1
|
Jin M, Cho HJ, Na YG, Yun TS, Song B, Lee SR, Je S, Oh HG, Park YG, Rho J, Yang MJ, Shin JS, Baek JS, Lee HK, Kim TW, Cho CW. Practical approach to development of GS-445124-loaded PLGA nanoparticles for the long-term treatment of feline infectious peritonitis caused by feline coronavirus infection. Int J Pharm 2025; 674:125468. [PMID: 40107469 DOI: 10.1016/j.ijpharm.2025.125468] [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: 10/24/2024] [Revised: 02/13/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Feline infectious peritonitis virus (FIPV) caused by feline coronavirus (FCoV) infection leads to a high mortality rate when untreated. GS-441524, an antiviral agent effective against FIPV, is orally administered twice daily or through daily subcutaneous injections for approximately 12 weeks. While the short treatment period recuses concerns about adherence, frequent administrations may cause handling-related stress in cats. Therefore, it is essential to develop a long-acting formulation that requires only a single administration. In this study, we used polylactide-co-glycolide (PLGA) as a carrier, which was used to effectively encapsulate GS-441524 with sustained-release functionality and GS-441524-loaded PLGA nanoparticles (GS-PLGA NP) were prepared. The particle size of GS-PLGA NP was 216 nm, the encapsulation efficiency was 78 %, and the 7-day release was 92 %. When GS-PLGA NP was injected at 22 mg/kg in cats, higher systemic exposure can be expected compared to injecting GS-441524 at 4 mg/kg for one week (relative bioavailability, 152 %). As well as GS-PLGA NP showed lower toxicity, improved cellular uptake, and enhanced antiviral efficacy against FCoV compared to the pure GS-441524.
Collapse
Affiliation(s)
- Minki Jin
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; Center for Large Animals Convergence Research, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea
| | - Hyun-Jin Cho
- Center for Large Animals Convergence Research, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea; College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Young-Guk Na
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Taek-Seon Yun
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Bomin Song
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Sang-Rae Lee
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Seonho Je
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Hong-Geun Oh
- R&D Division, Huvet Co., Ltd, Iksan-si 54541, Republic of Korea
| | - Yang-Gyu Park
- R&D Division, Huvet Co., Ltd, Iksan-si 54541, Republic of Korea
| | - Jinhyung Rho
- Center for Translational Toxicologic Research, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea
| | - Mi-Jin Yang
- Center for Translational Toxicologic Research, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea
| | - Jin Soo Shin
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Jong-Suep Baek
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hong-Ki Lee
- College of Veterinary Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-Gu, Cheongju, Chungbuk 28644, Republic of Korea.
| | - Tae-Won Kim
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea.
| | - Cheong-Weon Cho
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea.
| |
Collapse
|
2
|
Fan X, Cheng D, Niu B, Wang X, Zhang P. Current research status, applications and challenges of ketorolac-based sustained-release and controlled-release formulations. Int J Pharm 2025; 670:125162. [PMID: 39793634 DOI: 10.1016/j.ijpharm.2024.125162] [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: 10/02/2024] [Revised: 12/11/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025]
Abstract
Ketorolac, a nonsteroidal anti-inflammatory drug, exhibits moderate antipyretic and anti-inflammatory properties, as well as potent analgesic effects. It is widely used in clinical practice for pain relief in cases of mild and severe pain such as postoperative pain, fractures, sprains, toothaches and cancer pain. Due to its relatively short half-life, patients experiencing pain often need frequent injections or oral medications, leading to poor patient compliance. Thus, it is crucial to create long-acting sustained-release formulations of ketorolac. This paper provides an overview of the research, applications, and challenges associated with ketorolac sustained-release formulations over the past decade, based on a comprehensive review of the literature. The aim is to provide fresh insights for the research and development of long-acting, sustained-release, and controlled-release formulations of ketorolac.
Collapse
Affiliation(s)
- Xiaoling Fan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China; Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, Shandong, PR China
| | - Dongfang Cheng
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, Shandong, PR China.
| | - Baohua Niu
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, Shandong, PR China
| | - Xiuzhi Wang
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, PR China
| | - Peng Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China.
| |
Collapse
|
3
|
Perra M, Castangia I, Aroffu M, Fulgheri F, Abi-Rached R, Manca ML, Cortés H, Del Prado-Audelo ML, Nomura-Contreras C, Romero-Montero A, Büsselberg D, Leyva-Gómez G, Sharifi-Rad J, Calina D. Maytansinoids in cancer therapy: advancements in antibody-drug conjugates and nanotechnology-enhanced drug delivery systems. Discov Oncol 2025; 16:73. [PMID: 39838217 PMCID: PMC11751265 DOI: 10.1007/s12672-025-01820-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/15/2025] [Indexed: 01/23/2025] Open
Abstract
Cancer remains the second leading cause of death globally, driving the need for innovative therapies. Among natural compounds, maytansinoids have shown significant promise, contributing to nearly 25% of recently approved anticancer drugs. Despite their potential, early clinical trials faced challenges due to severe side effects, prompting advancements in delivery systems such as antibody-maytansinoid conjugates (AMCs). This review highlights the anticancer activity of maytansinoids, with a focus on AMCs designed to target cancer cells specifically. Preclinical and clinical studies show that AMCs, including FDA-approved drugs like Kadcyla and Elahere, effectively inhibit tumor growth while reducing systemic toxicity. Key developments include improved synthesis methods, linker chemistry and payload design. Ongoing research aims to enhance the safety and efficacy of AMCs, integrate nanotechnology for drug delivery, and identify novel therapeutic targets. These advancements hold potential to transform maytansinoid-based cancer treatments in the future.
Collapse
Affiliation(s)
- Matteo Perra
- DISVA-Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. DeMonserrato-Sestu Km 0.700, 09042 CA, Monserrato, Italy
| | - Ines Castangia
- DISVA-Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. DeMonserrato-Sestu Km 0.700, 09042 CA, Monserrato, Italy
| | - Matteo Aroffu
- DISVA-Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. DeMonserrato-Sestu Km 0.700, 09042 CA, Monserrato, Italy
| | - Federica Fulgheri
- DISVA-Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. DeMonserrato-Sestu Km 0.700, 09042 CA, Monserrato, Italy
| | - Rita Abi-Rached
- DISVA-Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. DeMonserrato-Sestu Km 0.700, 09042 CA, Monserrato, Italy
| | - Maria Letizia Manca
- DISVA-Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. DeMonserrato-Sestu Km 0.700, 09042 CA, Monserrato, Italy.
| | - Hernán Cortés
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de Mexico, Mexico
| | | | | | - Alejandra Romero-Montero
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico
| | - Dietrich Büsselberg
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico.
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, 092301, Ecuador.
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| |
Collapse
|
4
|
Fernandes Q, Therachiyil L, Khan AQ, Bedhiafi T, Korashy HM, Bhat AA, Uddin S. Shrinking the battlefield in cancer therapy: Nanotechnology against cancer stem cells. Eur J Pharm Sci 2023; 191:106586. [PMID: 37729956 DOI: 10.1016/j.ejps.2023.106586] [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: 06/24/2023] [Revised: 09/07/2023] [Accepted: 09/18/2023] [Indexed: 09/22/2023]
Abstract
Cancer remains one of the leading causes of mortality worldwide, presenting a significant healthcare challenge owing to the limited efficacy of current treatments. The application of nanotechnology in cancer treatment leverages the unique optical, magnetic, and electrical attributes of nanomaterials to engineer innovative, targeted therapies. Specifically, manipulating nanomaterials allows for enhanced drug loading efficiency, improved bioavailability, and targeted delivery systems, reducing the non-specific cytotoxic effects characteristic of conventional chemotherapies. Furthermore, recent advances in nanotechnology have demonstrated encouraging results in specifically targeting CSCs, a key development considering the role of these cells in disease recurrence and resistance to treatment. Despite these breakthroughs, the clinical approval rates of nano-drugs have not kept pace with research advances, pointing to existing obstacles that must be addressed. In conclusion, nanotechnology presents a novel, powerful tool in the fight against cancer, particularly in targeting the elusive and treatment-resistant CSCs. This comprehensive review delves into the intricacies of nanotherapy, explicitly targeting cancer stem cells, their markers, and associated signaling pathways.
Collapse
Affiliation(s)
- Queenie Fernandes
- College of Medicine, Qatar University, Doha, Qatar; Translational Cancer Research Facility, Hamad Medical Corporation, National Center for Cancer Care and Research, PO. Box 3050, Doha, Qatar
| | - Lubna Therachiyil
- Academic Health System, Hamad Medical Corporation, Translational Research Institute, Doha 3050, Qatar; Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | - Abdul Q Khan
- Academic Health System, Hamad Medical Corporation, Translational Research Institute, Doha 3050, Qatar
| | - Takwa Bedhiafi
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | - Hesham M Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Shahab Uddin
- College of Medicine, Qatar University, Doha, Qatar; Academic Health System, Hamad Medical Corporation, Dermatology Institute, Doha 3050, Qatar; Laboratory of Animal Research Center, Qatar University, Doha 2713, Qatar; Department of Biosciences, Integral University, Lucknow, Uttar Pradesh 22602, India.
| |
Collapse
|
5
|
Wang Y, Li Z, Bao Y, Cui H, Li J, Song B, Wang M, Li H, Cui X, Chen Y, Chen W, Yang S, Yang Y, Jin Z, Si X, Li B. Colon-targeted delivery of polyphenols: construction principles, targeting mechanisms and evaluation methods. Crit Rev Food Sci Nutr 2023; 65:64-86. [PMID: 37823723 DOI: 10.1080/10408398.2023.2266842] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Polyphenols have received considerable attention for their promotive effects on colonic health. However, polyphenols are mostly sensitive to harsh gastrointestinal environments, thus, must be protected. It is necessary to design and develop a colon-targeted delivery system to improve the stability, colon-targeting and bioavailability of polyphenols. This paper mainly introduces research on colon-targeted controlled release of polyphenols. The physiological features affecting the dissolution, release and absorption of polyphenol-loaded delivery systems in the colon are first discussed. Simultaneously, the types of colon-targeted carriers with different release mechanisms are described, and colon-targeting assessment models that have been studied so far and their advantages and limitations are summarized. Based on the current research on polyphenols colon-targeting, outlook and reflections are proposed, with the goal of inspiring strategic development of new colon-targeted therapeutics to ensure that the polyphenols reach the colon with complete bioactivity.
Collapse
Affiliation(s)
- Yidi Wang
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Zhiying Li
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Yiwen Bao
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Huijun Cui
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Jiaxin Li
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Baoge Song
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Mengzhu Wang
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Haikun Li
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Xingyue Cui
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Yi Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
| | - Wei Chen
- Faculty of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | - Shufang Yang
- Zhejiang Lanmei Technology Co., Ltd, Zhu-ji City, Zhejiang Province, China
| | - Yiyun Yang
- Zhejiang Lanmei Technology Co., Ltd, Zhu-ji City, Zhejiang Province, China
| | - Zhufeng Jin
- Zhejiang Lanmei Technology Co., Ltd, Zhu-ji City, Zhejiang Province, China
| | - Xu Si
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Bin Li
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| |
Collapse
|
6
|
Ren Y, Nie L, Luo C, Zhu S, Zhang X. Advancement in Therapeutic Intervention of Prebiotic-Based Nanoparticles for Colonic Diseases. Int J Nanomedicine 2022; 17:6639-6654. [PMID: 36582460 PMCID: PMC9793785 DOI: 10.2147/ijn.s390102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Intestinal flora has become a therapeutic target for the intervention of colonic diseases (CDs) with better understanding of the interplay between microbiota and CDs. Depending on unique properties and prominent ability of regulating the intestinal flora, prebiotics can not only achieve a colon-specific drug delivery but also maintain the intestinal homeostasis, thus playing a positive role in the intervention of CDs. Currently, different studies on prebiotic-based nanoparticles have been contrived for colonic drug delivery and have shown great potential in curing various CDs, such as colitis and colorectal cancer. Nevertheless, there is a lack of systematic survey on the use of prebiotic nanoparticles for the treatment of CDs. This review aims to generalize the state-of-the-art of prebiotic nanomedicines specific for CDs. The species and function of intestinal flora and various kinds of prebiotics available as well as their regulating effects on intestinal flora were expounded. A variety of prebiotic nanoparticles pertinent to colon-targeted drug delivery systems were illustrated with particular emphasis on their curative activities on CDs. The efficacy and safety of prebiotic-based colonic drug delivery systems (p-CDDs) were also analyzed. In conclusion, the synergy between prebiotic nanoparticles and their cargos may hold promise for the treatment and intervention of CDs.
Collapse
Affiliation(s)
- Yuehong Ren
- Department of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, People’s Republic of China
| | - Linghui Nie
- ASD Medical Rehabilitation Center, the Second People’s Hospital of Guangdong Province, Guangzhou, People’s Republic of China
| | - Chunhua Luo
- Newborn Intensive Care Unit, Guangzhou Women and Children’s Medical Center, Guangzhou, People’s Republic of China
| | - Shiping Zhu
- Department of Chinese Traditional Medicine, the First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China,Shiping Zhu, Department of Chinese Traditional Medicine, the First Affiliated Hospital of Jinan University, 613 West Huangpu Avenue, Guangzhou, 513630, People’s Republic of China, Email
| | - Xingwang Zhang
- Department of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, People’s Republic of China,Correspondence: Xingwang Zhang, Department of Pharmaceutics, College of Pharmacy, Jinan University, No. 855 East Xingye Avenue, Guangzhou, 511443, People’s Republic of China, Email
| |
Collapse
|
7
|
Stalder T, Zaiter T, El-Basset W, Cornu R, Martin H, Diab-Assaf M, Béduneau A. Interaction and toxicity of ingested nanoparticles on the intestinal barrier. Toxicology 2022; 481:153353. [DOI: 10.1016/j.tox.2022.153353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/05/2022] [Accepted: 10/13/2022] [Indexed: 11/28/2022]
|
8
|
Li Y, Zhang W, Zhao R, Zhang X. Advances in oral peptide drug nanoparticles for diabetes mellitus treatment. Bioact Mater 2022; 15:392-408. [PMID: 35386357 PMCID: PMC8958389 DOI: 10.1016/j.bioactmat.2022.02.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 02/18/2022] [Accepted: 02/18/2022] [Indexed: 12/11/2022] Open
Abstract
Peptide drugs play an important role in diabetes mellitus treatment. Oral administration of peptide drugs is a promising strategy for diabetes mellitus because of its convenience and high patient compliance compared to parenteral administration routes. However, there are a series of formidable unfavorable conditions present in the gastrointestinal (GI) tract after oral administration, which result in the low oral bioavailability of these peptide drugs. To overcome these challenges, various nanoparticles (NPs) have been developed to improve the oral absorption of peptide drugs due to their unique in vivo properties and high design flexibility. This review discusses the unfavorable conditions present in the GI tract and provides the corresponding strategies to overcome these challenges. The review provides a comprehensive overview on the NPs that have been constructed for oral peptide drug delivery in diabetes mellitus treatment. Finally, we will discuss the rational application and give some suggestions that can be utilized for the development of oral peptide drug NPs. Our aim is to provide a systemic and comprehensive review of oral peptide drug NPs that can overcome the challenges in GI tract for efficient treatment of diabetes mellitus. •Oral administration of peptide drugs is a promising strategy for diabetes mellitus treatment •A series of formidable unfavorable conditions in gastrointestinal tract result in the low oral bioavailability of peptide drugs •Nanoparticles can improve the oral bioavailability of peptide drugs
Collapse
Affiliation(s)
- Yan Li
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, PR China
| | - Wen Zhang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, PR China
| | - Ruichen Zhao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, PR China.,School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Xin Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, PR China
| |
Collapse
|
9
|
Curcumin-Induced Stabilization of Protein-Based Nano-Delivery Vehicles Reduces Disruption of Zwitterionic Giant Unilamellar Vesicles. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27061941. [PMID: 35335305 PMCID: PMC8956123 DOI: 10.3390/molecules27061941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/15/2022] [Accepted: 03/15/2022] [Indexed: 11/17/2022]
Abstract
Curcumin-loaded native and succinylated pea protein nanoparticles, as well as zwitterionic giant unilamellar vesicles were used in this study as model bioactive compound loaded-nanoparticles and biomembranes, respectively, to assess bio-nano interactions. Curcumin-loaded native protein-chitosan and succinylated protein-chitosan complexes, as well as native protein-chitosan and succinylated protein-chitosan hollow, induced leakage of the calcein encapsulated in the giant unilamellar vesicles. The leakage was more pronounced with hollow protein-chitosan complexes. However, curcumin-loaded native protein and curcumin-loaded succinylated protein nanoparticles induced calcein fluorescence quenching. Dynamic light scattering measurements showed that the interaction of curcumin-loaded native protein, curcumin-loaded succinylated protein, native protein-chitosan, and succinylated protein-chitosan complexes with the giant unilamellar vesicles caused a major reduction in the size of the lipid vesicles. Confocal and widefield fluorescence microscopy showed rupturing of the unilamellar vesicles after treatment with native pea protein-chitosan and succinylated pea protein-chitosan complexes. The nature of interaction between the curcumin-loaded protein nanoparticles and the biomembranes, at the bio-nano interface, is influenced by the encapsulated curcumin. Findings from this study showed that, as the protein plays a crucial role in stabilizing the bioactive compound from chemical and photodegradation, the encapsulated nutraceutical stabilizes the protein nanoparticle to reduce its interaction with biomembranes.
Collapse
|
10
|
Mirza-Aghazadeh-Attari M, Mihanfar A, Yousefi B, Majidinia M. Nanotechnology-based advances in the efficient delivery of melatonin. Cancer Cell Int 2022; 22:43. [PMID: 35093076 PMCID: PMC8800219 DOI: 10.1186/s12935-022-02472-7] [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: 11/02/2021] [Accepted: 01/16/2022] [Indexed: 01/09/2023] Open
Abstract
N-[2-(5-methoxy-1H-indol-3-yl) ethyl] or simply melatonin is a biogenic amine produced by pineal gland and recently recognized various other organs. Because of a broad range of biological function melatonin is considered as a therapeutic agent with high efficacy in the treatment of multiple disorders, such as cancer, degenerative disorders and immune disease. However, since melatonin can affect receptors on the cellular membrane, in the nucleus and can act as an anti-oxidant molecule, some unwanted effects may be observed after administration. Therefore, the entrapment of melatonin in biocompatible, biodegradable and safe nano-delivery systems can prevent its degradation in circulation; decrease its toxicity with increased half-life, enhanced pharmacokinetic profile leading to improved patient compliance. Because of this, nanoparticles have been used to deliver melatonin in multiple studies, and the present article aims to cumulatively illustrate their findings.
Collapse
Affiliation(s)
| | - Ainaz Mihanfar
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Bahman Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Orjhans Street, Resalat Blvd, Urmia, Iran.
| |
Collapse
|
11
|
Yue P, Chen B, Lv X, Zou Y, Cao H, Ma Y, Wang L, Liu Z, Zheng Y, Duan B, Wu S, Ye Q. Biocompatible Composite Microspheres of Chitin/Ordered Mesoporous Carbon CMK3 for Bilirubin Adsorption and Cell Microcarrier Culture. Macromol Biosci 2022; 22:e2100412. [PMID: 35007390 DOI: 10.1002/mabi.202100412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 12/12/2021] [Indexed: 11/12/2022]
Abstract
Extra bilirubin in the blood can provoke serious illness in patients with severe liver disease. Hemoperfusion is an effective method to remove the extra bilirubin, but its application is limited by the low adsorption efficiency and poor biocompatibility of available adsorbent materials. In this study, chitin/ordered mesoporous carbon CMK3 (Ch/CMK3) microspheres were successfully prepared. Results of characterization experiments indicated that these composite microspheres possess a multilayered porous nanofibrous structure with an extremely large specific surface area (300.19 m2 g-1 ) and large pore size. Notably, the Ch/CMK3 microspheres demonstrated a high bilirubin adsorption capacity (228.19 mg g-1 ) in phosphate buffer solution, and an outstanding bilirubin removal ratio (76.78%±4.40%) in the plasma of rabbits with hyperbilirubinemia without affecting the protein components. More importantly, the Ch/CMK3 microspheres showed no effect on other blood components, no cytotoxicity, and no systemic toxicity to mice. Cell coculture experiments revealed that the microspheres could provide a three-dimensional (3D) space to promote cell adhesion, proliferation, and nutrient exchange. These Ch/CMK3 microspheres featuring a strong ability for bilirubin adsorption and good biocompatibility could be a promising candidate in biomedical applications such as hemoperfusion, cell microcarrier, and 3D tissue engineering. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Pengpeng Yue
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, Wuhan, 430071, China
| | - Biao Chen
- Department of Transplant surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xiaoyan Lv
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yongkang Zou
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, Wuhan, 430071, China
| | - Hankun Cao
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, Wuhan, 430071, China
| | - Yongsheng Ma
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, Wuhan, 430071, China
| | - Lizhe Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, Wuhan, 430071, China
| | - Zhongzhong Liu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, Wuhan, 430071, China
| | - Yiran Zheng
- College of Chemistry and Molecular Sciences, Hubei Engineering Center of Natural Polymer-based Medical Materials, Key Laboratory of Biomedical Polymers of Ministry of Education, Wuhan University, Wuhan, 430072, China
| | - Bo Duan
- College of Chemistry and Molecular Sciences, Hubei Engineering Center of Natural Polymer-based Medical Materials, Key Laboratory of Biomedical Polymers of Ministry of Education, Wuhan University, Wuhan, 430072, China
| | - Shuangquan Wu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, Wuhan, 430071, China
| | - Qifa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, Wuhan, 430071, China.,The Third Xiangya Hospital of Central South University, Research Center of National Health Ministry on Transplantation Medicine Engineering and Technology, Changsha, 410013, China
| |
Collapse
|
12
|
Zong C, Bronckaers A, Vande Velde G, Willems G, Cadenas de Llano‐Pérula M. In Vivo Micro-Computerized Tomography Tracking of Human Periodontal Ligament Stem Cells Labeled with Gold Nanocomplexes. Adv Healthc Mater 2022; 11:e2101133. [PMID: 34704382 DOI: 10.1002/adhm.202101133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 09/29/2021] [Indexed: 12/15/2022]
Abstract
Gold nanocomplexes have been proposed as contrast agents for computerized tomography (CT) and cell tracking, which is especially useful in stem cell therapy. However, their potential for long-term in vivo cell detection is still unknown. This study proposes an optimized approach to labeling human periodontal ligament stem cells (hPDLSCs) with gold nanocomplexes to evaluate their detection with micro-CT after transplantation at four different rat tissues. The gold nanocomplexes of 0.05 mg mL-1 do not affect cell viability nor osteogenic differentiation capacity, but render fluorescent and radiopaque hPDLSCs. Excellent linear correlation with the number of labeled cells is shown over a wide range (r = 0.99, P < 0.01), with a detection limit of ≈1.2 × 103 cells/µL. In vivo, strong, and durable detection of transplanted labeled cells within 5 days at all investigated areas is seen by micro-CT and immunohistochemical assay. This approach confirms the potential of gold nanocomplexes in longitudinal in vivo cell tracking, which may facilitate their application in CT image-guided interventions commonly used in oromaxillofacial or systemic applications of stem cell therapy.
Collapse
Affiliation(s)
- Chen Zong
- Department of Oral Health Sciences‐Orthodontics KU Leuven and Dentistry University Hospitals Leuven Leuven 3000 Belgium
| | - Annelies Bronckaers
- Biomedical Research Institute Faculty of Life Sciences University of Hasselt Diepenbeek 3590 Belgium
| | - Greetje Vande Velde
- Biomedical MRI/Molecular Small Animal Imaging Center (MoSAIC) KU Leuven Leuven 3000 Belgium
| | - Guy Willems
- Department of Oral Health Sciences‐Orthodontics KU Leuven and Dentistry University Hospitals Leuven Leuven 3000 Belgium
| | - Maria Cadenas de Llano‐Pérula
- Department of Oral Health Sciences‐Orthodontics KU Leuven and Dentistry University Hospitals Leuven Leuven 3000 Belgium
| |
Collapse
|
13
|
Cassini C, Zatti PH, Angeli VW, Branco CS, Salvador M. Mutual effects of free and nanoencapsulated phenolic compounds on human microbiota. Curr Med Chem 2021; 29:3160-3178. [PMID: 34720074 DOI: 10.2174/0929867328666211101095131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/08/2021] [Accepted: 08/26/2021] [Indexed: 11/22/2022]
Abstract
Phenolic compounds (PC) have many health benefits such as antioxidant, anticarcinogenic, neuroprotective, and anti-inflammatory activities. All of these activities depend on their chemical structures and their interaction with biological targets in the body. PC occur naturally in polymerized form, linked to glycosides and requires metabolic transformation from their ingestion to their absorption. The gut microbiota can transform PC into more easily absorbed metabolites. The PC, in turn, have prebiotic and antimicrobial actions on the microbiota. Despite this, their low oral bioavailability still compromises biological performance. Therefore, the use of nanocarriers has been demonstrated to be a useful strategy to improve PC absorption and, consequently, their health effects. Nanotechnology is an excellent alternative able to overcome the limits of oral bioavailability of PC, since it offers protection from degradation during their passage through the gastrointestinal tract. Moreover, nanotechnology is also capable of promoting controlled PC release and modulating the interaction between PC and the microbiota. However, little is known about the impact of the nanotechnology on PC effects on the gut microbiota. This review highlights the use of nanotechnology for PC delivery on gut microbiota, focusing on the ability of such formulations to enhance oral bioavailability by applying nanocarriers (polymeric nanoparticles, nanostructured lipid carriers, solid lipid nanoparticles). In addition, the effects of free and nanocarried PC or nanocarriers per se on gut microbiota are also described.
Collapse
Affiliation(s)
- Carina Cassini
- Institute of Biotechnology, University of Caxias do Sul, Caxias do Sul. Brazil
| | | | | | - Catia Santos Branco
- Institute of Biotechnology, University of Caxias do Sul, Caxias do Sul. Brazil
| | - Mirian Salvador
- Institute of Biotechnology, University of Caxias do Sul, Caxias do Sul. Brazil
| |
Collapse
|
14
|
Silver Nanoparticles Agglomerate Intracellularly Depending on the Stabilizing Agent: Implications for Nanomedicine Efficacy. NANOMATERIALS 2020; 10:nano10101953. [PMID: 33007984 PMCID: PMC7601179 DOI: 10.3390/nano10101953] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 09/22/2020] [Accepted: 09/27/2020] [Indexed: 12/12/2022]
Abstract
Engineered nanoparticles are utilized as drug delivery carriers in modern medicine due to their high surface area and tailorable surface functionality. After in vivo administration, nanoparticles distribute and interact with biomolecules, such as polar proteins in serum, lipid membranes in cells, and high ionic conditions during digestion. Electrostatic forces and steric hindrances in a nanoparticle population are disturbed and particles agglomerate in biological fluids. Little is known about the stability of nanoparticles in relation to particle surface charge. Here, we compared three different surface-stabilized silver nanoparticles (50 nm) for intracellular agglomeration in human hepatocellular carcinoma cells (HepG2). Nanoparticles stabilized with branched polyethyleneimine conferred a positive surface charge, particles stabilized with lipoic acid conferred a negative surface charge, and particles stabilized with polyethylene glycol conferred a neutral surface charge. Particles were incubated in fetal bovine serum, simulated lung surfactant fluid, and simulated stomach digestion fluid. Each nanoparticle system was characterized via microscopic (transmission electron, fluorescence, and enhanced darkfield) and spectroscopic (hyperspectral, dynamic light scattering, and ultraviolet-visible absorption) techniques. Results showed that nanoparticle transformation included cellular internalization, agglomeration, and degradation and that these changes were dependent upon surface charge and incubation matrix. Hyperspectral analyses showed that positively charged silver nanoparticles red-shifted in spectral analysis after transformations, whereas negatively charged silver nanoparticles blue-shifted. Neutrally charged silver nanoparticles did not demonstrate significant spectral shifts. Spectral shifting indicates de-stabilization in particle suspension, which directly affects agglomeration intracellularly. These characteristics are translatable to critical quality attributes and can be exploited when developing nano-carriers for nanomedicine.
Collapse
|
15
|
Cartaxo da Costa Urtiga S, Rodrigues Marcelino H, Sócrates Tabosa do Egito E, Eleamen Oliveira E. Xylan in drug delivery: A review of its engineered structures and biomedical applications. Eur J Pharm Biopharm 2020; 151:199-208. [DOI: 10.1016/j.ejpb.2020.04.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 04/17/2020] [Accepted: 04/22/2020] [Indexed: 12/15/2022]
|
16
|
Enea M, Pereira E, Silva DD, Costa J, Soares ME, de Lourdes Bastos M, Carmo H. Study of the intestinal uptake and permeability of gold nanoparticles using both in vitro and in vivo approaches. NANOTECHNOLOGY 2020; 31:195102. [PMID: 31962292 DOI: 10.1088/1361-6528/ab6dfb] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Gold nanoparticles (AuNPs) are highly attractive to biomedical applications. Here, we investigated the effects of (i) ca. 15 nm spherical AuNPs capped with citrate or 11-mercaptoundecanoic acid (MUA) and (ii) ca. 60 nm spherical citrate-capped AuNPs, and ca. 60 nm MUA-capped star-shaped AuNPs on the cytotoxicity, cellular uptake and permeability, using media supplemented or not with 1% fetal bovine serum (FBS) on caucasian colon adenocarcinoma Caco-2 cells. In addition, the colloidal stability of the nanoparticles in media (supplemented or not) was assessed after 24 h-incubations at 60 μM. The 60 nm gold nanospheres and stars were administrated orally to Wistar rats in order to evaluate their systemic absorption and biodistribution after 24 h. At non-supplemented media settings, citrate-capped gold nanoparticles seem to be more toxic than their MUA-capped counterparts. Also, smaller nanoparticles show higher toxicity than larger ones. The use of cell culture media with 1% FBS not only increased the stability of all AuNPs, as also significantly reduced their cytotoxicity. In the uptake studies, higher AuNPs incorporation was noticed in serum supplemented media, this effect being particularly significant for the 60 nm nanoparticles. Cellular incorporation depended also on the capping agent and size. None of the tested samples crossed the in vitro intestinal barrier. Confirming the in vitro results, the in vivo biodistribution study of the 60 nm AuNPs orally given to rats showed that their systemic absorption is low and that they are mainly eliminated through the faeces. Altogether, these preliminary results suggest that our novel AuNPs have high potential to be considered promising candidates for application in diagnostics or drug delivery at the intestinal level, showing high biocompatibility. However, unless it is desired that these nanomaterials avoid systemic absorption upon oral administration, additional functionalization should be sought to increase their low bioavailability.
Collapse
Affiliation(s)
- Maria Enea
- UCIBIO REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, Porto, 4050-313, Portugal. LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, s/n, Porto, 4169-007, Portugal
| | | | | | | | | | | | | |
Collapse
|
17
|
Moura RP, Pacheco C, Pêgo AP, des Rieux A, Sarmento B. Lipid nanocapsules to enhance drug bioavailability to the central nervous system. J Control Release 2020; 322:390-400. [PMID: 32247807 DOI: 10.1016/j.jconrel.2020.03.042] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 12/15/2022]
Abstract
The central nervous system (CNS), namely the brain, still remains as the hardest area of the human body to achieve adequate concentration levels of most drugs, mainly due to the limiting behavior of its physical and biological defenses. Lipid nanocapsules emerge as a versatile platform to tackle those barriers, and efficiently delivery different drug payloads due to their numerous advantages. They can be produced in a fast, solvent-free and scalable-up process, and their properties can be fine-tuned for to make an optimal brain drug delivery vehicle. Moreover, lipid nanocapsule surface modification can further improve their bioavailability towards the central nervous system. Coupling these features with alternative delivery methods that stem to disrupt or fully circumvent the blood-brain barrier may fully harness the therapeutic advance that lipid nanocapsules can supply to current treatment options. Thus, this review intends to critically address the development of lipid nanocapsules, as well as to highlight the key features that can be modulated to ameliorate their properties towards the central nervous system delivery, mainly through intravenous methods, and how the pathological microenvironment of the CNS can be taken advantage of. The different routes to promote drug delivery towards the brain parenchyma are also discussed, as well as the synergetic effect that can be obtained by combining modified lipid nanocapsules with new/smart administration routes.
Collapse
Affiliation(s)
- Rui Pedro Moura
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Catarina Pacheco
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Ana Paula Pêgo
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; FEUP - Faculdade de Engenharia da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| | - Anne des Rieux
- Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue E. Mounier 73, 1200 Brussels, Belgium
| | - Bruno Sarmento
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal.
| |
Collapse
|
18
|
Hort MA, Alves BDS, Ramires Júnior OV, Falkembach MC, Araújo GDMS, Fernandes CLF, Tavella RA, Bidone J, Dora CL, da Silva Júnior FMR. In vivo toxicity evaluation of nanoemulsions for drug delivery. Drug Chem Toxicol 2019; 44:585-594. [PMID: 31476915 DOI: 10.1080/01480545.2019.1659806] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Lipid nanocarriers (LNs), for example nanoemulsions (NE), are an emerging tool for drug delivery due to their ability to incorporate drugs, protect the drug from degradation, improve bioavailability, and control release. Although LNs are widely studied and applied, especially in the pharmaceutical field, knowledge about their toxicity is scarce. Moreover, the majority of studies focus on their efficiency rather than safety. Thus, the aim of this study was to evaluate the possible toxic effects of NE in vivo. Male Wistar rats (2 months old, 250 g) were treated once daily for 21 days with NE via oral or intraperitoneal delivery at 200, 400 or 800 mg lipid/kg body weight. At the end of the experiment, biochemical, hematological, oxidative stress, and genotoxicity parameters were analyzed. Our results showed that treatment with NE did not modify organ weight or biochemical parameters when compared to controls. The highest NE dose (800 mg/kg) via intraperitoneal injection caused changes in hematological parameters, namely increased plasma proteins, platelets, total leukocytes, and neutrophils, findings that suggest an inflammatory reaction. Further, the same dose evoked lipid peroxidation in the liver. Taken together, the results from this study suggest that NEs can be considered safe for oral administration, but high doses via the parenteral route can cause toxic effects. This study contributes to knowledge about NE toxicity and provides important data about their safe use in the pharmaceutical field.
Collapse
Affiliation(s)
- Mariana Appel Hort
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, Brazil.,Programa de Pós-graduação em Ciências da Saúde, Faculdade de Medicina, Universidade Federal do Rio Grande - FURG, Rio Grande, Brazil
| | - Barbara da Silva Alves
- Programa de Pós-graduação em Ciências da Saúde, Faculdade de Medicina, Universidade Federal do Rio Grande - FURG, Rio Grande, Brazil
| | - Osmar Vieira Ramires Júnior
- Programa de Pós-graduação em Ciências da Saúde, Faculdade de Medicina, Universidade Federal do Rio Grande - FURG, Rio Grande, Brazil
| | - Mariana Correa Falkembach
- Programa de Pós-graduação em Ciências da Saúde, Faculdade de Medicina, Universidade Federal do Rio Grande - FURG, Rio Grande, Brazil
| | - Gabriela de Moraes Soares Araújo
- Programa de Pós-graduação em Ciências da Saúde, Faculdade de Medicina, Universidade Federal do Rio Grande - FURG, Rio Grande, Brazil
| | - Caroline Lopes Feijo Fernandes
- Programa de Pós-graduação em Ciências da Saúde, Faculdade de Medicina, Universidade Federal do Rio Grande - FURG, Rio Grande, Brazil
| | - Ronan Adler Tavella
- Programa de Pós-graduação em Ciências da Saúde, Faculdade de Medicina, Universidade Federal do Rio Grande - FURG, Rio Grande, Brazil
| | - Juliana Bidone
- Departamento de Farmácia, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Cristiana Lima Dora
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, Brazil.,Programa de Pós-graduação em Ciências da Saúde, Faculdade de Medicina, Universidade Federal do Rio Grande - FURG, Rio Grande, Brazil
| | - Flavio Manoel Rodrigues da Silva Júnior
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, Brazil.,Programa de Pós-graduação em Ciências da Saúde, Faculdade de Medicina, Universidade Federal do Rio Grande - FURG, Rio Grande, Brazil
| |
Collapse
|
19
|
Yang H, Du L, Wu G, Wu Z, Keelan JA. Murine exposure to gold nanoparticles during early pregnancy promotes abortion by inhibiting ectodermal differentiation. Mol Med 2018; 24:62. [PMID: 30509178 PMCID: PMC6276159 DOI: 10.1186/s10020-018-0061-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 11/07/2018] [Indexed: 01/16/2023] Open
Abstract
Background Gold nanoparticles (AuNPs) have been widely studied for biomedical applications, although their safety and potential toxicity in pregnancy remains unknown. The aim of this study is to explore the effect of AuNPs maternal exposure at different gestational ages on fetal survival and development, as well as the potential mechanism of AuNPs affecting embryos and fetuses. Methods Thirty nm polyethylene glycol (PEG)-coated AuNPs (A30) were administered to pregnant mice via intravenous injection (5 μg Au/g body weight) over three days at either early or late pregnancy. Fetal abortion rate and morphological development in E16.5 were then detected in detail. The pregnant mice physiological states with A30 exposure were examined by biochemical, histological or imaging methods; and materno-fetal distribution of gold elements was assayed by electron microscopy and mass spectrometry. Murine embryonic stem cells derived embryoid-bodies or neuroectodermal cells were treated with A30 (0.0025 to 0.25 μg Au/mL) to examine A30 effects on expression levels of the germ differentiation marker genes. Tukey’s method was used for statistical analysis. Results Exposure to A30 during early (A30E) but not late (A30L) pregnancy caused a high abortion rate (53.5%), lower fetal survival rate and abnormal decidualization compared with non-exposed counterparts. The developmental damage caused by A30 followed an “all-or-nothing” pattern, as the non-aborted fetuses developed normally and pregnancies maintained normal endocrine values. A30 caused minor impairment of liver and kidney function of A30E but not A30L mice. TEM imaging of fetal tissue sections confirmed the transfer of A30 into fetal brain and live as aggregates. qPCR assays showed A30 suppressed the expression of ectodermal, but not mesodermal and endodermal differentiation markers. Conclusions These results illustrate that maternal A30 exposure in early pregnant results in A30 transfer into embryonic tissues, inhibiting ectodermal differentiation of embryonic stem cells, leading to abnormal embryonic development and abortion. While exposure to A30 during late pregnancy had little or no impact on dams and fetuses. These findings suggest the safety of biomedical applications employing AuNPs during pregnancy is strongly influenced by fetal maturity and gestational age at exposure and provide the clues for AuNPs safe application period in pregnancy. Electronic supplementary material The online version of this article (10.1186/s10020-018-0061-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hui Yang
- Immunology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Libo Du
- Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Guangjun Wu
- Immunology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Zhenyu Wu
- Immunology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Jeffrey A Keelan
- Division of Obstetrics & Gynaecology, Faculty of Health and Medical Sciences, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
20
|
In Vitro Evaluation of Antimicrobial Activity and Cytotoxicity of Different Nanobiotics Targeting Multidrug Resistant and Biofilm Forming Staphylococci. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7658238. [PMID: 30622962 PMCID: PMC6304202 DOI: 10.1155/2018/7658238] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 10/24/2018] [Accepted: 11/05/2018] [Indexed: 12/15/2022]
Abstract
Antibiotic-resistant and biofilm-forming bacteria have surprisingly increased over recent years. On the contrary, the rate of development of new antibiotics to treat these emerging superbugs is very slow. Therefore, the aim of this study was to prepare novel nanobiotic formulations to improve the antimicrobial activity of three antibiotics (linezolid, doxycycline, and clindamycin) against Staphylococci. Antibiotics were formulated as nanoemulsions and evaluated for their antimicrobial activities and cytotoxicities. Cytotoxicity of the conventional antibiotics and nanobiotics was analyzed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay on rat hepatocytes. Half-maximal inhibitory concentration (IC50) was estimated from an experimentally derived dose-response curve for each concentration using GraphPad Prism software. Upon quantitative assessment of Staphylococcus biofilm formation, eighty-four isolates (66.14 %) were biofilm forming. Linezolid and doxycycline nanobiotics exhibited promising antibacterial activities. On the contrary, clindamycin nanobiotic exhibited poor antibacterial activity. Minimum biofilm inhibitory concentrations showed that 73.68 %, 45.6%, and 5.2% of isolates were sensitive to linezolid, doxycycline, and clindamycin nanobiotics, respectively. Results of this study revealed that antibiotics loaded in nanosystems had a higher antimicrobial activity and lower cytotoxicities as compared to those of conventional free antibiotics, indicating their potential therapeutic values.
Collapse
|
21
|
Scioli Montoto S, Sbaraglini ML, Talevi A, Couyoupetrou M, Di Ianni M, Pesce GO, Alvarez VA, Bruno-Blanch LE, Castro GR, Ruiz ME, Islan GA. Carbamazepine-loaded solid lipid nanoparticles and nanostructured lipid carriers: Physicochemical characterization and in vitro/in vivo evaluation. Colloids Surf B Biointerfaces 2018; 167:73-81. [PMID: 29627680 DOI: 10.1016/j.colsurfb.2018.03.052] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 02/21/2018] [Accepted: 03/30/2018] [Indexed: 01/16/2023]
Abstract
Solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) represent promising alternatives for drug delivery to the central nervous system. In the present work, four different nanoformulations of the antiepileptic drug Carbamazepine (CBZ) were designed and prepared by the homogenization/ultrasonication method, with encapsulation efficiencies ranging from 82.8 to 93.8%. The formulations remained stable at 4 °C for at least 3 months. Physicochemical and microscopic characterization were performed by photon correlation spectroscopy (PCS), transmission electron microscopy (TEM), atomic force microscopy (AFM); thermal properties by differential scanning calorimetry (DSC), thermogravimetry (TGA) and X-ray diffraction analysis (XRD). The results indicated the presence of spherical shape nanoparticles with a mean particle diameter around 160 nm in a narrow size distribution; the entrapped CBZ displayed an amorphous state. The in vitro release profile of CBZ fitted into a Baker-Lonsdale model for spherical matrices and almost the 100% of the encapsulated drug was released in a controlled manner during the first 24 h. The apparent permeability of CBZ-loaded nanoparticles through a cell monolayer model was similar to that of the free drug. In vivo experiments in a mice model of seizure suggested protection by CBZ-NLC against seizures for at least 2 h after intraperitoneal administration. The developed CBZ-loaded lipid nanocarriers displayed optimal characteristics of size, shape and drug release and possibly represent a promising tool to improve the treatment of refractory epilepsy linked to efflux transporters upregulation.
Collapse
Affiliation(s)
- S Scioli Montoto
- Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP), Calle 47 y 115, B1900AJI, La Plata, Buenos Aires, Argentina
| | - M L Sbaraglini
- Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP), Calle 47 y 115, B1900AJI, La Plata, Buenos Aires, Argentina
| | - A Talevi
- Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP), Calle 47 y 115, B1900AJI, La Plata, Buenos Aires, Argentina
| | - M Couyoupetrou
- Departamento de Farmacología, Instituto Nacional de Medicamentos (INAME), Administración Nacional de Medicamentos, Alimentos y Tecnología Médica (ANMAT), CABA, Buenos Aires, Argentina
| | - M Di Ianni
- Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP), Calle 47 y 115, B1900AJI, La Plata, Buenos Aires, Argentina
| | - G O Pesce
- Departamento de Farmacología, Instituto Nacional de Medicamentos (INAME), Administración Nacional de Medicamentos, Alimentos y Tecnología Médica (ANMAT), CABA, Buenos Aires, Argentina
| | - V A Alvarez
- Grupo de Materiales Compuestos de Matriz Polimérica (CoMP), Instituto de Investigaciones en Ciencia y Tecnología de Materiales (INTEMA), Facultad de Ingeniería, Universidad Nacional de Mar del Plata (UNMDP) - CONICET, Solis 7575, B7608FDQ, Mar del Plata, Buenos Aires, Argentina
| | - L E Bruno-Blanch
- Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP), Calle 47 y 115, B1900AJI, La Plata, Buenos Aires, Argentina
| | - G R Castro
- Laboratorio de Nanobiomateriales, CINDEFI, Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP) -CONICET (CCT La Plata), Calle 47 y 115, B1900AJI, La Plata, Buenos Aires, Argentina
| | - M E Ruiz
- Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP), Calle 47 y 115, B1900AJI, La Plata, Buenos Aires, Argentina.
| | - G A Islan
- Laboratorio de Nanobiomateriales, CINDEFI, Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata (UNLP) -CONICET (CCT La Plata), Calle 47 y 115, B1900AJI, La Plata, Buenos Aires, Argentina.
| |
Collapse
|
22
|
Shrestha N, Bouttefeux O, Vanvarenberg K, Lundquist P, Cunarro J, Tovar S, Khodus G, Andersson E, Keita ÅV, Gonzalez Dieguez C, Artursson P, Préat V, Beloqui A. The stimulation of GLP-1 secretion and delivery of GLP-1 agonists via nanostructured lipid carriers. NANOSCALE 2018; 10:603-613. [PMID: 29235598 DOI: 10.1039/c7nr07736j] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Nanoparticulate based drug delivery systems have been extensively studied to efficiently encapsulate and deliver peptides orally. However, most of the existing data mainly focus on the nanoparticles as a drug carrier, but the ability of nanoparticles having a biological effect has not been exploited. Herein, we hypothesize that nanostructured lipid carriers (NLCs) could activate the endogenous glucagon-like peptide-1 (GLP-1) secretion and also act as oral delivery systems for GLP-1 analogs (exenatide and liraglutide). NLCs effectively encapsulated the peptides, the majority of which were only released under the intestinal conditions. NLCs, with and without peptide encapsulation, showed effective induction of GLP-1 secretion in vitro from the enteroendocrinal L-cells (GLUTag). NLCs also showed a 2.9-fold increase in the permeability of exenatide across the intestinal cell monolayer. The intestinal administration of the exenatide and liraglutide loaded NLCs did not demonstrate any glucose lowering effect on normal mice. Further, ex vivo studies depicted that the NLCs mainly adhered to the mucus layer. In conclusion, this study demonstrates that NLCs need further optimization to overcome the mucosal barrier in the intestine; nonetheless, this study also presents a promising strategy to use a dual-action drug delivery nanosystem which synergizes its own biological effect and that of the encapsulated drug molecule.
Collapse
Affiliation(s)
- Neha Shrestha
- Université catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200 Brussels, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Phase-Transition Nanodroplets for Real-Time Photoacoustic/Ultrasound Dual-Modality Imaging and Photothermal Therapy of Sentinel Lymph Node in Breast Cancer. Sci Rep 2017; 7:45213. [PMID: 28338071 PMCID: PMC5364557 DOI: 10.1038/srep45213] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 02/21/2017] [Indexed: 02/05/2023] Open
Abstract
Pathological status of lymph nodes (LNs) plays a critical role in staging and treatment for the patients with breast cancer. Sentinel lymph node biopsy has become the standard method in determining pathological status of axillary LNs. Therefore, the determination of sentinel lymph nodes (SLNs) and therapy of metastatic LNs are highly desirable in clinic. Herein, an unprecedented carbon nanoparticles (CNs)-incorporated liquid-gas phase-transition nanodroplets (CNPs) with strong near-infrared (NIR) absorption, good biocompatibility, excellent photoacoustic (PA) and ultrasound (US) contrast, and high photothermal-conversion efficiency are reported in this study. Upon laser irradiation, liquid-gas phase transition of the CNPs has been demonstrated to provide excellent contrasts for PA/US dual-modality imaging both in vitro and in vivo. Additionally, the CNPs are capable of staining lymph nodes, which can contribute significantly to the identification of LNs with naked eyes. With increased laser energy, the CNPs exhibit the high performance in killing the breast cancer cells both in vitro and in vivo, due to the photothermal effect induced from the CNs within CNPs. These results suggest that the developed multifunctional phase-transition nanodroplets have high potential to act as the theranostic agents in both SLNs detection and therapy of metastatic LNs.
Collapse
|
24
|
Griffin BT, Guo J, Presas E, Donovan MD, Alonso MJ, O'Driscoll CM. Pharmacokinetic, pharmacodynamic and biodistribution following oral administration of nanocarriers containing peptide and protein drugs. Adv Drug Deliv Rev 2016; 106:367-380. [PMID: 27320644 DOI: 10.1016/j.addr.2016.06.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 06/07/2016] [Accepted: 06/10/2016] [Indexed: 12/17/2022]
Abstract
The influence of nanoparticle (NP) formulations on the pharmacokinetic, pharmacodynamic and biodistribution profiles of peptide- and protein-like drugs following oral administration is critically reviewed. The possible mechanisms of absorption enhancement and the effects of the physicochemical properties of the NP are examined. The potential advantages and challenges of physiologically-based pharmacokinetic (PBPK) modelling to help predict efficacy in man are discussed. The importance of developing and expanding the regulatory framework to help translate the technology into the clinic and accelerate the availability of oral nanoparticulate formulations is emphasized. In conclusion, opportunities for future work to improve the state of the art of oral nanomedicines are identified.
Collapse
|
25
|
Assa F, Jafarizadeh-Malmiri H, Ajamein H, Vaghari H, Anarjan N, Ahmadi O, Berenjian A. Chitosan magnetic nanoparticles for drug delivery systems. Crit Rev Biotechnol 2016; 37:492-509. [DOI: 10.1080/07388551.2016.1185389] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
26
|
Lyubartsev AP, Rabinovich AL. Force Field Development for Lipid Membrane Simulations. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:2483-2497. [PMID: 26766518 DOI: 10.1016/j.bbamem.2015.12.033] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 12/21/2015] [Accepted: 12/23/2015] [Indexed: 02/04/2023]
Abstract
With the rapid development of computer power and wide availability of modelling software computer simulations of realistic models of lipid membranes, including their interactions with various molecular species, polypeptides and membrane proteins have become feasible for many research groups. The crucial issue of the reliability of such simulations is the quality of the force field, and many efforts, especially in the latest several years, have been devoted to parametrization and optimization of the force fields for biomembrane modelling. In this review, we give account of the recent development in this area, covering different classes of force fields, principles of the force field parametrization, comparison of the force fields, and their experimental validation. This article is part of a Special Issue entitled: Biosimulations edited by Ilpo Vattulainen and Tomasz Róg.
Collapse
Affiliation(s)
- Alexander P Lyubartsev
- Department of Materials and Environmental Chemistry, Stockholm University, SE 106 91, Stockholm, Sweden.
| | - Alexander L Rabinovich
- Institute of Biology, Karelian Research Center, Russian Academy of Sciences, Pushkinskaya 11, Petrozavodsk, 185910, Russian Federation.
| |
Collapse
|
27
|
Wang Y, Kaur G, Chen Y, Santos A, Losic D, Evdokiou A. Bioinert Anodic Alumina Nanotubes for Targeting of Endoplasmic Reticulum Stress and Autophagic Signaling: A Combinatorial Nanotube-Based Drug Delivery System for Enhancing Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2015; 7:27140-27151. [PMID: 26556288 DOI: 10.1021/acsami.5b07557] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Although nanoparticle-based targeted delivery systems have gained promising achievements for cancer therapy, the development of sophisticated strategies with effective combinatorial therapies remains an enduring challenge. Herein, we report the fabrication of a novel nanomaterial, so-called anodic alumina nanotubes (AANTs) for proof-of-concept cancer therapy by targeting cell signaling networks. This strategy is to target autophagic and endoplasmic reticulum (ER) stress signaling by using thapsigargin (TG)-loaded AANTs cotreated with an autophagy inhibitor 3-methyladenine (3-MA). We first show that AANTs are nontoxic and can activate autophagy in different cell types including human fibroblast cells (HFF), human monocyte cells (THP-1), and human breast cancer cells (MDA-MB 231-TXSA). Treatment with 3-MA at a nontoxic dose reduced the level of autophagy induced by AANTs, and consequently sensitized breast cancer cells to AANTs-induced cellular stresses. To target autophagic and ER stress signaling networking, breast cancer cells were treated with 3-MA together with AANTs loaded with the prototype ER stress inducer TG. We demonstrated that 3-MA enhanced the cancer cell killing effect of AANTs loaded with TG. This effect was associated with enhanced ER stress signaling due to the combination effect of TG and 3-MA. These findings not only demonstrate the excellent biocompatibility of AANTs as novel biomaterials but also provide new opportunities for developing ER- and autophagy-targeted delivery systems for future clinical cancer therapy.
Collapse
Affiliation(s)
- Ye Wang
- School of Chemical Engineering and ‡Discipline of Surgery, Basil Hetzel Institute, The University of Adelaide , Adelaide, South Australia 5005, Australia
| | - Gagandeep Kaur
- School of Chemical Engineering and ‡Discipline of Surgery, Basil Hetzel Institute, The University of Adelaide , Adelaide, South Australia 5005, Australia
| | - Yuting Chen
- School of Chemical Engineering and ‡Discipline of Surgery, Basil Hetzel Institute, The University of Adelaide , Adelaide, South Australia 5005, Australia
| | - Abel Santos
- School of Chemical Engineering and ‡Discipline of Surgery, Basil Hetzel Institute, The University of Adelaide , Adelaide, South Australia 5005, Australia
| | - Dusan Losic
- School of Chemical Engineering and ‡Discipline of Surgery, Basil Hetzel Institute, The University of Adelaide , Adelaide, South Australia 5005, Australia
| | - Andreas Evdokiou
- School of Chemical Engineering and ‡Discipline of Surgery, Basil Hetzel Institute, The University of Adelaide , Adelaide, South Australia 5005, Australia
| |
Collapse
|
28
|
Competitive photometric enzyme immunoassay for fullerene C60 and its derivatives using a fullerene conjugated to horseradish peroxidase. Mikrochim Acta 2015. [DOI: 10.1007/s00604-015-1621-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
29
|
Araújo F, Shrestha N, Shahbazi MA, Liu D, Herranz-Blanco B, Mäkilä EM, Salonen JJ, Hirvonen JT, Granja PL, Sarmento B, Santos HA. Microfluidic Assembly of a Multifunctional Tailorable Composite System Designed for Site Specific Combined Oral Delivery of Peptide Drugs. ACS NANO 2015; 9:8291-8302. [PMID: 26235314 DOI: 10.1021/acsnano.5b02762] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Multifunctional tailorable composite systems, specifically designed for oral dual-delivery of a peptide (glucagon-like peptide-1) and an enzymatic inhibitor (dipeptidyl peptidase 4 (DPP4)), were assembled through the microfluidics technique. Both drugs were coloaded into these systems for a synergistic therapeutic effect. The systems were composed of chitosan and cell-penetrating peptide modified poly(lactide-co-glycolide) and porous silicon nanoparticles as nanomatrices, further encapsulated in an enteric hydroxypropylmethylcellulose acetylsuccinate polymer. The developed multifunctional systems were pH-sensitive, inherited by the enteric polymer, enabling the release of the nanoparticles only in the simulated intestinal conditions. Moreover, the encapsulation into this polymer prevented the degradation of the nanoparticles' modifications. These nanoparticles showed strong and higher interactions with the intestinal cells in comparison with the nonmodified ones. The presence of DPP4 inhibitor enhanced the peptide permeability across intestinal cell monolayers. Overall, this is a promising platform for simultaneously delivering two drugs from a single formulation. Through this approach peptides are expected to increase their bioavailability and efficiency in vivo both by their specific release at the intestinal level and also by the reduced enzymatic activity. The use of this platform, specifically in combination of the two antidiabetic drugs, has clinical potential for the therapy of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Francisca Araújo
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki , FI-00014 Helsinki, Finland
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto , 4150-180 Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, University of Porto , 4150-180 Porto, Portugal
- ICBAS - Instituto Ciências Biomédicas Abel Salazar, University of Porto , 4150-180 Porto, Portugal
| | - Neha Shrestha
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki , FI-00014 Helsinki, Finland
| | - Mohammad-Ali Shahbazi
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki , FI-00014 Helsinki, Finland
| | - Dongfei Liu
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki , FI-00014 Helsinki, Finland
| | - Bárbara Herranz-Blanco
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki , FI-00014 Helsinki, Finland
| | - Ermei M Mäkilä
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki , FI-00014 Helsinki, Finland
- Laboratory of Industrial Physics, University of Turku , FI-20014 Turku, Finland
| | - Jarno J Salonen
- Laboratory of Industrial Physics, University of Turku , FI-20014 Turku, Finland
| | - Jouni T Hirvonen
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki , FI-00014 Helsinki, Finland
| | - Pedro L Granja
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto , 4150-180 Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, University of Porto , 4150-180 Porto, Portugal
- ICBAS - Instituto Ciências Biomédicas Abel Salazar, University of Porto , 4150-180 Porto, Portugal
| | - Bruno Sarmento
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto , 4150-180 Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, University of Porto , 4150-180 Porto, Portugal
- CESPU , Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, 4585-116 Gandra, Portugal
| | - Hélder A Santos
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki , FI-00014 Helsinki, Finland
| |
Collapse
|
30
|
Kumar S, Randhawa JK. Solid lipid nanoparticles of stearic acid for the drug delivery of paliperidone. RSC Adv 2015. [DOI: 10.1039/c5ra10642g] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Paliperidone is an antipsychotic drug having poor water solubility and bioavailability. Solid lipid nanoparticles of stearic acid loaded with paliperidone were prepared to enhance the bioavailability.
Collapse
Affiliation(s)
- Sacheen Kumar
- Centre for Material Science and Engineering
- National Institute of Technology
- India-177005
| | | |
Collapse
|