] [Accepted: 10/05/2023] [Indexed: 11/06/2023]
Abstract
The selection of effective antibiotics is becoming increasingly limited due to the emergence of bacterial resistance. Designing and developing nanoscale antibacterials is a strategy for effectively addressing the antibiotic crisis. In this work, AgNPs@AMP nanoparticles were synthesized to take advantage of the synergistic antibacterial activity of the (LLRR)3 antimicrobial peptide (AMP) and silver nanoparticles (AgNPs). Based on morphological structure characterization and biocompatibility analysis, the inhibitory properties of AgNPs@AMP on Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus) were evaluated. The results demonstrated that AMP and AgNPs were physically bound to form AgNPs@AMP nanoparticles, which had better solution stability, improved nanomaterial properties, and overcame the hemolytic activity of AMP and the cytotoxicity of AgNPs. The inhibitory activity of AgNPs@AMP against E. coli and S. aureus was significantly higher than that of AMP and AgNPs. It was capable of disrupting the morphology and internal structure of cells, damaging the cell membrane, and inhibiting the activity of enzymes related to the material-energy metabolism of the tricarboxylic acid cycle. Compared to AMP and AgNPs, AgNPs@AMP were found to effectively inhibit the infection of mouse wounds and promote their healing. Therefore, AMP-modified AgNPs can enhance their biocompatibility and antibacterial activity, and they can be further developed as a potential antimicrobial agent.
Collapse
Affiliation(s)
- Rongyu Li
- School of Basic Medical Sciences, Wannan Medical College, Wuhu, 241002, China
| | - Jiaqing Mao
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, 241000, China
| | - Peng Zheng
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, 241000, China
| | - Ruonan Wang
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, 241000, China
| | - Zicheng Yang
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, 241000, China
| | - Senhe Qian
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, 241000, China.
| |
Collapse
|
12
|
Paul P, Chacko L, Dua TK, Chakraborty P, Paul U, Phulchand V, Jha NK, Jha SK, Kandimalla R, Dewanjee S. Nanomedicines for the management of diabetic nephropathy: present progress and prospects. Front Endocrinol (Lausanne) 2023; 14:1236686. [PMID: 38027185 PMCID: PMC10656621 DOI: 10.3389/fendo.2023.1236686] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Diabetic nephropathy (DN) is a serious microvascular consequence of diabetes mellitus (DM), posing an encumbrance to public health worldwide. Control over the onset and progress of DN depend heavily on early detection and effective treatment. DN is a major contributor to end-stage renal disease, and a complete cure is yet to be achieved with currently available options. Though some therapeutic molecules have exhibited promise in treating DN complications, their poor solubility profile, low bioavailability, poor permeation, high therapeutic dose and associated toxicity, and low patient compliance apprehend their clinical usefulness. Recent research has indicated nano-systems as potential theranostic platforms displaying futuristic promise in the diagnosis and treatment of DN. Early and accurate diagnosis, site-specific delivery and retention by virtue of ligand conjugation, and improved pharmacokinetic profile are amongst the major advantages of nano-platforms, defining their superiority. Thus, the emergence of nanoparticles has offered fresh approaches to the possible diagnostic and therapeutic strategies regarding DN. The present review corroborates an updated overview of different types of nanocarriers regarding potential approaches for the diagnosis and therapy of DN.
Collapse
Affiliation(s)
- Paramita Paul
- Department of Pharmaceutical Technology, University of North Bengal, Darjeeling, India
| | - Leena Chacko
- BioAnalytical Lab, Meso Scale Discovery, Rockville, MD, United States
| | - Tarun K. Dua
- Department of Pharmaceutical Technology, University of North Bengal, Darjeeling, India
| | - Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Udita Paul
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Vishwakarma Vishal Phulchand
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Niraj K. Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Saurabh K. Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Ramesh Kandimalla
- Department of Biochemistry, Kakatiya Medical College, Warangal, Telangana, India
- Department of Applied Biology, Indian Institute of Technology, Council of Scientific & Industrial Research (CSIR), Hyderabad, India
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| |
Collapse
|
13
|
Won Lee J, Kyu Shim M, Kim H, Jang H, Lee Y, Hwa Kim S. RNAi therapies: Expanding applications for extrahepatic diseases and overcoming delivery challenges. Adv Drug Deliv Rev 2023; 201:115073. [PMID: 37657644 DOI: 10.1016/j.addr.2023.115073] [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: 04/27/2023] [Revised: 07/31/2023] [Accepted: 08/20/2023] [Indexed: 09/03/2023]
Abstract
The era of RNA medicine has become a reality with the success of messenger RNA (mRNA) vaccines against COVID-19 and the approval of several RNA interference (RNAi) agents in recent years. Particularly, therapeutics based on RNAi offer the promise of targeting intractable and previously undruggable disease genes. Recent advances have focused in developing delivery systems to enhance the poor cellular uptake and insufficient pharmacokinetic properties of RNAi therapeutics and thereby improve its efficacy and safety. However, such approach has been mainly achieved via lipid nanoparticles (LNPs) or chemical conjugation with N-Acetylgalactosamine (GalNAc), thus current RNAi therapy has been limited to liver diseases, most likely to encounter liver-targeting limitations. Hence, there is a huge unmet medical need for intense evolution of RNAi therapeutics delivery systems to target extrahepatic tissues and ultimately extend their indications for treating various intractable diseases. In this review, challenges of delivering RNAi therapeutics to tumors and major organs are discussed, as well as their transition to clinical trials. This review also highlights innovative and promising preclinical RNAi-based delivery platforms for the treatment of extrahepatic diseases.
Collapse
Affiliation(s)
- Jong Won Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Man Kyu Shim
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hyosuk Kim
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hochung Jang
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Yuhan Lee
- Department of Anesthesiology, Perioperative, and Pain Medicine, Center for Accelerated Medical Innovation & Center for Nanomedicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Sun Hwa Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
| |
Collapse
|
14
|
Sahu A, Min K, Jeon SH, Kwon K, Tae G. Self-assembled hemin-conjugated heparin with dual-enzymatic cascade reaction activities for acute kidney injury. Carbohydr Polym 2023; 316:121088. [PMID: 37321716 DOI: 10.1016/j.carbpol.2023.121088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/16/2023] [Accepted: 06/01/2023] [Indexed: 06/17/2023]
Abstract
Nanozymes have prominent catalytic activities with high stability as a substitute for unstable and expensive natural enzymes. However, most nanozymes are metal/inorganic nanomaterials, facing difficulty in clinical translation due to their unproven biosafety and limited biodegradability issues. Hemin, an organometallic porphyrin, was newly found to possess superoxide dismutase (SOD) mimetic activity along with previously known catalase (CAT) mimetic activity. However, hemin has poor bioavailability due to its low water solubility. Therefore, a highly biocompatible and biodegradable organic-based nanozyme system with SOD/CAT mimetic cascade reaction activity was developed by conjugating hemin to heparin (HepH) or chitosan (CS-H). Between them, Hep-H formed a smaller (<50 nm) and more stable self-assembled nanostructure and even possessed much higher and more stable SOD and CAT activities as well as the cascade reaction activity compared to CS-H and free hemin. Hep-H also showed a better cell protection effect against reactive oxygen species (ROS) compared to CS-H and hemin in vitro. Furthermore, Hep-H was selectively delivered to the injured kidney upon intravenous administration at the analysis time point (24 h) and exhibited excellent therapeutic effects on an acute kidney injury model by efficiently removing ROS, reducing inflammation, and minimizing structural and functional damage to the kidney.
Collapse
Affiliation(s)
- Abhishek Sahu
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Kiyoon Min
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Sae Hyun Jeon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Kiyoon Kwon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| |
Collapse
|
15
|
Hidayat M, Hasan K, Yusuf M, Sriwidodo S, Panatarani C, Joni IM. Nano Delivery Chitosan-Protein/Hydrolysate of Green Peas Bromelain (PHGPB) Synthesized by Colloidal-Spray Drying Method. Polymers (Basel) 2023; 15:polym15112546. [PMID: 37299345 DOI: 10.3390/polym15112546] [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: 04/29/2023] [Revised: 05/19/2023] [Accepted: 05/27/2023] [Indexed: 06/12/2023] Open
Abstract
Patients with chronic kidney disease (CKD) suffer persistent decreased kidney function. Previous study of protein hydrolysate of green pea (Pisum sativum) bromelain (PHGPB) has shown promising results as an antifibrotic in glucose-induced renal mesangial culture cells, by decreasing their TGF-β levels. To be effective, protein derived from PHGPB must provide adequate protein intake and reach the target organs. This paper presents a drug delivery system for the formulation of PHGPB using chitosan as polymeric nanoparticles. A PHGPB nano delivery system was synthesized by precipitation with fixed chitosan 0.1 wt.%, followed by a spray drying process at different aerosol flow rates of 1, 3, and 5 L/min. FTIR results showed that the PHGPB was entrapped in the chitosan polymer particles. Homogeneous size and spherical morphology of NDs were obtained for the chitosan-PHGPB with a flow rate of 1 L/min. Our in vivo study showed that the highest entrapment efficiency, solubility, and sustained release were achieved by the delivery system method at 1 L/min. It was concluded that the chitosan-PHGPB delivery system developed in this study improves pharmacokinetics compared to pure PHGPB.
Collapse
Affiliation(s)
- Meilinah Hidayat
- Department of Nutrition, Faculty of Medicine, Universitas Kristen Maranatha, Jalan Suria Sumantri 65, Bandung 40164, West Java, Indonesia
| | - Khomaini Hasan
- Department of Biochemistry, Faculty of Medicine, Universitas Jenderal Achmad Yani, Jalan Terusan Jenderal Sudirman, P.O. Box 148, Cimahi 40531, West Java, Indonesia
| | - Muhamad Yusuf
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jalan Raya Bandung_Sumedang KM 21, Sumedang 45363, West Java, Indonesia
| | - Sriwidodo Sriwidodo
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jalan Raya Bandung-Sumedang KM 21, Jatinangor 45363, West Java, Indonesia
| | - Camellia Panatarani
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jalan Raya Bandung_Sumedang KM 21, Sumedang 45363, West Java, Indonesia
- Functional Nano Powder (FiNder), University Center of Excellence, Universitas Padjadjaran, Jalan Raya Bandung_Sumedang KM 21, Sumedang 45363, West Java, Indonesia
| | - I Made Joni
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jalan Raya Bandung_Sumedang KM 21, Sumedang 45363, West Java, Indonesia
- Functional Nano Powder (FiNder), University Center of Excellence, Universitas Padjadjaran, Jalan Raya Bandung_Sumedang KM 21, Sumedang 45363, West Java, Indonesia
| |
Collapse
|
16
|
Sharma A, Choi HK, Lee HJ. Carbon Dots for the Treatment of Inflammatory Diseases: An Appraisal of In Vitro and In Vivo Studies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:3076119. [PMID: 37273553 PMCID: PMC10234732 DOI: 10.1155/2023/3076119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/16/2023] [Accepted: 04/25/2023] [Indexed: 06/06/2023]
Abstract
In recent decades, several studies demonstrating various applications of carbon dots (C-dots), including metal sensing, bioimaging, pH sensing, and antimicrobial activities, have been published. Recent developments have shifted this trend toward biomedical applications that target various biomarkers relevant to chronic diseases. However, relevant developments and research results regarding the anti-inflammatory properties of C-dots against inflammation-associated diseases have not been systematically reviewed. Hence, this review discusses the anti-inflammatory effects of C-dots in in vivo and in vitro models of LPS-induced inflammation, gout, cartilage tissue engineering, drug-induced inflammation, spinal cord injury, wound healing, liver diseases, stomach cancer, gastric ulcers, acute kidney and lung injury, psoriasis, fever or hypothermia, and bone tissue regeneration. The compiled studies demonstrate the promising potential of C-dots as anti-inflammatory agents for the development of new drugs.
Collapse
Affiliation(s)
- Anshul Sharma
- College of Bionanotechnology, Department of Food and Nutrition, Gachon University, Gyeonggi-do 13120, Republic of Korea
| | - Hyo-Kyoung Choi
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, Republic of Korea 55365
| | - Hae-Jeung Lee
- College of Bionanotechnology, Department of Food and Nutrition, Gachon University, Gyeonggi-do 13120, Republic of Korea
- Institute for Aging and Clinical Nutrition Research, Gachon University, Gyeonggi-do 13120, Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
17
|
Trac N, Ashraf A, Giblin J, Prakash S, Mitragotri S, Chung EJ. Spotlight on Genetic Kidney Diseases: A Call for Drug Delivery and Nanomedicine Solutions. ACS NANO 2023; 17:6165-6177. [PMID: 36988207 PMCID: PMC10145694 DOI: 10.1021/acsnano.2c12140] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/08/2023] [Indexed: 06/19/2023]
Abstract
Nanoparticles as drug delivery carriers have benefited diseases, including cancer, since the 1990s, and more recently, their promise to quickly and efficiently be mobilized to fight against global diseases such as in the COVID-19 pandemic have been proven. Despite these success stories, there are limited nanomedicine efforts for chronic kidney diseases (CKDs), which affect 844 million people worldwide and can be linked to a variety of genetic kidney diseases. In this Perspective, we provide a brief overview of the clinical status of genetic kidney diseases, background on kidney physiology and a summary of nanoparticle design that enable kidney access and targeting, and emerging technological strategies that can be applied for genetic kidney diseases, including rare and congenital kidney diseases. Finally, we conclude by discussing gaps in knowledge remaining in both genetic kidney diseases and kidney nanomedicine and collective efforts that are needed to bring together stakeholders from diverse expertise and industries to enable the development of the most relevant drug delivery strategies that can make an impact in the clinic.
Collapse
Affiliation(s)
- Noah Trac
- Department
of Biomedical Engineering, University of
Southern California, Los Angeles, California 90089, United States
| | - Anisa Ashraf
- Department
of Biomedical Engineering, University of
Southern California, Los Angeles, California 90089, United States
| | - Joshua Giblin
- Department
of Biomedical Engineering, University of
Southern California, Los Angeles, California 90089, United States
| | - Supriya Prakash
- John
A. Paulson School of Engineering & Applied Sciences, Harvard University, Allston, Massachusetts 02134, United States
- Wyss
Institute for Biologically Inspired Engineering, Boston, Massachusetts 02115, United States
| | - Samir Mitragotri
- John
A. Paulson School of Engineering & Applied Sciences, Harvard University, Allston, Massachusetts 02134, United States
- Wyss
Institute for Biologically Inspired Engineering, Boston, Massachusetts 02115, United States
| | - Eun Ji Chung
- Department
of Biomedical Engineering, University of
Southern California, Los Angeles, California 90089, United States
- Division
of Nephrology and Hypertension, Department of Medicine, Keck School
of Medicine, University of Southern California, Los Angeles, California 90033, United States
- Norris
Comprehensive Cancer Center, University
of Southern California, Los Angeles, California 90033, United States
- Eli and Edythe
Broad Center for Regenerative Medicine and Stem Cell Research, Keck
School of Medicine, University of Southern
California, Los Angeles, California 90033, United States
- Division
of Vascular Surgery and Endovascular Therapy, Department of Surgery,
Keck School of Medicine, University of Southern
California, Los Angeles, California 90033, United States
- Mork
Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90089, United States
| |
Collapse
|
18
|
Peres RAS, Silva-Aguiar RP, Teixeira DE, Peruchetti DB, Alves SAS, Leal ABC, Castro GF, Ribeiro NBS, Guimarães FV, Pinheiro AAS, Silva PMRE, Martins MA, Caruso-Neves C. Gold nanoparticles reduce tubule-interstitial injury and proteinuria in a murine model of subclinical acute kidney injury. Biochim Biophys Acta Gen Subj 2023; 1867:130314. [PMID: 36693453 DOI: 10.1016/j.bbagen.2023.130314] [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: 08/26/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Subclinical acute kidney injury (subAKI) is characterized by tubule-interstitial injury without significant changes in glomerular function. SubAKI is associated with the pathogenesis and progression of acute and chronic kidney diseases. Currently, therapeutic strategies to treat subAKI are limited. The use of gold nanoparticles (AuNPs) has shown promising benefits in different models of diseases. However, their possible effects on subAKI are still unknown. Here, we investigated the effects of AuNPs on a mouse model of subAKI. Animals with subAKI showed increased functional and histopathologic markers of tubular injury. There were no changes in glomerular function and structure. The animals with subAKI also presented an inflammatory profile demonstrated by activation of Th1 and Th17 cells in the renal cortex. This phenotype was associated with decreased megalin-mediated albumin endocytosis and expression of proximal tubular megalin. AuNP treatment prevented tubule-interstitial injury induced by subAKI. This effect was associated with a shift to an anti-inflammatory Th2 response. Furthermore, AuNP treatment preserved megalin-mediated albumin endocytosis in vivo and in vitro. AuNPs were not nephrotoxic in healthy mice. These results suggest that AuNPs have a protective effect in the tubule-interstitial injury observed in subAKI, highlighting a promising strategy as a future antiproteinuric treatment.
Collapse
Affiliation(s)
- Rodrigo A S Peres
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo P Silva-Aguiar
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Douglas E Teixeira
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diogo B Peruchetti
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sarah A S Alves
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anna Beatriz C Leal
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Guilherme F Castro
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Natalia B S Ribeiro
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Fernanda V Guimarães
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Ana Acacia S Pinheiro
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
| | - Patrícia M R E Silva
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
| | - Marco A Martins
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
| | - Celso Caruso-Neves
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.
| |
Collapse
|
19
|
Passively-targeted mitochondrial tungsten-based nanodots for efficient acute kidney injury treatment. Bioact Mater 2023; 21:381-393. [PMID: 36185743 PMCID: PMC9483742 DOI: 10.1016/j.bioactmat.2022.08.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/05/2022] [Accepted: 08/20/2022] [Indexed: 12/25/2022] Open
Abstract
Acute kidney injury (AKI) can lead to loss of kidney function and a substantial increase in mortality. The burst of reactive oxygen species (ROS) plays a key role in the pathological progression of AKI. Mitochondrial-targeted antioxidant therapy is very promising because mitochondria are the main source of ROS in AKI. Antioxidant nanodrugs with actively targeted mitochondria have achieved encouraging success in many oxidative stress-induced diseases. However, most strategies to actively target mitochondria make the size of nanodrugs too large to pass through the glomerular system to reach the renal tubules, the main damage site of AKI. Here, an ultra-small Tungsten-based nanodots (TWNDs) with strong ROS scavenging can be very effective for treatment of AKI. TWNDs can reach the tubular site after crossing the glomerular barrier, and enter the mitochondria of the renal tubule without resorting to complex active targeting strategies. To our knowledge, this is the first time that ultra-small negatively charged nanodots can be used to passively target mitochondrial therapy for AKI. Through in-depth study of the therapeutic mechanism, such passive mitochondria-targeted TWNDs are highly effective in protecting mitochondria by reducing mitochondrial ROS and increasing mitophagy. In addition, TWNDs can also reduce the infiltration of inflammatory cells. This work provides a new way to passively target mitochondria for AKI, and give inspiration for the treatment of many major diseases closely related to mitochondria, such as myocardial infarction and cerebral infarction. Mitochondrial-targeted antioxidant therapy is a very promising treatment for AKI. TWNDs have a strong ability to scavenge ROS, and their small size allows them to cross the glomerular filtration barrier. TWNDs passively target the mitochondria of renal tubular cells and are highly effective in restoring renal function.
Collapse
|
20
|
Mourão CF, Lowenstein A. The Use of Alb-PRF as a Drug Delivery System for Malignant Lesion Treatment. REVISTA BRASILEIRA DE CANCEROLOGIA 2023. [DOI: 10.32635/2176-9745.rbc.2023v69n2.3762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023] Open
Abstract
Bioengineering is an area of biotechnology that offers tools for different therapeutic strategies, including tissue therapies. For this, cells are associated with growth factors and scaffolds composed of biocompatible biomaterials capable of promoting the regeneration of tissues, such as bones. Autologous biomaterials produced from human peripheral blood have a proven ability to produce/release activating factors for tissue repair, with potential application as a vehicle for carrying substances for local release. The present idea proposes the use of a new blood-derived concentrate, Alb-PRF, an activated plasma albumin gel with the platelet-rich fibrin in the liquid phase (Liquid PRF). Alb-PRF can be able to stabilize the fibrin matrix of the scaffold, acting as a drug delivery system vehicle in malignant lesions.
Collapse
|
21
|
Almeida B, Domingues C, Mascarenhas-Melo F, Silva I, Jarak I, Veiga F, Figueiras A. The Role of Cyclodextrins in COVID-19 Therapy-A Literature Review. Int J Mol Sci 2023; 24:2974. [PMID: 36769299 PMCID: PMC9918006 DOI: 10.3390/ijms24032974] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
Coronavirus disease-19 (COVID-19) emerged in December 2019 and quickly spread, giving rise to a pandemic crisis. Therefore, it triggered tireless efforts to identify the mechanisms of the disease, how to prevent and treat it, and to limit and hamper its global dissemination. Considering the above, the search for prophylactic approaches has led to a revolution in the reglementary pharmaceutical pipeline, with the approval of vaccines against COVID-19 in an unprecedented way. Moreover, a drug repurposing scheme using regulatory-approved antiretroviral agents is also being pursued. However, their physicochemical characteristics or reported adverse events have sometimes limited their use. Hence, nanotechnology has been employed to potentially overcome some of these challenges, particularly cyclodextrins. Cyclodextrins are cyclic oligosaccharides that present hydrophobic cavities suitable for complexing several drugs. This review, besides presenting studies on the inclusion of antiviral drugs in cyclodextrins, aims to summarize some currently available prophylactic and therapeutic schemes against COVID-19, highlighting those that already make use of cyclodextrins for their complexation. In addition, some new therapeutic approaches are underscored, and the potential application of cyclodextrins to increase their promising application against COVID-19 will be addressed. This review describes the instances in which the use of cyclodextrins promotes increased bioavailability, antiviral action, and the solubility of the drugs under analysis. The potential use of cyclodextrins as an active ingredient is also covered. Finally, toxicity and regulatory issues as well as future perspectives regarding the use of cyclodextrins in COVID-19 therapy will be provided.
Collapse
Affiliation(s)
- Beatriz Almeida
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Cátia Domingues
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- LAQV-REQUIMTE, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Filipa Mascarenhas-Melo
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- LAQV-REQUIMTE, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Inês Silva
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ivana Jarak
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Francisco Veiga
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- LAQV-REQUIMTE, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Figueiras
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- LAQV-REQUIMTE, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
22
|
Saladino GM, Vogt C, Brodin B, Shaker K, Kilic NI, Andersson K, Arsenian-Henriksson M, Toprak MS, Hertz HM. XFCT-MRI hybrid multimodal contrast agents for complementary imaging. NANOSCALE 2023; 15:2214-2222. [PMID: 36625091 DOI: 10.1039/d2nr05829d] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Multimodal contrast agents in biomedical imaging enable the collection of more comprehensive diagnostic information. In the present work, we design hybrid ruthenium-decorated superparamagnetic iron oxide nanoparticles (NPs) as the contrast agents for both magnetic resonance imaging (MRI) and X-ray fluorescence computed tomography (XFCT). The NPs are synthesized via a one-pot polyol hot injection route, in diethylene glycol. In vivo preclinical studies demonstrate the possibility of correlative bioimaging with these contrast agents. The complementarity allows accurate localization, provided by the high contrast of the soft tissues in MRI combined with the elemental selectivity of XFCT, leading to NP detection with high specificity and resolution. We envision that this multimodal imaging could find future applications for early tumor diagnosis, improved long-term treatment monitoring, and enhanced radiotherapy planning.
Collapse
Affiliation(s)
- Giovanni Marco Saladino
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691 Stockholm, Sweden.
| | - Carmen Vogt
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691 Stockholm, Sweden.
| | - Bertha Brodin
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691 Stockholm, Sweden.
| | - Kian Shaker
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691 Stockholm, Sweden.
| | - Nuzhet Inci Kilic
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691 Stockholm, Sweden.
| | - Kenth Andersson
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691 Stockholm, Sweden.
| | - Marie Arsenian-Henriksson
- Department of Microbiology Tumor and Cell Biology (MTC), Karolinska Institute, SE 17165 Stockholm, Sweden
| | - Muhammet Sadaka Toprak
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691 Stockholm, Sweden.
| | - Hans Martin Hertz
- Department of Applied Physics, Biomedical and X-Ray Physics, KTH Royal Institute of Technology, SE 10691 Stockholm, Sweden.
| |
Collapse
|
23
|
Peng Z, Wang H, Zheng J, Wang J, Xiang Y, Liu C, Ji M, Liu H, Pan L, Qin X, Qu X. Is the proximal tubule the focus of tubulointerstitial fibrosis? Heliyon 2023; 9:e13508. [PMID: 36846656 PMCID: PMC9950842 DOI: 10.1016/j.heliyon.2023.e13508] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/15/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Tubulointerstitial fibrosis (TIF), a common end result of almost all progressive chronic kidney diseases (CKD), is also the best predictor of kidney survival. Almost all cells in the kidney are involved in the progression of TIF. Myofibroblasts, the primary producers of extracellular matrix, have previously received a great deal of attention; however, a large body of emerging evidence reveals that proximal tubule (PT) plays a central role in TIF progression. In response to injury, renal tubular epithelial cells (TECs) transform into inflammatory and fibroblastic cells, producing various bioactive molecules that drive interstitial inflammation and fibrosis. Here we reviewed the increasing evidence for the key role of the PT in promoting TIF in tubulointerstitial and glomerular injury and discussed the therapeutic targets and carrier systems involving the PT that holds particular promise for treating patients with fibrotic nephropathy.
Collapse
Affiliation(s)
- Zhi Peng
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Hui Wang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Jiaoyun Zheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jie Wang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Chi Liu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Ming Ji
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Huijun Liu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Lang Pan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Xiaoqun Qin
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| |
Collapse
|
24
|
Benešová M, Bernatová S, Mika F, Pokorná Z, Ježek J, Šiler M, Samek O, Růžička F, Rebrošová K, Zemánek P, Pilát Z. SERS-Tags: Selective Immobilization and Detection of Bacteria by Strain-Specific Antibodies and Surface-Enhanced Raman Scattering. BIOSENSORS 2023; 13:182. [PMID: 36831948 PMCID: PMC9954015 DOI: 10.3390/bios13020182] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/16/2023] [Accepted: 01/20/2023] [Indexed: 06/18/2023]
Abstract
Efficient separation and sensitive identification of pathogenic bacterial strains is essential for a prosperous modern society, with direct applications in medical diagnostics, drug discovery, biodefense, and food safety. We developed a fast and reliable method for antibody-based selective immobilization of bacteria from suspension onto a gold-plated glass surface, followed by detection using strain-specific antibodies linked to gold nanoparticles decorated with a reporter molecule. The reporter molecules are subsequently detected by surface-enhanced Raman spectroscopy (SERS). Such a multi-functionalized nanoparticle is called a SERS-tag. The presented procedure uses widely accessible and cheap materials for manufacturing and functionalization of the nanoparticles and the immobilization surfaces. Here, we exemplify the use of the produced SERS-tags for sensitive single-cell detection of opportunistic pathogen Escherichia coli, and we demonstrate the selectivity of our method using two other bacterial strains, Staphylococcus aureus and Serratia marcescens, as negative controls. We believe that the described approach has a potential to inspire the development of novel medical diagnostic tools for rapid identification of bacterial pathogens.
Collapse
Affiliation(s)
- Markéta Benešová
- Institute of Scientific Instruments of the Czech Academy of Sciences, v.v.i., Královopolská 147, 612 64 Brno, Czech Republic
| | - Silvie Bernatová
- Institute of Scientific Instruments of the Czech Academy of Sciences, v.v.i., Královopolská 147, 612 64 Brno, Czech Republic
| | - Filip Mika
- Institute of Scientific Instruments of the Czech Academy of Sciences, v.v.i., Královopolská 147, 612 64 Brno, Czech Republic
| | - Zuzana Pokorná
- Institute of Scientific Instruments of the Czech Academy of Sciences, v.v.i., Královopolská 147, 612 64 Brno, Czech Republic
| | - Jan Ježek
- Institute of Scientific Instruments of the Czech Academy of Sciences, v.v.i., Královopolská 147, 612 64 Brno, Czech Republic
| | - Martin Šiler
- Institute of Scientific Instruments of the Czech Academy of Sciences, v.v.i., Královopolská 147, 612 64 Brno, Czech Republic
| | - Ota Samek
- Institute of Scientific Instruments of the Czech Academy of Sciences, v.v.i., Královopolská 147, 612 64 Brno, Czech Republic
| | - Filip Růžička
- Department of Microbiology, Faculty of Medicine of Masaryk University and St. Anne’s University Hospital, Pekařská 53, 656 91 Brno, Czech Republic
| | - Katarina Rebrošová
- Department of Microbiology, Faculty of Medicine of Masaryk University and St. Anne’s University Hospital, Pekařská 53, 656 91 Brno, Czech Republic
| | - Pavel Zemánek
- Institute of Scientific Instruments of the Czech Academy of Sciences, v.v.i., Královopolská 147, 612 64 Brno, Czech Republic
| | - Zdeněk Pilát
- Institute of Scientific Instruments of the Czech Academy of Sciences, v.v.i., Královopolská 147, 612 64 Brno, Czech Republic
| |
Collapse
|
25
|
Nanoparticle-antibody conjugate-based immunoassays for detection of CKD-associated biomarkers. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
26
|
Hebert JF, Burfeind KG, Malinoski D, Hutchens MP. Molecular Mechanisms of Rhabdomyolysis-Induced Kidney Injury: From Bench to Bedside. Kidney Int Rep 2022; 8:17-29. [PMID: 36644345 PMCID: PMC9831947 DOI: 10.1016/j.ekir.2022.09.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/20/2022] [Accepted: 09/26/2022] [Indexed: 01/18/2023] Open
Abstract
Rhabdomyolysis-induced acute kidney injury (RIAKI) occurs following damage to the muscular sarcolemma sheath, resulting in the leakage of myoglobin and other metabolites that cause kidney damage. Currently, the sole recommended clinical treatment for RIAKI is aggressive fluid resuscitation, but other potential therapies, including pretreatments for those at risk for developing RIAKI, are under investigation. This review outlines the mechanisms and clinical significance of RIAKI, investigational treatments and their specific targets, and the status of ongoing research trials.
Collapse
Affiliation(s)
- Jessica F. Hebert
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA,Correspondence: Jessica F. Hebert, Oregon Health and Science University, Department of Anesthesiology and Perioperative Medicine, Portland, Oregon, USA.
| | - Kevin G. Burfeind
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Darren Malinoski
- Department of Surgery, Oregon Health and Science University, Portland, Oregon, USA,Operative Care Division, Portland Veterans Administration Medical Center, Portland, Oregon, USA
| | - Michael P. Hutchens
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, Oregon, USA,Operative Care Division, Portland Veterans Administration Medical Center, Portland, Oregon, USA
| |
Collapse
|
27
|
Obeidat WM, Gharaibeh SF, Jaradat A. The Influence of Drugs Solubilities and Chitosan-TPP Formulation Parameters on the Mean Hydrodynamic Diameters and Drugs Entrapment Efficiencies into Chitosan-TPP Nanoparticles. AAPS PharmSciTech 2022; 23:262. [PMID: 36138243 DOI: 10.1208/s12249-022-02420-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/09/2022] [Indexed: 11/30/2022] Open
Abstract
Chitosan is a natural, biocompatible polymer. The aim of this work was to study the influence of drug solubility in 2% v/v acetic acid, formulation parameters, on mean hydrodynamic (MHD) diameters and drug entrapment efficiencies (% EE) into chitosan-TPP nanoparticles (NPs). Drugs of different aqueous solubilities with nearly similar molecular weights were chosen and admixed at several concentrations in 2% acetic acid at different chitosan concentrations and at fixed chitosan to TPP concentrations/volumes ratios. The NPs were freeze-dried, and the supernatants were utilized to determine % EE. Theophylline- and antipyrine-loaded NPs showed the best short-term physical stability in terms of MHD diameters. Antipyrine-loaded NPs possessed the larger MHD diameters, while vitamin C-loaded NPs showed the smallest ones. The relationships between the ratio of drug concentration relative to their solubilities in acetic acid were almost linear for antipyrine and vitamin C-loaded NPs when plotted against and the MHD diameters of NPs, and linear for antipyrine- and theophylline-loaded NPs when plotted against % EE with antipyrine NPs possessing the highest % EE. However, vitamin C- and propylthiouracil-loaded NPs exhibited curvilinear patterns with comparatively lower % EE. The concentration of chitosan, drug solubility in dispersion medium, and the ratio of the concentration of admixed drug relative to its solubility in dispersion medium were found critical in determining % EE and MHD diameters of NPs. It was evident that drugs with extremely low or high solubilities in dispersion medium resulted in low % EE when admixed at both low and high concentrations.
Collapse
Affiliation(s)
- Wasfy M Obeidat
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan.
| | | | - Abdolelah Jaradat
- Faculty of Pharmacy, Applied Pharmaceutical Sciences and Clinical Pharmacy, Isra University, 11622, Al Hezam Road, Amman, Jordan
| |
Collapse
|
28
|
Xiang H, Zhang C, Xiong J. Emerging role of extracellular vesicles in kidney diseases. Front Pharmacol 2022; 13:985030. [PMID: 36172178 PMCID: PMC9510773 DOI: 10.3389/fphar.2022.985030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Many types of renal disease eventually progress to end-stage renal disease, which can only be maintained by renal replacement therapy. Therefore, kidney diseases now contribute significantly to the health care burden in many countries. Many new advances and strategies have been found in the research involving kidney diseases; however, there is still no efficient treatment. Extracellular vesicles (EVs) are cell-derived membrane structures, which contains proteins, lipids, and nucleic acids. After internalization by downstream cells, these components can still maintain functional activity and regulate the phenotype of downstream cells. EVs drive the information exchange between cells and tissues. Majority of the cells can produce EVs; however, its production, contents, and transportation may be affected by various factors. EVs have been proved to play an important role in the occurrence, development, and treatment of renal diseases. However, the mechanism and potential applications of EVs in kidney diseases remain unclear. This review summarizes the latest research of EVs in renal diseases, and provides new therapeutic targets and strategies for renal diseases.
Collapse
|
29
|
Liu C, Yan P, Xu X, Zhou W, Prakash DR, Wang S, Zhou J, Wang R, Huang H, Chen J, Zhang H, Shen J. In Vivo Kidney Allograft Endothelial Specific Scavengers for On-Site Inflammation Reduction under Antibody-Mediated Rejection. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106746. [PMID: 35235710 DOI: 10.1002/smll.202106746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/21/2022] [Indexed: 06/14/2023]
Abstract
Kidney transplantation is the most effective therapy for patients with end-stage renal disease. However, antibody-mediated rejection (ABMR) threatens long-term survival of renal grafts. Although ABMR can be controlled by donor-specific antibody clearance and B- or (and) plasma-cells inhibition, the treatment often causes severe side effects in patients. Therefore, there is need to explore site-specific scavengers. In this study, a nanovehicle carrying an anti-inflammatory drug is developed with complement component 4d targeting, a specific biomarker expressed on allograft endothelium under ABMR. Moreover, the nanovehicle is endowed with photothermal properties to control drug release. Analysis through systematic in vitro and in vivo toxicity, non-invasive targeted imaging, and in situ remote controlled drug release show the nanovehicle specifically targets allograft kidney endothelium, releases an anti-inflammatory drug, methylprednisolone, locally upon laser irradiation, and promotes recovery of injured endothelium, without affecting systemic inflammation or innate immune responses. This strategy has the potential for future clinical application in ABMR treatment.
Collapse
Affiliation(s)
- Chang Liu
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
| | - Pengpeng Yan
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Xiaoyu Xu
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200031, China
| | - Wenhui Zhou
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
| | | | - Shuqi Wang
- Institute for Translational Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310029, China
| | - Junnian Zhou
- Experimental Hematology and Biochemistry Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
- Stem Cell and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Rending Wang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Hongfeng Huang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jia Shen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| |
Collapse
|
30
|
Bazin D, Lucas IT, Rouzière S, Elkaim E, Mocuta C, Réguer S, Reid DG, Mathurin J, Dazzi A, Deniset-Besseau A, Petay M, Frochot V, Haymann JP, Letavernier E, Verpont MC, Foy E, Bouderlique E, Colboc H, Daudon M. Profile of an “at cutting edge” pathology laboratory for pathological human deposits: from nanometer to in vivo scale analysis on large scale facilities. CR CHIM 2022. [DOI: 10.5802/crchim.199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
31
|
Feng J, Liu Y, Pan X, Jin F, Wu L, Chen J, Wan B, Zhang X, Rodrigues LR, Zhang Y. Acid-Directed Electrostatic Self-Assembly Generates Charge-Reversible Bacteria for Enhanced Tumor Targeting and Low Tissue Trapping. ACS APPLIED MATERIALS & INTERFACES 2022; 14:36411-36424. [PMID: 35917371 DOI: 10.1021/acsami.2c08684] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Despite recent preclinical progress with oncolytic bacteria in cancer therapy, dose-limiting toxicity has been a long-standing challenge for clinical application. Genetic and chemical modifications for enhancing the bacterial tumor-targeting ability have been unable to establish a balance between increasing its specificity and effectiveness while decreasing side effects. Herein, we report a simple, highly efficient method for rapidly self-assembling a clinically used lipid on bacterium and for reducing its minimum effective dose and toxicity to normal organs. The resultant bacteria present the ability to reverse-charge between neutral and acidic solutions, thus enabling weak interactions with the negatively charged normal cells, hence increasing their biocompatibility with blood cells and with the immune system. Additionally, the lipid-coated bacteria exhibit a longer blood circulation lifetime and low tissue trapping compared with the wild-type strains. Thereby, the engineered bacteria show enhanced tumor specificity and effectiveness even at low doses. Multiple visualization techniques are used for vividly demonstrating the time course of bacterial circulation in the blood and normal organs after intravenous administration. We believe that these methods for biointerfacial lipid self-assembly and evaluation of bacterial systemic circulation possess vast potential in exquisitely fabricating engineered bacteria for cancer therapy in the future.
Collapse
Affiliation(s)
- Jing Feng
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, P. R. China
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211100, P. R. China
| | - Yiting Liu
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, P. R. China
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211100, P. R. China
| | - Xia Pan
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, P. R. China
| | - Fa Jin
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, P. R. China
| | - Liangquan Wu
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, P. R. China
| | - Jianquan Chen
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, P. R. China
| | - Bing Wan
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, P. R. China
| | - Xiuwei Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, P. R. China
| | - Lígia R Rodrigues
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Yunlei Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, P. R. China
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211100, P. R. China
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, P. R. China
| |
Collapse
|
32
|
Genetic Kidney Diseases (GKDs) Modeling Using Genome Editing Technologies. Cells 2022; 11:cells11091571. [PMID: 35563876 PMCID: PMC9105797 DOI: 10.3390/cells11091571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 02/05/2023] Open
Abstract
Genetic kidney diseases (GKDs) are a group of rare diseases, affecting approximately about 60 to 80 per 100,000 individuals, for which there is currently no treatment that can cure them (in many cases). GKDs usually leads to early-onset chronic kidney disease, which results in patients having to undergo dialysis or kidney transplant. Here, we briefly describe genetic causes and phenotypic effects of six GKDs representative of different ranges of prevalence and renal involvement (ciliopathy, glomerulopathy, and tubulopathy). One of the shared characteristics of GKDs is that most of them are monogenic. This characteristic makes it possible to use site-specific nuclease systems to edit the genes that cause GKDs and generate in vitro and in vivo models that reflect the genetic abnormalities of GKDs. We describe and compare these site-specific nuclease systems (zinc finger nucleases (ZFNs), transcription activator-like effect nucleases (TALENs) and regularly clustered short palindromic repeat-associated protein (CRISPR-Cas9)) and review how these systems have allowed the generation of cellular and animal GKDs models and how they have contributed to shed light on many still unknown fields in GKDs. We also indicate the main obstacles limiting the application of these systems in a more efficient way. The information provided here will be useful to gain an accurate understanding of the technological advances in the field of genome editing for GKDs, as well as to serve as a guide for the selection of both the genome editing tool and the gene delivery method most suitable for the successful development of GKDs models.
Collapse
|
33
|
Advancements in nanomedicines for the detection and treatment of diabetic kidney disease. BIOMATERIALS AND BIOSYSTEMS 2022; 6:100047. [PMID: 36824160 PMCID: PMC9934479 DOI: 10.1016/j.bbiosy.2022.100047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/22/2022] [Accepted: 03/27/2022] [Indexed: 12/18/2022] Open
Abstract
In the diabetic kidneys, morbidities such as accelerated ageing, hypertension and hyperglycaemia create a pro-inflammatory microenvironment characterised by extensive fibrogenesis. Radiological techniques are not yet optimised generating inconsistent and non-reproducible data. The gold standard procedure to assess renal fibrosis is kidney biopsy, followed by histopathological assessment. However, this method is risky, invasive, subjective and examines less than 0.01% of kidney tissue resulting in diagnostic errors. As such, less than 10% of patients undergo kidney biopsy, limiting the accuracy of the current diabetic kidney disease (DKD) staging method. Standard treatments suppress the renin-angiotensin system to control hypertension and use of pharmaceuticals aimed at controlling diabetes have shown promise but can cause hypoglycaemia, diuresis and malnutrition as a result of low caloric intake. New approaches to both diagnosis and treatment are required. Nanoparticles (NPs) are an attractive candidate for managing DKD due to their ability to act as theranostic tools that can carry drugs and enhance image contrast. NP-based point-of-care systems can provide physiological information previously considered unattainable and provide control over the rate and location of drug release. Here we discuss the use of nanotechnology in renal disease, its application to both the treatment and diagnosis of DKD. Finally, we propose a new method of NP-based DKD classification that overcomes the current systems limitations.
Collapse
|
34
|
Long-Term Clearance and Biodistribution of Magnetic Nanoparticles Assessed by AC Biosusceptometry. MATERIALS 2022; 15:ma15062121. [PMID: 35329574 PMCID: PMC8948936 DOI: 10.3390/ma15062121] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/08/2021] [Accepted: 12/16/2021] [Indexed: 12/12/2022]
Abstract
Once administered in an organism, the physiological parameters of magnetic nanoparticles (MNPs) must be addressed, as well as their possible interactions and retention and elimination profiles. Alternating current biosusceptometry (ACB) is a biomagnetic detection system used to detect and quantify MNPs. The aims of this study were to evaluate the biodistribution and clearance of MNPs profiles through long-time in vivo analysis and determine the elimination time carried out by the association between the ACB system and MnFe2O4 nanoparticles. The liver, lung, spleen, kidneys, and heart and a blood sample were collected for biodistribution analysis and, for elimination analysis, and over 60 days. During the period analyzed, the animal’s feces were also collectedd. It was possible to notice a higher uptake by the liver and the spleen due to their characteristics of retention and uptake. In 60 days, we observed an absence of MNPs in the spleen and a significant decay in the liver. We also determined the MNPs’ half-life through the liver and the spleen elimination. The data indicated a concentration decay profile over the 60 days, which suggests that, in addition to elimination via feces, there is an endogenous mechanism of metabolization or possible agglomeration of MNPs, resulting in loss of ACB signal intensity.
Collapse
|
35
|
Zhou L, Ye Z, Zhang E, Chen L, Hou Y, Lin J, Huang F, Yuan Z. Co-Delivery of Dexamethasone and Captopril by α8 Integrin Antibodies Modified Liposome-PLGA Nanoparticle Hybrids for Targeted Anti-Inflammatory/Anti-Fibrosis Therapy of Glomerulonephritis. Int J Nanomedicine 2022; 17:1531-1547. [PMID: 35388271 PMCID: PMC8978694 DOI: 10.2147/ijn.s347164] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/10/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Mesangial cells-mediated glomerulonephritis refers to a category of immunologically mediated glomerular injuries characterized by infiltration of circulating inflammatory cells, proliferation of mesangial cells, and the common pathological manifestation to the later stage is renal fibrosis, accompanied by excessive accumulation of extracellular matrix (ECM). Treatment regimens include glucocorticoids and immunosuppressive agents, but their off-target distribution causes severe systemic toxicity. Hence, specific co-delivery of “anti-inflammatory/anti-fibrosis” drugs to the glomerular mesangial cell (MC) region is expected to produce better therapeutic effects. Methods A novel kidney-targeted nanocarrier drug delivery system targeting MCs was constructed using passive targeting resulting from the difference in pore size between the glomerular endothelial layer and the basement membrane, and active targeting based on the specific binding of antibodies and antigens. Specifically, a liposome-nanoparticle hybrid (PLGA-LNHy) was formed by coating the surface of PLGA nanoparticles (NPs) with a phospholipid bilayer, and then PLGA-LNHy was co-modified with PEG and α8 integrin antibodies to obtain PLGA immunoliposomes (PLGA-ILs). Results The results showed that the obtained NPs had a core-shell structure, uniform and suitable particle size (119.1 ± 2.31 nm), low cytotoxicity, and good mesangial cell-entry ability, which can successfully accumulate in the glomerular MC region. Both dexamethasone (DXMS) and captopril (CAP) were loaded onto PLGA-ILs with a drug loading of 10.22 ± 1.00% for DXMS and 6.37 ± 0.25% for CAP (DXMS/CAP@PLGA-ILs). In vivo pharmacodynamics showed that DXMS/CAP@PLGA-ILs can effectively improve the pathological changes in the mesangial area and positive expression of proliferating cell nuclear antigen (PCNA) in glomeruli as well as reduce the expression of inflammatory factors, fibrotic factors and reactive oxygen species (ROS). Thus, renal inflammation and fibrosis were relieved. Conclusion We have provided a strategy to increase nanoparticle accumulation in MCs with the potential to implement regulatory effects of anti-inflammatory and anti-fibrosis in glomerulonephritis (GN).
Collapse
Affiliation(s)
- Liuting Zhou
- Department of Osteoporosis, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, People’s Republic of China
| | - Zhenyan Ye
- School of Clinical Medical; Chengdu Medical College, Chengdu, People’s Republic of China
| | - E Zhang
- Officers college of PAP, Chengdu, Sichuan, People’s Republic of China
| | - Li Chen
- Department of Osteoporosis, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Yitong Hou
- Department of Osteoporosis, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - JuChun Lin
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, People’s Republic of China
| | - Fenglan Huang
- Department of Osteoporosis, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- Correspondence: Fenglan Huang, Email
| | - Zhixiang Yuan
- College of Pharmacy, Southwest Minzu University, Chengdu, Sichuan, People’s Republic of China
| |
Collapse
|
36
|
Emergent players in renovascular disease. Clin Sci (Lond) 2022; 136:239-256. [PMID: 35129198 DOI: 10.1042/cs20210509] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 02/07/2023]
Abstract
Renovascular disease (RVD) remains a common etiology of secondary hypertension. Recent clinical trials revealed unsatisfactory therapeutic outcomes of renal revascularization, leading to extensive investigation to unravel key pathophysiological mechanisms underlying irreversible functional loss and structural damage in the chronically ischemic kidney. Research studies identified complex interactions among various players, including inflammation, fibrosis, mitochondrial injury, cellular senescence, and microvascular remodeling. This interplay resulted in a shift of our understanding of RVD from a mere hemodynamic disorder to a pro-inflammatory and pro-fibrotic pathology strongly influenced by systemic diseases like metabolic syndrome (MetS), hypertension, diabetes mellitus, and hyperlipidemia. Novel diagnostic approaches have been tested for early detection and follow-up of RVD progression, using new imaging techniques and biochemical markers of renal injury and dysfunction. Therapies targeting some of the pathological pathways governing the development of RVD have shown promising results in animal models, and a few have moved from bench to clinical research. This review summarizes evolving understanding in chronic ischemic kidney injury.
Collapse
|
37
|
Gao C, Zhang Q, Yang Y, Li Y, Lin W. Recent trends in therapeutic application of engineered blood purification materials for kidney disease. Biomater Res 2022; 26:5. [PMID: 35120554 PMCID: PMC8815201 DOI: 10.1186/s40824-022-00250-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 01/10/2022] [Indexed: 12/11/2022] Open
Abstract
Blood purification is a commonly used method to remove excess metabolic waste in the blood in renal replacement therapy. The sufficient removal of these toxins from blood can reduce complications and improve survival lifetime in dialysis patients. However, the current biological blood purification materials in clinical practice are not ideal, where there is an unmet need for producing novel materials that have better biocompatibility, reduced toxicity, and, in particular, more efficient toxin clearance rates and a lower cost of production. Given this, this review has carefully summarized newly developed engineered different structural biomedical materials for blood purification in terms of types and structure characteristics of blood purification materials, the production process, as well as interfacial chemical adsorption properties or mechanisms. This study may provide a valuable reference for fabricating a user-friendly purification device that is more suitable for clinical blood purification applications in dialysis patients.
Collapse
Affiliation(s)
- Cui Gao
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China
| | - Qian Zhang
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China
| | - Yi Yang
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China.
- Department of Nephology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China.
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China.
| | - Yangyang Li
- Key Laboratory of Women's Reproductive Health Research of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| | - Weiqiang Lin
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China.
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China.
| |
Collapse
|
38
|
Su CT, See DHW, Huang JW. Lipid-Based Nanocarriers in Renal RNA Therapy. Biomedicines 2022; 10:283. [PMID: 35203492 PMCID: PMC8869454 DOI: 10.3390/biomedicines10020283] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 01/27/2023] Open
Abstract
Kidney disease is a multifactorial problem, with a growing prevalence and an increasing global burden. With the latest worldwide data suggesting that chronic kidney disease (CKD) is the 12th leading cause of death, it is no surprise that CKD remains a public health problem that requires urgent attention. Multiple factors contribute to kidney disease, each with its own pathophysiology and pathogenesis. Furthermore, microRNAs (miRNAs) have been linked to several types of kidney diseases. As dysregulation of miRNAs is often seen in some diseases, there is potential in the exploitation of this for therapeutic applications. In addition, uptake of interference RNA has been shown to be rapid in kidneys making them a good candidate for RNA therapy. The latest advancements in RNA therapy and lipid-based nanocarriers have enhanced the effectiveness and efficiency of RNA-related drugs, thereby making RNA therapy a viable treatment option for renal disease. This is especially useful for renal diseases, for which a suitable treatment is not yet available. Moreover, the high adaptability of RNA therapy combined with the low risk of lipid-based nanocarriers make for an attractive treatment choice. Currently, there are only a small number of RNA-based drugs related to renal parenchymal disease, most of which are in different stages of clinical trials. We propose the use of miRNAs or short interfering RNAs coupled with a lipid-based nanocarrier as a delivery vehicle for managing renal disease.
Collapse
Affiliation(s)
- Chi-Ting Su
- Department of Medicine, National Taiwan University Cancer Centre, Taipei 10672, Taiwan; (C.-T.S.); (D.H.W.S.)
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Douliu 640, Taiwan
| | - Daniel H. W. See
- Department of Medicine, National Taiwan University Cancer Centre, Taipei 10672, Taiwan; (C.-T.S.); (D.H.W.S.)
| | - Jenq-Wen Huang
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Douliu 640, Taiwan
| |
Collapse
|
39
|
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: 16] [Impact Index Per Article: 5.3] [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
|
40
|
Paluszkiewicz P, Martuszewski A, Zaręba N, Wala K, Banasik M, Kepinska M. The Application of Nanoparticles in Diagnosis and Treatment of Kidney Diseases. Int J Mol Sci 2021; 23:ijms23010131. [PMID: 35008556 PMCID: PMC8745391 DOI: 10.3390/ijms23010131] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/16/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Nanomedicine is currently showing great promise for new methods of diagnosing and treating many diseases, particularly in kidney disease and transplantation. The unique properties of nanoparticles arise from the diversity of size effects, used to design targeted nanoparticles for specific cells or tissues, taking renal clearance and tubular secretion mechanisms into account. The design of surface particles on nanoparticles offers a wide range of possibilities, among which antibodies play an important role. Nanoparticles find applications in encapsulated drug delivery systems containing immunosuppressants and other drugs, in imaging, gene therapies and many other branches of medicine. They have the potential to revolutionize kidney transplantation by reducing and preventing ischemia-reperfusion injury, more efficiently delivering drugs to the graft site while avoiding systemic effects, accurately localizing and visualising the diseased site and enabling continuous monitoring of graft function. So far, there are known nanoparticles with no toxic effects on human tissue, although further studies are still needed to confirm their safety.
Collapse
Affiliation(s)
- Patrycja Paluszkiewicz
- Department of Emergency Medical Service, Wroclaw Medical University, Bartla 5, 50-367 Wroclaw, Poland;
| | - Adrian Martuszewski
- Department of Population Health, Division of Environmental Health and Occupational Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368 Wroclaw, Poland;
| | - Natalia Zaręba
- Department of Pharmaceutical Biochemistry, Division of Biomedical and Environmental Analysis, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wrocław, Poland;
| | - Kamila Wala
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland;
| | - Mirosław Banasik
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
- Correspondence: (M.B.); (M.K.); Tel.: +48-71-733-2500 (M.B.); +48-71-784-0171 (M.K.)
| | - Marta Kepinska
- Department of Pharmaceutical Biochemistry, Division of Biomedical and Environmental Analysis, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wrocław, Poland;
- Correspondence: (M.B.); (M.K.); Tel.: +48-71-733-2500 (M.B.); +48-71-784-0171 (M.K.)
| |
Collapse
|
41
|
Wu Q, Wang J, Wang Y, Xiang L, Tan Y, Feng J, Zhang Z, Zhang L. Targeted delivery of celastrol to glomerular endothelium and podocytes for chronic kidney disease treatment. NANO RESEARCH 2021; 15:3556-3568. [PMID: 34925707 PMCID: PMC8666268 DOI: 10.1007/s12274-021-3894-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/07/2021] [Accepted: 09/17/2021] [Indexed: 06/14/2023]
Abstract
UNLABELLED The etiology of chronic kidney disease (CKD) is complex and diverse, which could be briefly categorized to glomerular- or tubular-originated. However, the final outcomes of CKD are mainly glomerular sclerosis, endothelial dysfunction and injury, and chronic inflammation. Thus, targeted delivery of drugs to the glomeruli in order to ameliorate glomerular endothelial damage may help alleviate CKD and help enrich our knowledge. The herb tripterygium wilfordii shows therapeutic effect on kidney disease, and celastrol (CLT) is one of its active ingredients but with strong toxicity. Therefore, based on the unique structure and pathological characteristics of the glomerulus, we designed a targeted delivery system named peptides coupled CLT-phospholipid lipid nanoparticles (PC-PLNs) to efficiently deliver CLT to damaged endothelial cells and podocytes in the glomerulus for CKD treatment and research. PC-PLNs could effectively inhibit inflammation, reduce endothelial damage, alleviate CKD severity, and reduce the toxicity of CLT. We also studied the mechanism of CLT in the treatment of nephropathy and found that CLT can increase the level of NO by increasing eNOS while inhibiting the expression of VCAM-1, thus provides an anti-inflammatory effect. Therefore, our study not only offered an efficient CKD drug formulation for further development, but also provided new medical knowledge about CKD. ELECTRONIC SUPPLEMENTARY MATERIAL Supplementary material (attached with all the supporting tables and figures mentioned in this work) is available in the online version of this article at 10.1007/s12274-021-3894-x.
Collapse
Affiliation(s)
- Qingsi Wu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 China
- Med-X Center for Materials, Sichuan University, No. 14 Section 3 South Renmin Road, Jinjiang District, Chengdu, 610000 China
| | - Jiading Wang
- College of Polymer Science and Engineering, Sichuan University, No. 24, South Block 1, First Ring Road, Chengdu, 610065 China
- Med-X Center for Materials, Sichuan University, No. 14 Section 3 South Renmin Road, Jinjiang District, Chengdu, 610000 China
| | - Yuanfang Wang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 China
- Med-X Center for Materials, Sichuan University, No. 14 Section 3 South Renmin Road, Jinjiang District, Chengdu, 610000 China
| | - Ling Xiang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 China
- Med-X Center for Materials, Sichuan University, No. 14 Section 3 South Renmin Road, Jinjiang District, Chengdu, 610000 China
| | - Yulu Tan
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 China
- Med-X Center for Materials, Sichuan University, No. 14 Section 3 South Renmin Road, Jinjiang District, Chengdu, 610000 China
| | - Jiaxing Feng
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 China
- Med-X Center for Materials, Sichuan University, No. 14 Section 3 South Renmin Road, Jinjiang District, Chengdu, 610000 China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 China
- Med-X Center for Materials, Sichuan University, No. 14 Section 3 South Renmin Road, Jinjiang District, Chengdu, 610000 China
| | - Ling Zhang
- College of Polymer Science and Engineering, Sichuan University, No. 24, South Block 1, First Ring Road, Chengdu, 610065 China
- Med-X Center for Materials, Sichuan University, No. 14 Section 3 South Renmin Road, Jinjiang District, Chengdu, 610000 China
| |
Collapse
|
42
|
Tan J, Yin K, Ouyang Z, Wang R, Pan H, Wang Z, Zhao C, Guo W, Gu X. Real-Time Monitoring Renal Impairment Due to Drug-Induced AKI and Diabetes-Caused CKD Using an NAG-Activatable NIR-II Nanoprobe. Anal Chem 2021; 93:16158-16165. [PMID: 34813273 DOI: 10.1021/acs.analchem.1c03926] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Real-time in vivo optical imaging of kidney function is important for the diagnosis of renal diseases, such as acute kidney injury (AKI) and chronic kidney disease (CKD), with high morbidity and mortality worldwide. However, the reported optical imaging agents still have limitations for identifying AKI or CKD in the early stage due to their low sensitivity, poor tissue penetration, and significant background interference. Herein, an N-acetyl-β-d-glucosaminidase (NAG)-activatable second near-infrared (NIR-II) fluorescent nanoprobe (BOD-II-NAG-NP) is developed for monitoring the progression of drug-induced AKI and in vivo imaging of diabetes-caused CKD. NAG, as a biomarker of renal diseases, is able to specifically activate BOD-II-NAG-NP to release NIR-II fluorescence signals, enabling in vivo imaging of kidney dysfunctions in living mice. Importantly, such an active imaging mechanism allows BOD-II-NAG-NP to noninvasively detect the onset of drug-induced AKI at least 32 h earlier than the most existing assays, which indicates that BOD-II-NAG-NP has the potential to be an optical imaging agent for the early diagnosis of AKI. Moreover, NIR-II fluorescence produced by BOD-II-NAG-NP could deeply penetrate into the relatively thick layers of fat in diabetic nephropathy mice and provide in vivo imaging with high resolution, indicating that BOD-II-NAG-NP has clinical potential for precision diagnosis of CKD.
Collapse
Affiliation(s)
- Jiahui Tan
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Kai Yin
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Zhirong Ouyang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Rongchen Wang
- Key Laboratory for Advanced Materials and Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Hongming Pan
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Zhijun Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Chuchang Zhao
- Key Laboratory for Advanced Materials and Feringa Nobel Prize Scientist Joint Research Center, Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Wei Guo
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Xianfeng Gu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| |
Collapse
|
43
|
Zhong Y, Wang K, Zhang Y, Yin Q, Li S, Wang J, Zhang X, Han H, Yao K. Ocular Wnt/β-Catenin Pathway Inhibitor XAV939-Loaded Liposomes for Treating Alkali-Burned Corneal Wound and Neovascularization. Front Bioeng Biotechnol 2021; 9:753879. [PMID: 34765592 PMCID: PMC8576519 DOI: 10.3389/fbioe.2021.753879] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022] Open
Abstract
Corneal wound involves a series of complex and coordinated physiological processes, leading to persistent epithelial defects and opacification. An obstacle in the treatment of ocular diseases is poor drug delivery and maintenance. In this study, we constructed a Wnt/β-catenin pathway inhibitor, XAV939-loaded liposome (XAV939 NPs), and revealed its anti-inflammatory and antiangiogenic effects. The XAV939 NPs possessed excellent biocompatibility in corneal epithelial cells and mouse corneas. In vitro corneal wound healing assays demonstrated their antiangiogenic effect, and LPS-induced expressions of pro-inflammatory genes of IL-1β, IL-6, and IL-17α were significantly suppressed by XAV939 NPs. In addition, the XAV939 NPs significantly ameliorated alkali-burned corneas with slight corneal opacity, reduced neovascularization, and faster recovery, which were attributed to the decreased gene expressions of angiogenic and inflammatory cytokines. The findings supported the potential of XAV939 NPs in ameliorating corneal wound and suppressing neovascularization, providing evidence for their clinical application in ocular vascular diseases.
Collapse
Affiliation(s)
- Yueyang Zhong
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kai Wang
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yin Zhang
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qichuan Yin
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Su Li
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiaming Wang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, State Key Laboratory of Experimental Hematology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaobo Zhang
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haijie Han
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ke Yao
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
44
|
Núñez-Leyva JM, Kolosovas-Machuca ES, Sánchez J, Guevara E, Cuadrado A, Alda J, González FJ. Computational and Experimental Analysis of Gold Nanorods in Terms of Their Morphology: Spectral Absorption and Local Field Enhancement. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:1696. [PMID: 34203448 PMCID: PMC8308185 DOI: 10.3390/nano11071696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 01/04/2023]
Abstract
A nanoparticle's shape and size determine its optical properties. Nanorods are nanoparticles that have double absorption bands associated to surface plasmon oscillations along their two main axes. In this work, we analize the optical response of gold nanorods with numerical simulations and spectral absorption measurements to evaluate their local field enhancement-which is key for surface-enhanced Raman spectroscopic (SERS) applications. Our experimental results are in good agreement with finite element method (FEM) simulations for the spectral optical absorption of the nanoparticles. We also observed a strong dependence of the optical properties of gold nanorods on their geometrical dimension and shape. Our numerical simulations helped us reveal the importance of the nanorods' morphology generated during the synthesis stage in the evaluation of absorption and local field enhancement. The application of these gold nanorods in surface-enhancement Raman spectroscopy is analyzed numerically, and results in a 5.8×104 amplification factor when comparing the values obtained for the nanorod deposited on a dielectric substrate compared to the nanorod immersed in water.
Collapse
Affiliation(s)
- Juan Manuel Núñez-Leyva
- Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología, Universidad Autónoma de San Luis Potosí, 550 Sierra Leona Ave, San Luis Potosí 78210, Mexico; (J.M.N.-L.); (E.S.K.-M.); (J.S.); (E.G.); (F.J.G.)
- Doctorado Institucional en Ingeniería y Ciencia de Materiales (DICIM-UASLP), Universidad Autónoma de San Luis Potosí, 550 Sierra Leona Ave, San Luis Potosí 78210, Mexico
| | - Eleazar Samuel Kolosovas-Machuca
- Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología, Universidad Autónoma de San Luis Potosí, 550 Sierra Leona Ave, San Luis Potosí 78210, Mexico; (J.M.N.-L.); (E.S.K.-M.); (J.S.); (E.G.); (F.J.G.)
| | - John Sánchez
- Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología, Universidad Autónoma de San Luis Potosí, 550 Sierra Leona Ave, San Luis Potosí 78210, Mexico; (J.M.N.-L.); (E.S.K.-M.); (J.S.); (E.G.); (F.J.G.)
| | - Edgar Guevara
- Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología, Universidad Autónoma de San Luis Potosí, 550 Sierra Leona Ave, San Luis Potosí 78210, Mexico; (J.M.N.-L.); (E.S.K.-M.); (J.S.); (E.G.); (F.J.G.)
| | - Alexander Cuadrado
- Escuela Superior de Ciencias Experimentales y Tecnología, Universidad Rey Juan Carlos, C/ Tulipán s/n, Móstoles, 28933 Madrid, Spain;
| | - Javier Alda
- Applied Optics Complutense Group, Faculty of Optics and Optometry, University Complutense of Madrid, 118 Arcos de Jalón Ave, 28037 Madrid, Spain
| | - Francisco Javier González
- Coordinación para la Innovación y Aplicación de la Ciencia y la Tecnología, Universidad Autónoma de San Luis Potosí, 550 Sierra Leona Ave, San Luis Potosí 78210, Mexico; (J.M.N.-L.); (E.S.K.-M.); (J.S.); (E.G.); (F.J.G.)
| |
Collapse
|
45
|
Eftekhari A, Maleki Dizaj S, Ahmadian E, Przekora A, Hosseiniyan Khatibi SM, Ardalan M, Zununi Vahed S, Valiyeva M, Mehraliyeva S, Khalilov R, Hasanzadeh M. Application of Advanced Nanomaterials for Kidney Failure Treatment and Regeneration. MATERIALS 2021; 14:ma14112939. [PMID: 34072461 PMCID: PMC8198057 DOI: 10.3390/ma14112939] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/15/2021] [Accepted: 05/27/2021] [Indexed: 11/18/2022]
Abstract
The implementation of nanomedicine not only provides enhanced drug solubility and reduced off-target adverse effects, but also offers novel theranostic approaches in clinical practice. The increasing number of studies on the application of nanomaterials in kidney therapies has provided hope in a more efficient strategy for the treatment of renal diseases. The combination of biotechnology, material science and nanotechnology has rapidly gained momentum in the realm of therapeutic medicine. The establishment of the bedrock of this emerging field has been initiated and an exponential progress is observed which might significantly improve the quality of human life. In this context, several approaches based on nanomaterials have been applied in the treatment and regeneration of renal tissue. The presented review article in detail describes novel strategies for renal failure treatment with the use of various nanomaterials (including carbon nanotubes, nanofibrous membranes), mesenchymal stem cells-derived nanovesicles, and nanomaterial-based adsorbents and membranes that are used in wearable blood purification systems and synthetic kidneys.
Collapse
Affiliation(s)
- Aziz Eftekhari
- Pharmacology and Toxicology Department, Maragheh University of Medical Sciences, Maragheh 7815155158, Iran;
- Russian Institute for Advanced Study, Moscow State Pedagogical University, 1/1, Malaya Pirogovskaya St., 119991 Moscow, Russia;
| | - Solmaz Maleki Dizaj
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz 5166614756, Iran;
| | - Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz 5166614756, Iran; (S.M.H.K.); (S.Z.V.)
- Correspondence: (E.A.); (A.P.); (M.A.); (M.H.); Tel.: +48-81-448-7026 (A.P.)
| | - Agata Przekora
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland
- Correspondence: (E.A.); (A.P.); (M.A.); (M.H.); Tel.: +48-81-448-7026 (A.P.)
| | | | - Mohammadreza Ardalan
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz 5166614756, Iran; (S.M.H.K.); (S.Z.V.)
- Correspondence: (E.A.); (A.P.); (M.A.); (M.H.); Tel.: +48-81-448-7026 (A.P.)
| | - Sepideh Zununi Vahed
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz 5166614756, Iran; (S.M.H.K.); (S.Z.V.)
| | - Mahbuba Valiyeva
- Department of Pharmaceutical Technology and Management, Azerbaijan Medical University, AZ 1022 Baku, Azerbaijan; (M.V.); (S.M.)
| | - Sevil Mehraliyeva
- Department of Pharmaceutical Technology and Management, Azerbaijan Medical University, AZ 1022 Baku, Azerbaijan; (M.V.); (S.M.)
| | - Rovshan Khalilov
- Russian Institute for Advanced Study, Moscow State Pedagogical University, 1/1, Malaya Pirogovskaya St., 119991 Moscow, Russia;
- Department of Biophysics and Biochemistry, Baku State University, AZ 1148 Baku, Azerbaijan
- Institute of Radiation Problems, Azerbaijan National Academy of Sciences, AZ 1001 Baku, Azerbaijan
| | - Mohammad Hasanzadeh
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz 5166614756, Iran
- Correspondence: (E.A.); (A.P.); (M.A.); (M.H.); Tel.: +48-81-448-7026 (A.P.)
| |
Collapse
|
46
|
Oligonucleotide-Based Therapies for Renal Diseases. Biomedicines 2021; 9:biomedicines9030303. [PMID: 33809425 PMCID: PMC8001091 DOI: 10.3390/biomedicines9030303] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 02/07/2023] Open
Abstract
The global burden of chronic kidney disease (CKD) is increasing every year and represents a great cost for public healthcare systems, as the majority of these diseases are progressive. Therefore, there is an urgent need to develop new therapies. Oligonucleotide-based drugs are emerging as novel and promising alternatives to traditional drugs. Their expansion corresponds with new knowledge regarding the molecular basis underlying CKD, and they are already showing encouraging preclinical results, with two candidates being evaluated in clinical trials. However, despite recent technological advances, efficient kidney delivery remains challenging, and the presence of off-targets and side-effects precludes development and translation to the clinic. In this review, we provide an overview of the various oligotherapeutic strategies used preclinically, emphasizing the most recent findings in the field, together with the different strategies employed to achieve proper kidney delivery. The use of different nanotechnological platforms, including nanocarriers, nanoparticles, viral vectors or aptamers, and their potential for the development of more specific and effective treatments is also outlined.
Collapse
|