1
|
Caselli L, Du G, Micciulla S, Traini T, Sebastiani F, Diedrichsen RG, Köhler S, Skoda MWA, van der Plas MJA, Malmsten M. Photocatalytic Degradation of Bacterial Lipopolysaccharides by Peptide-Coated TiO 2 Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2024; 16:60056-60069. [PMID: 39443826 PMCID: PMC11551910 DOI: 10.1021/acsami.4c15706] [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: 09/13/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
In this study, we report the degradation of smooth and rough lipopolysaccharides (LPS) from Gram-negative bacteria and of lipoteichoic acid (LTA) from Gram-positive bacteria by peptide-coated TiO2 nanoparticles (TiO2 NPs). While bare TiO2 NPs displayed minor binding to both LPS and LTA, coating TiO2 NPs with the antimicrobial peptide LL-37 dramatically increased the level of binding to both LPS and LTA, decorating these uniformly. Importantly, peptide coating did not suppress reactive oxygen species generation of TiO2 NPs; hence, UV illumination triggered pronounced degradation of LPS and LTA by peptide-coated TiO2 NPs. Structural consequences of oxidative degradation were examined by neutron reflectometry for smooth LPS, showing that degradation occurred preferentially in its outer O-antigen tails. Furthermore, cryo-TEM and light scattering showed lipopolysaccharide fragments resulting from degradation to be captured by the NP/lipopolysaccharide coaggregates. The capacity of LL-37-TiO2 NPs to capture and degrade LPS and LTA was demonstrated to be of importance for their ability to suppress lipopolysaccharide-induced activation in human monocytes at simultaneously low toxicity. Together, these results suggest that peptide-coated photocatalytic NPs offer opportunities for the confinement of infection and inflammation.
Collapse
Affiliation(s)
- Lucrezia Caselli
- Department
of Physical Chemistry 1, Lund University, Lund SE-22100, Sweden
| | - Guanqun Du
- Department
of Physical Chemistry 1, Lund University, Lund SE-22100, Sweden
| | | | - Tanja Traini
- Department
of Pharmacy, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Federica Sebastiani
- Department
of Physical Chemistry 1, Lund University, Lund SE-22100, Sweden
- Department
of Pharmacy, University of Copenhagen, Copenhagen DK-2100, Denmark
| | | | - Sebastian Köhler
- LINXS
Institute of Advanced Neutron and X-ray Science, Scheelevagen 19, Lund 22370, Sweden
| | - Maximilian W. A. Skoda
- ISIS
Pulsed Neutron and Muon Source, Rutherford
Appleton Laboratory, Harwell OX11 0QX, U.K.
| | | | - Martin Malmsten
- Department
of Physical Chemistry 1, Lund University, Lund SE-22100, Sweden
- Department
of Pharmacy, University of Copenhagen, Copenhagen DK-2100, Denmark
| |
Collapse
|
2
|
Zhou J, Jiang Z, Sun R, Pan D, Du Q, Zhou X, Chen Y, Chen Y, Peng J. Comparison of cell delivery and cell membrane camouflaged PLGA nanoparticles in the delivery of shikonin for colorectal cancer treatment. Colloids Surf B Biointerfaces 2024; 241:114017. [PMID: 38865869 DOI: 10.1016/j.colsurfb.2024.114017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
Inspired by the "natural camouflage" strategy, cell-based biomimetic drug delivery systems (BDDS) have shown great potential in cancer therapy. Red blood cell (RBC) delivery vehicles and red blood cell membrane (RBCm)-camouflaged vehicles were commonly used strategies for drug delivery. We prepared shikonin-encapsulated PLGA nanoparticles (PLGA/SK) with different surface charges to obtain both RBC delivery and RBCm-camouflaged PLGA NPs. The physicochemical properties, in vivo circulation and antitumor effects of these biomimetic preparations were studied. Since the positive PLGA NPs may affect the morphology and function of RBCs, the biomimetic preparations prepared by the negative PLGA NPs showed better in vitro stability. However, positive PLGA NP-based biomimetic preparations exhibited longer circulation time and higher tumor region accumulation, leading to stronger anti-tumor effects. Meanwhile, the RBC delivery PLGA(+) NPs possessed better in vitro cytotoxicity, longer circulation time and higher tumor accumulation than RBCm-camouflaged PLGA(+) NPs. Collectively, RBC delivery vehicles possessed more potential than RBCm-camouflaged vehicles on drug delivery for tumor treatment, especially with positive NPs-loaded.
Collapse
Affiliation(s)
- Jia Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Zhaohui Jiang
- The First People's Hospital of Guiyang, Guizhou 550002, China
| | - Runbin Sun
- Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Jiangsu 210008, China
| | - Di Pan
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Qianming Du
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Jiangsu 210006, China
| | - Xiang Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Yi Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China.
| | - Yan Chen
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China.
| | - Jianqing Peng
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China.
| |
Collapse
|
3
|
Yew JS, Ong SK, Lim HX, Tan SH, Ong KC, Wong KT, Poh CL. Immunogenicity of trivalent DNA vaccine candidate encapsulated in Chitosan-TPP nanoparticles against EV-A71 and CV-A16. Nanomedicine (Lond) 2024; 19:1779-1799. [PMID: 39140594 PMCID: PMC11418279 DOI: 10.1080/17435889.2024.2372243] [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: 03/08/2024] [Accepted: 06/21/2024] [Indexed: 08/15/2024] Open
Abstract
Aim: To develop a trivalent DNA vaccine candidate encapsulated in Chitosan-TPP nanoparticles against hand foot and mouth disease (HFMD) and assess its immunogenicity in mice.Materials & methods: Trivalent plasmid carrying the VP1 and VP2 genes of EV-A71, VP1 gene of CV-A16 was encapsulated in Chitosan-TPP nanoparticles through ionic gelation. In vitro characterization and in vivo immunization studies of the CS-TPP-NPs (pIRES-VP121) were performed.Results: Mice administered with CS-TPP NPs (pIRES-VP121) intramuscularly were observed to have the highest IFN-γ response. Sera from mice immunized with the naked pDNA and CS-TPP-NPs (pIRES-VP121) demonstrated good viral clearance against wild-type EV-A71 and CV-A16 in RD cells.Conclusion: CS-TPP-NPs (pIRES-VP121) could serve as a prototype for future development of multivalent HFMD DNA vaccine candidates.
Collapse
Affiliation(s)
- Jia Sheng Yew
- Centre for Virus & Vaccine Research, School of Medical & Life Sciences, Sunway University, Petaling Jaya, 47500, Malaysia
| | - Seng-Kai Ong
- Department of Biological science, School of Medical & Life Sciences, Sunway University, Petaling Jaya, 47500, Malaysia
| | - Hui Xuan Lim
- Centre for Virus & Vaccine Research, School of Medical & Life Sciences, Sunway University, Petaling Jaya, 47500, Malaysia
- Sunway Microbiome Centre, School of Medical & Life Sciences, Sunway University, Petaling Jaya, 47500, Malaysia
| | - Soon Hao Tan
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Federal Territory of Kuala Lumpur, Kuala Lumpur, 50603, Malaysia
| | - Kien Chai Ong
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Federal Territory of Kuala Lumpur, Kuala Lumpur, 50603, Malaysia
| | - Kum Thong Wong
- Department of Pathology, Faculty of Medicine, Universiti Malaya, Federal Territory of Kuala Lumpur, Kuala Lumpur, 50603, Malaysia
| | - Chit Laa Poh
- Centre for Virus & Vaccine Research, School of Medical & Life Sciences, Sunway University, Petaling Jaya, 47500, Malaysia
- ALPS Global Holding Berhad, The ICON, No.1, Off Jalan Tun Razak, Kuala Lumpur, 50400, Malaysia
| |
Collapse
|
4
|
Caselli L, Parra-Ortiz E, Micciulla S, Skoda MWA, Häffner SM, Nielsen EM, van der Plas MJA, Malmsten M. Boosting Membrane Interactions and Antimicrobial Effects of Photocatalytic Titanium Dioxide Nanoparticles by Peptide Coating. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309496. [PMID: 38402437 DOI: 10.1002/smll.202309496] [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: 10/19/2023] [Revised: 01/30/2024] [Indexed: 02/26/2024]
Abstract
Photocatalytic nanoparticles offer antimicrobial effects under illumination due to the formation of reactive oxygen species (ROS), capable of degrading bacterial membranes. ROS may, however, also degrade human cell membranes and trigger toxicity. Since antimicrobial peptides (AMPs) may display excellent selectivity between human cells and bacteria, these may offer opportunities to effectively "target" nanoparticles to bacterial membranes for increased selectivity. Investigating this, photocatalytic TiO2 nanoparticles (NPs) are coated with the AMP LL-37, and ROS generation is found by C11-BODIPY to be essentially unaffected after AMP coating. Furthermore, peptide-coated TiO2 NPs retain their positive ζ-potential also after 1-2 h of UV illumination, showing peptide degradation to be sufficiently limited to allow peptide-mediated targeting. In line with this, quartz crystal microbalance measurements show peptide coating to promote membrane binding of TiO2 NPs, particularly so for bacteria-like anionic and cholesterol-void membranes. As a result, membrane degradation during illumination is strongly promoted for such membranes, but not so for mammalian-like membranes. The mechanisms of these effects are elucidated by neutron reflectometry. Analogously, LL-37 coating promoted membrane rupture by TiO2 NPs for Gram-negative and Gram-positive bacteria, but not for human monocytes. These findings demonstrate that AMP coating may selectively boost the antimicrobial effects of photocatalytic NPs.
Collapse
Affiliation(s)
- Lucrezia Caselli
- Department of Pharmacy, University of Copenhagen, Copenhagen, DK-2100, Denmark
- Department of Physical Chemistry 1, Lund University, Lund, SE-22100, Sweden
| | - Elisa Parra-Ortiz
- Department of Pharmacy, University of Copenhagen, Copenhagen, DK-2100, Denmark
- Novonesis, Biologiens Vej 2, Lyngby, DK-2800 Kgs, Denmark
| | - Samantha Micciulla
- Institut Laue-Langevin, CS 20156, Grenoble Cedex 9, 38042, France
- Laboratoire Interdisciplinaire de Physique (LIPhy), Saint Martin d'Hères, 38402, France
- Centre National de la Recherche Scientifique (CNRS), Saint-Martin-d'Hères, Auvergne-Rhône-Alpes, France
| | - Maximilian W A Skoda
- ISIS Pulsed Neutron and Muon Source, Rutherford Appleton Laboratory, Harwell, OX11 0QX, UK
| | - Sara Malekkhaiat Häffner
- Department of Pharmacy, University of Copenhagen, Copenhagen, DK-2100, Denmark
- RISE Research Institutes of Sweden, Malvinas väg 3, Stockholm, 114 86, Sweden
| | | | | | - Martin Malmsten
- Department of Pharmacy, University of Copenhagen, Copenhagen, DK-2100, Denmark
- Department of Physical Chemistry 1, Lund University, Lund, SE-22100, Sweden
| |
Collapse
|
5
|
Caselli L, Köhler S, Schirone D, Humphreys B, Malmsten M. Conformational control of antimicrobial peptide amphiphilicity: consequences for boosting membrane interactions and antimicrobial effects of photocatalytic TiO 2 nanoparticles. Phys Chem Chem Phys 2024; 26:16529-16539. [PMID: 38828872 DOI: 10.1039/d4cp01724b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
This study reports on the effects of conformationally controlled amphiphilicity of antimicrobial peptides (AMPs) on their ability to coat TiO2 nanoparticles (NPs) and boost the photocatalytic antimicrobial effects of such NPs. For this, TiO2 NPs were combined with AMP EFK17 (EFKRIVQRIKDFLRNLV), displaying a disordered conformation in aqueous solution but helix formation on interaction with bacterial membranes. The membrane-bound helix is amphiphilic, with all polar and charged amino acid residues located at one side and all non-polar and hydrophobic residues on the other. In contrast, the d-enantiomer variant EFK17-d (E(dF)KR(dI)VQR(dI)KD(dF)LRNLV) is unable to form the amphiphilic helix on bacterial membrane interaction, whereas the W-residues in EFK17-W (EWKRWVQRWKDFLRNLV) boost hydrophobic interactions of the amphiphilic helix. Circular dichroism results showed the effects displayed for the free peptide, to also be present for peptide-coated TiO2 NPs, causing peptide binding to decrease in the order EFK17-W > EFK17 > EFK17-d. Notably, the formation of reactive oxygen species (ROS) by the TiO2 NPs was essentially unaffected by the presence of peptide coating, for all the peptides investigated, and the coatings stabilized over hours of UV exposure. Photocatalytic membrane degradation from TiO2 NPs coated with EFK17-W and EFK17 was promoted for bacteria-like model bilayers containing anionic phosphatidylglycerol but suppressed in mammalian-like bilayers formed by zwitterionic phosphatidylcholine and cholesterol. Structural aspects of these effects were further investigated by neutron reflectometry with clear variations observed between the bacteria- and mammalian-like model bilayers for the three peptides. Mirroring these results in bacteria-like model membranes, combining TiO2 NPs with EFK17-W and EFK17, but not with non-adsorbing EFK17-d, resulted in boosted antimicrobial effects of the resulting cationic composite NPs already in darkness, effects enhanced further on UV illumination.
Collapse
Affiliation(s)
- Lucrezia Caselli
- Department of Physical Chemistry 1, Lund University, SE-22100 Lund, Sweden.
| | - Sebastian Köhler
- LINXS Institute of Advanced Neutron and X-ray Science, Scheelevagen 19, 22370 Lund, Sweden
| | - Davide Schirone
- Department of Biomedical Sciences and Biofilms-Research Center for Biointerfaces (BRCB), Malmö University, 20506 Malmö, Sweden
| | - Ben Humphreys
- Institut Laue-Langevin, CS 20156, 38042 Grenoble Cedex 9, France
| | - Martin Malmsten
- Department of Physical Chemistry 1, Lund University, SE-22100 Lund, Sweden.
- Department of Pharmacy, University of Copenhagen, DK-2100 Copenhagen, Denmark
| |
Collapse
|
6
|
Zhang Y, Luo M, Jia Y, Gao T, Deng L, Gong T, Zhang Z, Cao X, Fu Y. Adipocyte-targeted delivery of rosiglitazone with localized photothermal therapy for the treatment of diet-induced obesity in mice. Acta Biomater 2024; 181:317-332. [PMID: 38643815 DOI: 10.1016/j.actbio.2024.04.029] [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: 02/20/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 04/23/2024]
Abstract
Obesity represents a growing public health concern and is closely associated with metabolic complications such as diabetes and fatty liver disease. Anti-obesity medications currently available have limited efficacy in weight loss and are often accompanied by adverse effects. This study proposes a localized photothermal therapy (PTT) combined with adipocyte-targeted delivery of rosiglitazone (RSG) to address obesity. Specifically, cationic albumin nanoparticles (cNPs) were synthesized to deliver RSG precisely to white adipocytes, stimulating the browning process. An IR780-loaded thermosensitive hydrogel was injected and allowed to gel in situ to afford a subcutaneous reservoir that enables localized PTT and controlled release of RSG cNPs. Notably, cNPs significantly enhanced the internalization efficiency in adipocytes in vitro and prolonged the therapeutic retention in the adipose tissue in vivo. Co-administration of RSG cNPs and PTT substantially reduced fat content, induced browning in white adipose tissue in diet-induced obese mice, and mitigated complications such as insulin resistance, fatty liver, and hyperlipidemia. The increased expression of uncoupling protein 1 contributes to enhancing energy expenditure and facilitating adipose metabolism, thereby effectively combating obesity. This therapeutic approach integrates localized PTT with adipocyte-targeted delivery to combat the global obesity epidemic thus offering a promising solution with reduced systemic toxicity and enhanced efficacy. STATEMENT OF SIGNIFICANCE: Cationic albumin nanoparticles are capable of efficient internalization in adipocytes, which may enhance drug targeting to adipose tissue. The combination of rosiglitazone-loaded cationic albumin nanoparticles and local hyperthermia effectively reduces lipid accumulation in adipocytes and induces an upregulated expression of uncoupling protein 1. The combination therapy effectively inhibits fat accumulation, induces adipocyte browning, and regulates systemic metabolism in diet-induced obese mice.
Collapse
Affiliation(s)
- Yunxiao Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Maoqi Luo
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yaxin Jia
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Tingting Gao
- School of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, and the Grade 3 Pharmaceutical Chemistry Laboratory of State Administrate of Traditional Chinese Medicine, Hefei, 230032, China
| | - Li Deng
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Tao Gong
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xi Cao
- School of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, and the Grade 3 Pharmaceutical Chemistry Laboratory of State Administrate of Traditional Chinese Medicine, Hefei, 230032, China.
| | - Yao Fu
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
7
|
Gemayel J, Chebly A, Kourie H, Hanna C, Mheidly K, Mhanna M, Karam F, Ghoussaini D, Najjar PE, Khalil C. Genome Engineering as a Therapeutic Approach in Cancer Therapy: A Comprehensive Review. ADVANCED GENETICS (HOBOKEN, N.J.) 2024; 5:2300201. [PMID: 38465225 PMCID: PMC10919288 DOI: 10.1002/ggn2.202300201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Indexed: 03/12/2024]
Abstract
Cancer is one of the foremost causes of mortality. The human genome remains stable over time. However, human activities and environmental factors have the power to influence the prevalence of certain types of mutations. This goes to the excessive progress of xenobiotics and industrial development that is expanding the territory for cancers to develop. The mechanisms involved in immune responses against cancer are widely studied. Genome editing has changed the genome-based immunotherapy process in the human body and has opened a new era for cancer treatment. In this review, recent cancer immunotherapies and the use of genome engineering technology are largely focused on.
Collapse
Affiliation(s)
- Jack Gemayel
- Faculty of SciencesBalamand UniversityBeirutLebanon
- FMPS Holding BIOTECKNO s.a.l. Research and Quality SolutionsNaccashBeirut60 247Lebanon
| | - Alain Chebly
- Center Jacques Loiselet for Medical Genetics and Genomics (CGGM), Faculty of MedicineSaint Joseph UniversityBeirutLebanon
- Higher Institute of Public HealthSaint Joseph UniversityBeirutLebanon
| | - Hampig Kourie
- Center Jacques Loiselet for Medical Genetics and Genomics (CGGM), Faculty of MedicineSaint Joseph UniversityBeirutLebanon
- Faculty of MedicineSaint Joseph UniversityBeirutLebanon
| | - Colette Hanna
- Faculty of MedicineLebanese American University Medical CenterRizk HospitalBeirutLebanon
| | | | - Melissa Mhanna
- Faculty of MedicineParis Saclay University63 Rue Gabriel PériLe Kremlin‐Bicêtre94270France
| | - Farah Karam
- Faculty of MedicineBalamand UniversityBeirutLebanon
| | | | - Paula El Najjar
- FMPS Holding BIOTECKNO s.a.l. Research and Quality SolutionsNaccashBeirut60 247Lebanon
- Department of Agricultural and Food Engineering, School of EngineeringHoly Spirit University of KaslikJounieh446Lebanon
| | - Charbel Khalil
- Reviva Regenerative Medicine CenterBsalimLebanon
- Bone Marrow Transplant UnitBurjeel Medical CityAbu DhabiUAE
- Lebanese American University School of MedicineBeirutLebanon
| |
Collapse
|
8
|
Zhang T, Luo X, Xu K, Zhong W. Peptide-containing nanoformulations: Skin barrier penetration and activity contribution. Adv Drug Deliv Rev 2023; 203:115139. [PMID: 37951358 DOI: 10.1016/j.addr.2023.115139] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/21/2023] [Accepted: 11/08/2023] [Indexed: 11/14/2023]
Abstract
Transdermal drug delivery presents a less invasive pathway, circumventing the need to pass through the gastrointestinal tract and liver, thereby reducing drug breakdown, initial metabolism, and gastrointestinal discomfort. Nevertheless, the unique composition and dense structure of the stratum corneum present a significant barrier to transdermal delivery. This article presents an overview of the current developments in peptides and nanotechnology to address this challenge. Initially, we sum up peptide-containing nanoformulations for transdermal drug delivery, examining them through the lenses of both inorganic and organic materials. Particular emphasis is placed on the diverse roles that peptides play within these nanoformulations, including conferring functionality upon nanocarriers and enhancing the biological efficacy of drugs. Subsequently, we summarize innovative strategies for enhancing skin penetration, categorizing them into passive and active approaches. Lastly, we discuss the therapeutic potential of peptide-containing nanoformulations in addressing a range of diseases, drawing insights from the biological activities and functions of peptides. Furthermore, the challenges hindering clinical translation are also discussed, providing valuable insights for future advancements in transdermal drug delivery.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Xuan Luo
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Keming Xu
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing 210009, China.
| | - Wenying Zhong
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
9
|
Seemann S, Dubs M, Koczan D, Salapare HS, Ponche A, Pieuchot L, Petithory T, Wartenberg A, Staehlke S, Schnabelrauch M, Anselme K, Nebe JB. Response of Osteoblasts on Amine-Based Nanocoatings Correlates with the Amino Group Density. Molecules 2023; 28:6505. [PMID: 37764281 PMCID: PMC10534789 DOI: 10.3390/molecules28186505] [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] [Revised: 08/30/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Increased life expectancy in industrialized countries is causing an increased incidence of osteoporosis and the need for bioactive bone implants. The integration of implants can be improved physically, but mainly by chemical modifications of the material surface. It was recognized that amino-group-containing coatings improved cell attachment and intracellular signaling. The aim of this study was to determine the role of the amino group density in this positive cell behavior by developing controlled amino-rich nanolayers. This work used covalent grafting of polymer-based nanocoatings with different amino group densities. Titanium coated with the positively-charged trimethoxysilylpropyl modified poly(ethyleneimine) (Ti-TMS-PEI), which mostly improved cell area after 30 min, possessed the highest amino group density with an N/C of 32%. Interestingly, changes in adhesion-related genes on Ti-TMS-PEI could be seen after 4 h. The mRNA microarray data showed a premature transition of the MG-63 cells into the beginning differentiation phase after 24 h indicating Ti-TMS-PEI as a supportive factor for osseointegration. This amino-rich nanolayer also induced higher bovine serum albumin protein adsorption and caused the cells to migrate slower on the surface after a more extended period of cell settlement as an indication of a better surface anchorage. In conclusion, the cell spreading on amine-based nanocoatings correlated well with the amino group density (N/C).
Collapse
Affiliation(s)
- Susanne Seemann
- Institute for Cell Biology, Rostock University Medical Center, 18057 Rostock, Germany (J.B.N.)
| | - Manuela Dubs
- Department of Biomaterials, INNOVENT e.V., 07745 Jena, Germany; (M.D.); (A.W.); (M.S.)
| | - Dirk Koczan
- Department of Immunology, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Hernando S. Salapare
- Institut de Science des Matériaux de Mulhouse (IS2M), CNRS, Université de Haute-Alsace, UMR 7361, 68100 Mulhouse, France (A.P.); (L.P.); (T.P.); (K.A.)
| | - Arnaud Ponche
- Institut de Science des Matériaux de Mulhouse (IS2M), CNRS, Université de Haute-Alsace, UMR 7361, 68100 Mulhouse, France (A.P.); (L.P.); (T.P.); (K.A.)
| | - Laurent Pieuchot
- Institut de Science des Matériaux de Mulhouse (IS2M), CNRS, Université de Haute-Alsace, UMR 7361, 68100 Mulhouse, France (A.P.); (L.P.); (T.P.); (K.A.)
| | - Tatiana Petithory
- Institut de Science des Matériaux de Mulhouse (IS2M), CNRS, Université de Haute-Alsace, UMR 7361, 68100 Mulhouse, France (A.P.); (L.P.); (T.P.); (K.A.)
| | - Annika Wartenberg
- Department of Biomaterials, INNOVENT e.V., 07745 Jena, Germany; (M.D.); (A.W.); (M.S.)
| | - Susanne Staehlke
- Institute for Cell Biology, Rostock University Medical Center, 18057 Rostock, Germany (J.B.N.)
| | | | - Karine Anselme
- Institut de Science des Matériaux de Mulhouse (IS2M), CNRS, Université de Haute-Alsace, UMR 7361, 68100 Mulhouse, France (A.P.); (L.P.); (T.P.); (K.A.)
| | - J. Barbara Nebe
- Institute for Cell Biology, Rostock University Medical Center, 18057 Rostock, Germany (J.B.N.)
- Department Life, Light & Matter, Interdisciplinary Faculty, University of Rostock, 18059 Rostock, Germany
| |
Collapse
|
10
|
Wu R, Prachyathipsakul T, Zhuang J, Liu H, Han Y, Liu B, Gong S, Qiu J, Wong S, Ribbe A, Medeiros J, Bhagabati J, Gao J, Wu P, Dutta R, Herrera R, Faraci S, Xiao H, Thayumanavan S. Conferring liver selectivity to a thyromimetic using a novel nanoparticle increases therapeutic efficacy in a diet-induced obesity animal model. PNAS NEXUS 2023; 2:pgad252. [PMID: 37649581 PMCID: PMC10465086 DOI: 10.1093/pnasnexus/pgad252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 07/21/2023] [Indexed: 09/01/2023]
Abstract
Optimization of metabolic regulation is a promising solution for many pathologies, including obesity, dyslipidemia, type 2 diabetes, and inflammatory liver disease. Synthetic thyroid hormone mimics-based regulation of metabolic balance in the liver showed promise but was hampered by the low biocompatibility and harmful effects on the extrahepatic axis. In this work, we show that specifically directing the thyromimetic to the liver utilizing a nanogel-based carrier substantially increased therapeutic efficacy in a diet-induced obesity mouse model, evidenced by the near-complete reversal of body weight gain, liver weight and inflammation, and cholesterol levels with no alteration in the thyroxine (T4) / thyroid stimulating hormone (TSH) axis. Mechanistically, the drug acts by binding to thyroid hormone receptor β (TRβ), a ligand-inducible transcription factor that interacts with thyroid hormone response elements and modulates target gene expression. The reverse cholesterol transport (RCT) pathway is specifically implicated in the observed therapeutic effect. Overall, the study demonstrates a unique approach to restoring metabolic regulation impacting obesity and related metabolic dysfunctions.
Collapse
Affiliation(s)
- Ruiling Wu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Theeraphop Prachyathipsakul
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jiaming Zhuang
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Hongxu Liu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Yanhui Han
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Bin Liu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Shuai Gong
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jingyi Qiu
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA
| | - Siu Wong
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Alexander Ribbe
- Department of Polymer Science and Engineering, University of Massachusetts, Amherst, MA 01003, USA
| | - Jewel Medeiros
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jayashree Bhagabati
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jingjing Gao
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Peidong Wu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Ranit Dutta
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | | | | | - Hang Xiao
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| | - S Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
11
|
Carroll E, Kunte N, McGraw E, Gautam S, Range R, Noveron-Nunez JA, Held DW, Avila LA. Gene silencing in adult Popillia japonica through feeding of double-stranded RNA (dsRNA) complexed with branched amphiphilic peptide capsules (BAPCs). FRONTIERS IN INSECT SCIENCE 2023; 3:1151789. [PMID: 38469482 PMCID: PMC10926504 DOI: 10.3389/finsc.2023.1151789] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/28/2023] [Indexed: 03/13/2024]
Abstract
Gene silencing by feeding double-stranded (dsRNA) holds promise as a novel pest management strategy. Nonetheless, degradation of dsRNA in the environment and within the insect gut, as well as inefficient systemic delivery are major limitations to applying this strategy. Branched amphiphilic peptide capsules (BAPCs) complexed with dsRNA have been used to successfully target genes outside and inside the gut epithelium upon ingestion. This suggests that BAPCs can protect dsRNA from degradation in the gut environment and successfully shuttle it across gut epithelium. In this study, our objectives were to 1) Determine whether feeding on BAPC-dsRNA complexes targeting a putative peritrophin gene of P. japonica would result in the suppression of gut peritrophin synthesis, and 2) gain insight into the cellular uptake mechanisms and transport of BAPC-dsRNA complexes across the larval midgut of P. japonica. Our results suggest that BAPC-dsRNA complexes are readily taken up by the midgut epithelium, and treatment of the tissue with endocytosis inhibitors effectively suppresses intracellular transport. Further, assessment of gene expression in BAPC- peritrophin dsRNA fed beetles demonstrated significant downregulation in mRNA levels relative to control and/or dsRNA alone. Our results demonstrated that BAPCs increase the efficacy of gene knockdown relative to dsRNA alone in P. japonica adults. To our knowledge, this is the first report on nanoparticle-mediated dsRNA delivery through feeding in P. japonica.
Collapse
Affiliation(s)
- Elijah Carroll
- Department of Entomology and Plant Pathology, Auburn University, Auburn, AL, United States
| | - Nitish Kunte
- Department of Biological Sciences, Auburn University, Auburn, AL, United States
| | - Erin McGraw
- Department of Biological Sciences, Auburn University, Auburn, AL, United States
| | - Sujan Gautam
- Department of Biological Sciences, Auburn University, Auburn, AL, United States
| | - Ryan Range
- Department of Biological Sciences, Auburn University, Auburn, AL, United States
| | | | - David W. Held
- Department of Biological Sciences, Auburn University, Auburn, AL, United States
| | - L. Adriana Avila
- Department of Entomology and Plant Pathology, Auburn University, Auburn, AL, United States
| |
Collapse
|
12
|
Ajayi TO, Liu S, Rosen C, Rinaldi-Ramos CM, Allen KD, Sharma B. Application of magnetic particle imaging to evaluate nanoparticle fate in rodent joints. J Control Release 2023; 356:347-359. [PMID: 36868518 PMCID: PMC11565467 DOI: 10.1016/j.jconrel.2023.02.038] [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: 10/20/2022] [Revised: 02/16/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023]
Abstract
Nanoparticles are a promising approach for improving intra-articular drug delivery and tissue targeting. However, techniques to non-invasively track and quantify their concentration in vivo are limited, resulting in an inadequate understanding of their retention, clearance, and biodistribution in the joint. Currently, fluorescence imaging is often used to track nanoparticle fate in animal models; however, this approach has limitations that impede long-term quantitative assessment of nanoparticles over time. The goal of this work was to evaluate an emerging imaging modality, magnetic particle imaging (MPI), for intra-articular tracking of nanoparticles. MPI provides 3D visualization and depth-independent quantification of superparamagnetic iron oxide nanoparticle (SPION) tracers. Here, we developed and characterized a polymer-based magnetic nanoparticle system incorporated with SPION tracers and cartilage targeting properties. MPI was then used to longitudinally assess nanoparticle fate after intra-articular injection. Magnetic nanoparticles were injected into the joints of healthy mice, and evaluated for nanoparticle retention, biodistribution, and clearance over 6 weeks using MPI. In parallel, the fate of fluorescently tagged nanoparticles was tracked using in vivo fluorescence imaging. The study was concluded at day 42, and MPI and fluorescence imaging demonstrated different profiles in nanoparticle retention and clearance from the joint. MPI signal was persistent over the study duration, suggesting NP retention of at least 42 days, much longer than the 14 days observed based on fluorescence signal. These data suggest that the type of tracer - SPIONs or fluorophores - and modality of imaging can affect interpretation of nanoparticle fate in the joint. Given that understanding particle fate over time is paramount for attaining insights about therapeutic profiles in vivo, our data suggest MPI may yield a quantitative and robust method to non-invasively track nanoparticles following intra-articular injection on an extended timeline.
Collapse
Affiliation(s)
- Tolulope O Ajayi
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Sitong Liu
- Department of Chemical Engineering, University of Florida, Gainesville, FL, USA
| | - Chelsea Rosen
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Carlos M Rinaldi-Ramos
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; Department of Chemical Engineering, University of Florida, Gainesville, FL, USA
| | - Kyle D Allen
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Blanka Sharma
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
13
|
Ruijter N, Soeteman-Hernández LG, Carrière M, Boyles M, McLean P, Catalán J, Katsumiti A, Cabellos J, Delpivo C, Sánchez Jiménez A, Candalija A, Rodríguez-Llopis I, Vázquez-Campos S, Cassee FR, Braakhuis H. The State of the Art and Challenges of In Vitro Methods for Human Hazard Assessment of Nanomaterials in the Context of Safe-by-Design. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:472. [PMID: 36770432 PMCID: PMC9920318 DOI: 10.3390/nano13030472] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 06/18/2023]
Abstract
The Safe-by-Design (SbD) concept aims to facilitate the development of safer materials/products, safer production, and safer use and end-of-life by performing timely SbD interventions to reduce hazard, exposure, or both. Early hazard screening is a crucial first step in this process. In this review, for the first time, commonly used in vitro assays are evaluated for their suitability for SbD hazard testing of nanomaterials (NMs). The goal of SbD hazard testing is identifying hazard warnings in the early stages of innovation. For this purpose, assays should be simple, cost-effective, predictive, robust, and compatible. For several toxicological endpoints, there are indications that commonly used in vitro assays are able to predict hazard warnings. In addition to the evaluation of assays, this review provides insights into the effects of the choice of cell type, exposure and dispersion protocol, and the (in)accurate determination of dose delivered to cells on predictivity. Furthermore, compatibility of assays with challenging advanced materials and NMs released from nano-enabled products (NEPs) during the lifecycle is assessed, as these aspects are crucial for SbD hazard testing. To conclude, hazard screening of NMs is complex and joint efforts between innovators, scientists, and regulators are needed to further improve SbD hazard testing.
Collapse
Affiliation(s)
- Nienke Ruijter
- National Institute for Public Health & the Environment (RIVM), 3721 MA Bilthoven, The Netherlands
| | | | - Marie Carrière
- Univ. Grenoble-Alpes, CEA, CNRS, SyMMES-CIBEST, 17 rue des Martyrs, 38000 Grenoble, France
| | - Matthew Boyles
- Institute of Occupational Medicine (IOM), Edinburgh EH14 4AP, UK
| | - Polly McLean
- Institute of Occupational Medicine (IOM), Edinburgh EH14 4AP, UK
| | - Julia Catalán
- Finnish Institute of Occupational Health, 00250 Helsinki, Finland
- Department of Anatomy, Embryology and Genetics, University of Zaragoza, 50013 Zaragoza, Spain
| | - Alberto Katsumiti
- GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), 48170 Zamudio, Spain
| | | | | | | | | | - Isabel Rodríguez-Llopis
- GAIKER Technology Centre, Basque Research and Technology Alliance (BRTA), 48170 Zamudio, Spain
| | | | - Flemming R. Cassee
- National Institute for Public Health & the Environment (RIVM), 3721 MA Bilthoven, The Netherlands
- Institute for Risk Assessment Sciences (IRAS), Utrecht University, 3584 CS Utrecht, The Netherlands
| | - Hedwig Braakhuis
- National Institute for Public Health & the Environment (RIVM), 3721 MA Bilthoven, The Netherlands
| |
Collapse
|
14
|
Lei F, Cai J, Lyu R, Shen H, Xu Y, Wang J, Shuai Y, Xu Z, Mao C, Yang M. Efficient Tumor Immunotherapy through a Single Injection of Injectable Antigen/Adjuvant-Loaded Macroporous Silk Fibroin Microspheres. ACS APPLIED MATERIALS & INTERFACES 2022; 14:42950-42962. [PMID: 36112417 DOI: 10.1021/acsami.2c11286] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Synthetic or natural materials have been used as vaccines in cancer immunotherapy. However, using them as vaccines necessitates multiple injections or surgical implantations. To tackle such daunting challenges, we develop an injectable macroporous Bombyx mori (B. mori) silk fibroin (SF) microsphere loaded with antigens and immune adjuvants to suppress established tumors with only a single injection. SF microspheres can serve as a scaffold by injection and avoid surgical injury as seen in traditional scaffold vaccines. The macroporous structure of the vaccine facilitates the recruitment of immune cells and promotes the activation of dendritic cells (DCs), resulting in a favorable immune microenvironment that further induces strong humoral and cellular immunity. We have also modified the vaccine into a booster version by simply allowing the antigens to be adsorbed onto the SF microspheres. The booster vaccine highly efficiently suppresses tumor growth by improving the cytotoxic T lymphocyte (CTL) response. In general, these results demonstrate that the macroporous SF microspheres can serve as a facile platform for tumor vaccine therapy in the future. Since the SF microspheres are also potential scaffolds for tissue regeneration, their use as a vaccine platform will enable their applications in eradicating tumors while regenerating healthy tissue to heal the tumor-site cavity.
Collapse
Affiliation(s)
- Fang Lei
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, Zhejiang 310058, P.R. China
| | - Jiangfeng Cai
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, Zhejiang 310058, P.R. China
| | - Ruyin Lyu
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, Zhejiang 310058, P.R. China
| | - Honglan Shen
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, Zhejiang 310058, P.R. China
| | - Yalan Xu
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, Zhejiang 310058, P.R. China
| | - Jie Wang
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, Zhejiang 310058, P.R. China
| | - Yajun Shuai
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, Zhejiang 310058, P.R. China
| | - Zongpu Xu
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, Zhejiang 310058, P.R. China
| | - Chuanbin Mao
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, P.R. China
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Mingying Yang
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, Zhejiang 310058, P.R. China
| |
Collapse
|
15
|
Hausig-Punke F, Richter F, Hoernke M, Brendel JC, Traeger A. Tracking the Endosomal Escape: A Closer Look at Calcein and Related Reporters. Macromol Biosci 2022; 22:e2200167. [PMID: 35933579 DOI: 10.1002/mabi.202200167] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/19/2022] [Indexed: 11/11/2022]
Abstract
Crossing the cellular membrane and delivering active pharmaceuticals or biologicals into the cytosol of cells is an essential step in the development of nanomedicines. One of the most important intracellular processes regarding the cellular uptake of biologicals is the endolysosomal pathway. Sophisticated nanocarriers have been developed overcoming a major hurdle, the endosomal entrapment, and delivering their cargo to the required site of action. In parallel, in vitro assays have been established analyzing the performance of these nanocarriers. Among them, the release of the membrane-impermeable dye calcein has become a popular and straightforward method. It is accessible for most researchers worldwide, allows for rapid conclusions about the release potential, and enables the study of release mechanisms. This review is intended to provide an overview and guidance for scientists applying the calcein release assay. It comprises a survey of several applications in the study of endosomal escape, considerations of potential pitfalls, challenges and limitations of the assay, and a brief summary of complementary methods. Based on this review, we hope to encourage further research groups to take advantage of the calcein release assay for their own purposes and help to create a database for more efficient cross-correlations between nanocarriers. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Franziska Hausig-Punke
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany.,Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Friederike Richter
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany.,Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Maria Hoernke
- Chemistry and Pharmacy, Albert-Ludwigs-Universität Freiburg, Hermann-Herder-Str. 9, 79104, Freiburg i.Br., Germany
| | - Johannes C Brendel
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany.,Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Anja Traeger
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany.,Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| |
Collapse
|
16
|
Colapicchioni V, Millozzi F, Parolini O, Palacios D. Nanomedicine, a valuable tool for skeletal muscle disorders: Challenges, promises, and limitations. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1777. [PMID: 35092179 PMCID: PMC9285803 DOI: 10.1002/wnan.1777] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/24/2021] [Accepted: 01/06/2022] [Indexed: 12/15/2022]
Abstract
Muscular dystrophies are a group of rare genetic disorders characterized by progressive muscle weakness, which, in the most severe forms, leads to the patient's death due to cardiorespiratory problems. There is still no cure available for these diseases and significant effort is being placed into developing new strategies to either correct the genetic defect or to compensate muscle loss by stimulating skeletal muscle regeneration. However, the vast anatomical extension of the target tissue poses great challenges to these goals, highlighting the need for complementary strategies. Nanomedicine is an actively evolving field that merges nanotechnologies with biomedical and pharmaceutical sciences. It holds great potential in regenerative medicine, both in supporting tissue engineering and regeneration, and in optimizing drug and oligonucleotide delivery and gene therapy strategies. In this review, we will summarize the state‐of‐the‐art in the field of nanomedicine applied to skeletal muscle regeneration. We will discuss the recent work toward the development of nanopatterned scaffolds for tissue engineering, the efforts in the synthesis of organic and inorganic nanoparticles for gene therapy and drug delivery applications, as well as their use as immune modulators. Although nanomedicine holds great promise for muscle and other degenerative diseases, many challenges still need to be systematically addressed to assure a smooth transition from the bench to the bedside. This article is categorized under:Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement
Collapse
Affiliation(s)
- Valentina Colapicchioni
- Italian National Research Council, Institute for Atmospheric Pollution Research (CNR-IIA), Rome, Italy.,Mhetra LLC, Miami, Florida, USA
| | - Francesco Millozzi
- Histology and Embryology Unit, DAHFMO, Sapienza University, Rome, Italy.,IRCCS Santa Lucia Foundation, Rome, Italy
| | - Ornella Parolini
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy.,IRCCS Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Daniela Palacios
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy.,IRCCS Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
17
|
Xie B, Du K, Huang F, Lin Z, Wu L. Cationic Nanomaterials for Autoimmune Diseases Therapy. Front Pharmacol 2022; 12:762362. [PMID: 35126109 PMCID: PMC8813968 DOI: 10.3389/fphar.2021.762362] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 12/30/2021] [Indexed: 01/14/2023] Open
Abstract
Cationic nanomaterials are defined as nanoscale structures smaller than 100 nm bearing positive charges. They have been investigated to apply to many aspects including clinical diagnosis, gene delivery, drug delivery, and tissue engineering for years. Recently, a novel concept has been made to use cationic nanomaterials as cell-free nucleic acid scavengers and inhibits the inflammatory responses in autoimmune diseases. Here, we highlighted different types of cationic materials which have the potential for autoimmune disease treatment and reviewed the strategy for autoimmune diseases therapy based on cationic nanoparticles. This review will also demonstrate the challenges and possible solutions that are encountered during the development of cationic materials-based therapeutics for autoimmune diseases.
Collapse
Affiliation(s)
- Baozhao Xie
- Division of Rheumatology, Department of Internal Medicine, the 7th Affiliated Hospital, Guang Xi Medical University, Wuzhou, China
| | - Keqian Du
- Department of Rheumatology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Fujian Huang
- Center for Chemical Biology and Drug Discovery, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhiming Lin
- Department of Rheumatology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Linping Wu
- Center for Chemical Biology and Drug Discovery, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
18
|
Noureddine A, Paffett ML, Franco S, Chan AE, Pallikkuth S, Lidke K, Serda RE. Endolysosomal Mesoporous Silica Nanoparticle Trafficking along Microtubular Highways. Pharmaceutics 2021; 14:pharmaceutics14010056. [PMID: 35056951 PMCID: PMC8781846 DOI: 10.3390/pharmaceutics14010056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/19/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
This study examines intra- and intercellular trafficking of mesoporous silica nanoparticles along microtubular highways, with an emphasis on intercellular bridges connecting interphase and telophase cells. The study of nanoparticle trafficking within and between cells during all phases of the cell cycle is relevant to payload destination and dilution, and impacts delivery of therapeutic or diagnostic agents. Super-resolution stochastic optical reconstruction and sub-airy unit image acquisition, the latter combined with Huygens deconvolution microscopy, enable single nanoparticle and microtubule resolution. Combined structural and functional data provide enhanced details on biological processes, with an example of mitotic inheritance during cancer cell trivision.
Collapse
Affiliation(s)
- Achraf Noureddine
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM 87131, USA;
| | - Michael L. Paffett
- Fluorescence Microscopy Shared Resource, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA;
| | - Stefan Franco
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, USA; (S.F.); (A.E.C.)
| | - Alfonso E. Chan
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, USA; (S.F.); (A.E.C.)
| | - Sandeep Pallikkuth
- Physics and Astronomy, University of New Mexico, Albuquerque, NM 87131, USA; (S.P.); (K.L.)
| | - Keith Lidke
- Physics and Astronomy, University of New Mexico, Albuquerque, NM 87131, USA; (S.P.); (K.L.)
| | - Rita E. Serda
- Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM 87131, USA;
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, USA; (S.F.); (A.E.C.)
- Correspondence: ; Tel.: +1-505-272-7698
| |
Collapse
|
19
|
Cancer vaccines from cryogenically silicified tumour cells functionalized with pathogen-associated molecular patterns. Nat Biomed Eng 2021; 6:19-31. [PMID: 34725505 DOI: 10.1038/s41551-021-00795-w] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 08/16/2021] [Indexed: 12/20/2022]
Abstract
The production of personalized cancer vaccines made from autologous tumour cells could benefit from mechanisms that enhance immunogenicity. Here we show that cancer vaccines can be made via the cryogenic silicification of tumour cells, which preserves tumour antigens within nanoscopic layers of silica, followed by the decoration of the silicified surface with pathogen-associated molecular patterns. These pathogen-mimicking cells activate dendritic cells and enhance the internalization, processing and presentation of tumour antigens to T cells. In syngeneic mice with high-grade ovarian cancer, a cell-line-based silicified cancer vaccine supported the polarization of CD4+ T cells towards the T-helper-1 phenotype in the tumour microenvironment, and induced tumour-antigen-specific T-cell immunity, resulting in complete tumour eradication and in long-term animal survival. In the setting of established disease and a suppressive tumour microenvironment, the vaccine synergized with cisplatin. Silicified and surface-modified cells from tumour samples are amenable to dehydration and room-temperature storage without loss of efficacy and may be conducive to making individualized cancer vaccines across tumour types.
Collapse
|
20
|
Tang H, Li Q, Yan W, Jiang X. Reversing the Chirality of Surface Ligands Can Improve the Biosafety and Pharmacokinetics of Cationic Gold Nanoclusters. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202101609] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Hao Tang
- Department of Biomedical Engineering Southern University of Science and Technology No. 1088 Xueyuan Rd, Nanshan District Shenzhen Guangdong 518055 P. R. China
| | - Qizhen Li
- Department of Biomedical Engineering Southern University of Science and Technology No. 1088 Xueyuan Rd, Nanshan District Shenzhen Guangdong 518055 P. R. China
| | - Weixiao Yan
- Department of Biomedical Engineering Southern University of Science and Technology No. 1088 Xueyuan Rd, Nanshan District Shenzhen Guangdong 518055 P. R. China
| | - Xingyu Jiang
- Department of Biomedical Engineering Southern University of Science and Technology No. 1088 Xueyuan Rd, Nanshan District Shenzhen Guangdong 518055 P. R. China
| |
Collapse
|
21
|
Tang H, Li Q, Yan W, Jiang X. Reversing the Chirality of Surface Ligands Can Improve the Biosafety and Pharmacokinetics of Cationic Gold Nanoclusters. Angew Chem Int Ed Engl 2021; 60:13829-13834. [PMID: 33755292 DOI: 10.1002/anie.202101609] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Indexed: 12/12/2022]
Abstract
Severe toxicity and rapid in vivo clearance of cationic nanomaterials seriously hinder their clinical translation. Present strategies to improve the biosafety and in vivo performance of cationic nanomaterials require neutralization of positive charge, which often compromises their efficacy. Herein, we report that substituting L-glutathione (L-GSH) on cationic gold nanoclusters (GNCs) with its D-counterpart can effectively improve the biosafety and pharmacokinetics. Compared with L-GNCs, D-GNCs do not exhibit cellular cytotoxicity, hemolysis, or acute damage to organs. Cationic D-GNCs show less cell internalization than L-GNCs, and do not induce cellular apoptosis. In vivo, the chirality of surface ligands distinctly affects the pharmacokinetics and tumor targeting abilities of D-/L-GNCs. D-GNCs show higher extended circulation time in blood plasma compared to similarly-sized and poly (ethylene glycol)-modified gold nanoparticles. This work demonstrates that the choice of chirality of surface ligands can determine toxicities and pharmacokinetics of cationic nanomaterials.
Collapse
Affiliation(s)
- Hao Tang
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd, Nanshan District, Shenzhen, Guangdong, 518055, P. R. China
| | - Qizhen Li
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd, Nanshan District, Shenzhen, Guangdong, 518055, P. R. China
| | - Weixiao Yan
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd, Nanshan District, Shenzhen, Guangdong, 518055, P. R. China
| | - Xingyu Jiang
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Rd, Nanshan District, Shenzhen, Guangdong, 518055, P. R. China
| |
Collapse
|
22
|
Zahmatkesh E, Ghanian MH, Zarkesh I, Farzaneh Z, Halvaei M, Heydari Z, Moeinvaziri F, Othman A, Ruoß M, Piryaei A, Gramignoli R, Yakhkeshi S, Nüssler A, Najimi M, Baharvand H, Vosough M. Tissue-Specific Microparticles Improve Organoid Microenvironment for Efficient Maturation of Pluripotent Stem-Cell-Derived Hepatocytes. Cells 2021; 10:1274. [PMID: 34063948 PMCID: PMC8224093 DOI: 10.3390/cells10061274] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/16/2022] Open
Abstract
Liver organoids (LOs) are receiving considerable attention for their potential use in drug screening, disease modeling, and transplantable constructs. Hepatocytes, as the key component of LOs, are isolated from the liver or differentiated from pluripotent stem cells (PSCs). PSC-derived hepatocytes are preferable because of their availability and scalability. However, efficient maturation of the PSC-derived hepatocytes towards functional units in LOs remains a challenging subject. The incorporation of cell-sized microparticles (MPs) derived from liver extracellular matrix (ECM), could provide an enriched tissue-specific microenvironment for further maturation of hepatocytes inside the LOs. In the present study, the MPs were fabricated by chemical cross-linking of a water-in-oil dispersion of digested decellularized sheep liver. These MPs were mixed with human PSC-derived hepatic endoderm, human umbilical vein endothelial cells, and mesenchymal stromal cells to produce homogenous bioengineered LOs (BLOs). This approach led to the improvement of hepatocyte-like cells in terms of gene expression and function, CYP activities, albumin secretion, and metabolism of xenobiotics. The intraperitoneal transplantation of BLOs in an acute liver injury mouse model led to an enhancement in survival rate. Furthermore, efficient hepatic maturation was demonstrated after ex ovo transplantation. In conclusion, the incorporation of cell-sized tissue-specific MPs in BLOs improved the maturation of human PSC-derived hepatocyte-like cells compared to LOs. This approach provides a versatile strategy to produce functional organoids from different tissues and offers a novel tool for biomedical applications.
Collapse
Affiliation(s)
- Ensieh Zahmatkesh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (E.Z.); (Z.F.); (Z.H.); (F.M.); (S.Y.)
- Department of Developmental Biology, University of Science and Culture, Tehran 1665659911, Iran
| | - Mohammad Hossein Ghanian
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (M.H.G.); (I.Z.); (M.H.)
| | - Ibrahim Zarkesh
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (M.H.G.); (I.Z.); (M.H.)
| | - Zahra Farzaneh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (E.Z.); (Z.F.); (Z.H.); (F.M.); (S.Y.)
| | - Majid Halvaei
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (M.H.G.); (I.Z.); (M.H.)
| | - Zahra Heydari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (E.Z.); (Z.F.); (Z.H.); (F.M.); (S.Y.)
- Department of Developmental Biology, University of Science and Culture, Tehran 1665659911, Iran
| | - Farideh Moeinvaziri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (E.Z.); (Z.F.); (Z.H.); (F.M.); (S.Y.)
- Department of Developmental Biology, University of Science and Culture, Tehran 1665659911, Iran
| | - Amnah Othman
- Department of Traumatology, Siegfried Weller Institute, University of Tübingen, 72076 Tübingen, Germany; (A.O.); (M.R.); (A.N.)
| | - Marc Ruoß
- Department of Traumatology, Siegfried Weller Institute, University of Tübingen, 72076 Tübingen, Germany; (A.O.); (M.R.); (A.N.)
| | - Abbas Piryaei
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran;
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran
| | - Roberto Gramignoli
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, 17177 Stockholm, Sweden;
| | - Saeed Yakhkeshi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (E.Z.); (Z.F.); (Z.H.); (F.M.); (S.Y.)
| | - Andreas Nüssler
- Department of Traumatology, Siegfried Weller Institute, University of Tübingen, 72076 Tübingen, Germany; (A.O.); (M.R.); (A.N.)
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental & Clinical Research, Université Catholique de Louvain, B-1200 Brussels, Belgium
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (E.Z.); (Z.F.); (Z.H.); (F.M.); (S.Y.)
- Department of Developmental Biology, University of Science and Culture, Tehran 1665659911, Iran
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (E.Z.); (Z.F.); (Z.H.); (F.M.); (S.Y.)
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran
| |
Collapse
|
23
|
Yamada Y, Fichman G, Schneider JP. Serum Protein Adsorption Modulates the Toxicity of Highly Positively Charged Hydrogel Surfaces. ACS APPLIED MATERIALS & INTERFACES 2021; 13:8006-8014. [PMID: 33590757 PMCID: PMC9169696 DOI: 10.1021/acsami.0c21596] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Hydrogels formed from peptide self-assembly are a class of materials that are being explored for their utility in tissue engineering, drug and cell delivery, two- and three-dimensional cell culture, and as adjuvants in surgical procedures. Most self-assembled peptide gels can be syringe-injected in vivo to facilitate the local delivery of payloads, including cells, directly to the targeted tissue. Herein, we report that highly positively charged peptide gels are inherently toxic to cells, which would seem to limit their utility. However, adding media containing fetal bovine serum, a common culture supplement, directly transforms these toxic gels into cytocompatible materials capable of sustaining cell viability even in the absence of added nutrients. Multistage mass spectrometry showed that at least 40 serum proteins can absorb to a gel's surface through electrostatic attraction ameliorating its toxicity. Further, cell-based studies employing model gels having only bovine serum albumin, fetuin-A, or vitronectin absorbed to the gel surface showed that single protein additives can also be effective depending on the identity of the cell line. Separate studies employing these model gels showed that the mechanism(s) responsible for mitigating apoptosis involve both the pacification of gel surface charge and adsorbed protein-mediated cell signaling events that activate both the PI3/Akt and MAPK/ERK pathways which are known to facilitate resistance to stress-induced apoptosis and overall cell survival.
Collapse
Affiliation(s)
- Yuji Yamada
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Galit Fichman
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Joel P Schneider
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, National Institutes of Health, Frederick, Maryland 21702, United States
| |
Collapse
|
24
|
Nabil G, Alzhrani R, Alsaab HO, Atef M, Sau S, Iyer AK, Banna HE. CD44 Targeted Nanomaterials for Treatment of Triple-Negative Breast Cancer. Cancers (Basel) 2021; 13:cancers13040898. [PMID: 33672756 PMCID: PMC7924562 DOI: 10.3390/cancers13040898] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/15/2021] [Accepted: 02/11/2021] [Indexed: 12/12/2022] Open
Abstract
Identified as the second leading cause of cancer-related deaths among American women after lung cancer, breast cancer of all types has been the focus of numerous research studies. Even though triple-negative breast cancer (TNBC) represents 15-20% of the number of breast cancer cases worldwide, its existing therapeutic options are fairly limited. Due to the pivotal role of the presence/absence of specific receptors to luminal A, luminal B, HER-2+, and TNBC in the molecular classification of breast cancer, the lack of these receptors has accounted for the aforementioned limitation. Thereupon, in an attempt to participate in the ongoing research endeavors to overcome such a limitation, the conducted study adopts a combination strategy as a therapeutic paradigm for TNBC, which has proven notable results with respect to both: improving patient outcomes and survivability rates. The study hinges upon an investigation of a promising NPs platform for CD44 mediated theranostic that can be combined with JAK/STAT inhibitors for the treatment of TNBC. The ability of momelotinib (MMB), which is a JAK/STAT inhibitor, to sensitize the TNBC to apoptosis inducer (CFM-4.16) has been evaluated in MDA-MB-231 and MDA-MB-468. MMB + CFM-4.16 combination with a combination index (CI) ≤0.5, has been selected for in vitro and in vivo studies. MMB has been combined with CD44 directed polymeric nanoparticles (PNPs) loaded with CFM-4.16, namely CD44-T-PNPs, which selectively delivered the payload to CD44 overexpressing TNBC with a significant decrease in cell viability associated with a high dose reduction index (DRI). The mechanism underlying their synergism is based on the simultaneous downregulation of P-STAT3 and the up-regulation of CARP-1, which has induced ROS-dependent apoptosis leading to caspase 3/7 elevation, cell shrinkage, DNA damage, and suppressed migration. CD44-T-PNPs showed a remarkable cellular internalization, demonstrated by uptake of a Rhodamine B dye in vitro and S0456 (NIR dye) in vivo. S0456 was conjugated to PNPs to form CD44-T-PNPs/S0456 that simultaneously delivered CFM-4.16 and S0456 parenterally with selective tumor targeting, prolonged circulation, minimized off-target distribution.
Collapse
Affiliation(s)
- Ghazal Nabil
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt; (G.N.); (M.A.)
- Use-Inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA; (R.A.); (H.O.A.); (S.S.)
| | - Rami Alzhrani
- Use-Inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA; (R.A.); (H.O.A.); (S.S.)
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, P.O. Box 11099, Taif 21944, Egypt
| | - Hashem O. Alsaab
- Use-Inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA; (R.A.); (H.O.A.); (S.S.)
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, P.O. Box 11099, Taif 21944, Egypt
| | - Mohammed Atef
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt; (G.N.); (M.A.)
| | - Samaresh Sau
- Use-Inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA; (R.A.); (H.O.A.); (S.S.)
| | - Arun K. Iyer
- Use-Inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA; (R.A.); (H.O.A.); (S.S.)
- Molecular Imaging Program, Barbara Ann Karmanos Cancer Institute, Wayne State University, School of Medicine, Detroit, MI 48201, USA
- Correspondence: (A.K.I.); (H.E.B.); Tel.: +1-3135775875 (A.K.I.); +2-01004552557 (H.E.B.)
| | - Hossny El Banna
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt; (G.N.); (M.A.)
- Correspondence: (A.K.I.); (H.E.B.); Tel.: +1-3135775875 (A.K.I.); +2-01004552557 (H.E.B.)
| |
Collapse
|
25
|
Wang Z, Tang M. Research progress on toxicity, function, and mechanism of metal oxide nanoparticles on vascular endothelial cells. J Appl Toxicol 2020; 41:683-700. [DOI: 10.1002/jat.4121] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/05/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Zhihui Wang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health Southeast University Nanjing China
| | - Meng Tang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health Southeast University Nanjing China
| |
Collapse
|
26
|
Wright L, Joyce P, Barnes TJ, Lundmark R, Bergström CAS, Hubert M, Prestidge CA. A Comparison of Chitosan, Mesoporous Silica and Poly(lactic-co-glycolic) Acid Nanocarriers for Optimising Intestinal Uptake of Oral Protein Therapeutics. J Pharm Sci 2020; 110:217-227. [PMID: 32979363 DOI: 10.1016/j.xphs.2020.09.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/17/2020] [Indexed: 12/26/2022]
Abstract
Efficacious oral delivery of therapeutic proteins remains challenging and nanoparticulate approaches are gaining interest for enhancing their permeability. In this study, we explore the ability for three comparably sized nanocarriers, with diverse physicochemical properties [i.e., chitosan (CSNP), mesoporous silica nanoparticles (MSNP) and poly(lactic-co-glycolic) acid (PLGA-NP)], to successfully facilitate epithelial uptake of a model protein, ovalbumin (OVA). We report the effect of nanoparticle surface chemistry and nanostructure on protein release, cell toxicity and the uptake mechanism in a Madin Darby Canine Kidney (MDCK) cell model of the intestinal epithelium. All nanocarriers exhibited bi-phasic OVA release kinetics with sustained and incomplete release after 4 days, and more pronounced release from MSNP than either polymeric nanocarriers. CSNP and MSNP displayed the highest cellular uptake, however CSNP was prone to significant dose-dependent toxicity attributed to the cationic surface charge. Approximately 25% of MSNP uptake was governed by a clathrin-independent endocytic mechanism, while CSNP and PLGA-NP uptake was not controlled via any endocytic mechanisms investigated herein. Furthermore, endosomal localisation was observed for CSNP and MSNP, but not for PLGA-NP. These findings may assist in the optimal choice and engineering of nanocarriers for specific intestinal permeation enhancement for oral protein delivery.
Collapse
Affiliation(s)
- Leah Wright
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, Australia; ARC Centre of Excellence in Bio-Nano Science, Adelaide, Australia
| | - Paul Joyce
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, Australia; ARC Centre of Excellence in Bio-Nano Science, Adelaide, Australia
| | - Timothy J Barnes
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Richard Lundmark
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Christel A S Bergström
- Department of Pharmacy, Uppsala University, Uppsala, Sweden; The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Madlen Hubert
- Department of Pharmacy, Uppsala University, Uppsala, Sweden.
| | - Clive A Prestidge
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, Australia; ARC Centre of Excellence in Bio-Nano Science, Adelaide, Australia.
| |
Collapse
|
27
|
Kelly IB, Fletcher RB, McBride JR, Weiss SM, Duvall CL. Tuning Composition of Polymer and Porous Silicon Composite Nanoparticles for Early Endosome Escape of Anti-microRNA Peptide Nucleic Acids. ACS APPLIED MATERIALS & INTERFACES 2020; 12:39602-39611. [PMID: 32805967 PMCID: PMC8356247 DOI: 10.1021/acsami.0c05827] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Porous silicon nanoparticles (PSNPs) offer tunable pore structure and easily modified surface chemistry, enabling high loading capacity for drugs with diverse chemicophysical properties. While PSNPs are also cytocompatible and degradable, PSNP integration into composite structures can be a useful approach to enhance carrier colloidal stability, drug-cargo loading stability, and endosome escape. Here, we explored PSNP polymer composites formed by coating of oxidized PSNPs with a series of poly[ethylene glycol-block-(dimethylaminoethyl methacrylate-co-butyl methacrylate)] (PEG-DB) diblock copolymers with varied molar ratios of dimethylaminoethyl methacrylate (D) and butyl methacrylate (B) in the random copolymer block. We screened and developed PSNP composites specifically toward intracellular delivery of microRNA inhibitory peptide nucleic acids (PNA). While a copolymer with 50 mol % B (50B) is optimal for early endosome escape in free polymer form, its pH switch was suppressed when it was formed into 50B polymer-coated PSNP composites (50BCs). We demonstrate that a lower mol % B (30BC) is the ideal PEG-DB composition for PSNP/PEG-DB nanocomposites based on having both the highest endosome disruption potential and miR-122 inhibitory activity. At a 1 mM PNA dose, 30BCs facilitated more potent inhibition of miR-122 in comparison to 40BC (p = 0.0095), 50BC (p < 0.0001), or an anti-miR-122 oligonucleotide delivered with the commercial transfection reagent Fugene 6. Using a live cell galectin 8-based endosome disruption reporter, 30BCs had greater endosomal escape than 40BCs and 50BCs within 2 h after treatment, suggesting that rapid endosome escape correlates with higher intracellular bioactivity. This study provides new insight on the polymer structure-dependent effects on stability, endosome escape, and cargo intracellular bioavailability for endosomolytic polymer-coated PSNPs.
Collapse
Affiliation(s)
- Isom B Kelly
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - R Brock Fletcher
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - James R McBride
- Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Sharon M Weiss
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| |
Collapse
|
28
|
Liu J, Miao L, Sui J, Hao Y, Huang G. Nanoparticle cancer vaccines: Design considerations and recent advances. Asian J Pharm Sci 2020; 15:576-590. [PMID: 33193861 PMCID: PMC7610208 DOI: 10.1016/j.ajps.2019.10.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 09/15/2019] [Accepted: 10/14/2019] [Indexed: 12/30/2022] Open
Abstract
Vaccines therapeutics manipulate host's immune system and have broad potential for cancer prevention and treatment. However, due to poor immunogenicity and limited safety, fewer cancer vaccines have been successful in clinical trials. Over the past decades, nanotechnology has been exploited to deliver cancer vaccines, eliciting long-lasting and effective immune responses. Compared to traditional vaccines, cancer vaccines delivered by nanomaterials can be tuned towards desired immune profiles by (1) optimizing the physicochemical properties of the nanomaterial carriers, (2) modifying the nanomaterials with targeting molecules, or (3) co-encapsulating with immunostimulators. In order to develop vaccines with desired immunogenicity, a thorough understanding of parameters that affect immune responses is required. Herein, we discussed the effects of physicochemical properties on antigen presentation and immune response, including but not limited to size, particle rigidity, intrinsic immunogenicity. Furthermore, we provided a detailed overview of recent preclinical and clinical advances in nanotechnology for cancer vaccines, and considerations for future directions in advancing the vaccine platform to widespread anti-cancer applications.
Collapse
Affiliation(s)
- Jingjing Liu
- The School of Pharmaceutical Sciences, Shandong University, Ji'nan 250012, China
| | - Lei Miao
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge MA 02139, USA
| | - Jiying Sui
- Affiliated Hospital of Shandong Academy of Medical Sciences, Ji'nan 250012, China
| | - Yanyun Hao
- The School of Pharmaceutical Sciences, Shandong University, Ji'nan 250012, China
| | - Guihua Huang
- The School of Pharmaceutical Sciences, Shandong University, Ji'nan 250012, China
| |
Collapse
|
29
|
Brown SB, Wang L, Jungels RR, Sharma B. Effects of cartilage-targeting moieties on nanoparticle biodistribution in healthy and osteoarthritic joints. Acta Biomater 2020; 101:469-483. [PMID: 31586725 DOI: 10.1016/j.actbio.2019.10.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/11/2019] [Accepted: 10/02/2019] [Indexed: 12/22/2022]
Abstract
Understanding intra-articular biodistribution is imperative as candidate osteoarthritis (OA) drugs become increasingly site-specific. Cartilage has been identified as opportunistic for therapeutic intervention, but poses numerous barriers to drug delivery. To facilitate drug delivery to cartilage, nanoscale vehicles have been designed with different features that target the tissue's matrix. However, it is unclear if these targeting strategies are influenced by OA and the associated structural changes that occur in cartilage. The goal of this work was to study the effectiveness of different cartilage-targeting nanomaterials with respect to cartilage localization and retention, and to determine how these outcomes change in OA. To address these questions, a nanoparticle (NP) system was developed, and the formulation was tuned to possess three distinct cartilage-targeting strategies: (1) passive targeting cationic NPs for electrostatic attraction to cartilage, (2) active targeting NPs with binding peptides for collagen type II, and (3) untargeted neutrally-charged NPs. Ex vivo analyses with bovine cartilage explants demonstrated that targeting strategies significantly improved NP associations with both healthy and OA-like cartilage. In vivo studies with collagenase-induced OA in rats revealed that disease state influenced joint biodistribution for all three NP formulations. Importantly, the extent of cartilage accumulation for each NP system was affected by disease differently; with active NPs, but not passive NPs, cartilage accumulation was increased in OA relative to healthy knees. Together, this work suggests that NPs can be strategically designed for site-specific OA drug delivery, but the biodistribution of the NPs are influenced by the disease conditions into which they are delivered. STATEMENT OF SIGNIFICANCE: As emerging drugs for osteoarthritis are becoming increasingly site-specific, the need for targeted intra-articular drug delivery has evolved. To improve drug delivery to cartilage, targeting strategies for nanomaterials have been developed, but the manner in which these targeted systems accumulate at different sites within the joint remains poorly understood. Moreover, it is unclear how nanomaterial-tissue interactions change in osteoarthritic conditions, as tissue structure and composition change after disease onset. By understanding how nanomaterials distribute within healthy and disease joints, we can advance targeted drug delivery strategies and improve therapeutic outcomes for emerging drugs.
Collapse
|
30
|
Chen D, Parayath N, Ganesh S, Wang W, Amiji M. The role of apolipoprotein- and vitronectin-enriched protein corona on lipid nanoparticles for in vivo targeted delivery and transfection of oligonucleotides in murine tumor models. NANOSCALE 2019; 11:18806-18824. [PMID: 31595922 DOI: 10.1039/c9nr05788a] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The application of lipid-based nanoparticle (LNP) delivery systems remains a popular strategy for the systemic delivery of gene therapies to specific disease targets, including solid tumors. It is now well acknowledged that upon systemic administration, biomolecules from blood will adsorb onto nanoparticles' surfaces, forming a "protein corona", affording nanoparticles a "biological identity" on top of their "synthetic identity". Detailed analysis of nanoparticle protein corona is gradually revealing the "missing link" between nanoparticle chemical properties and the biological identity. Nevertheless, the discovery of nanoparticle protein corona's impact on tumor delivery is limited. In this study, we demonstrate that protein corona can be manipulated by formulation composition and particle surface charge changes, and a single lipid switch could switch the nanoparticle protein corona profile. The protein corona composition differences had a profound impact on cell transfection, in vivo biodistribution as well as tumor-specific delivery efficiency. Nanoparticles with apolipoprotein-rich corona showed better delivery to hepatocellular carcinoma (HepG2) as compared to those with vitronectin-rich corona. In addition, we found that, the PEG conjugated lipid chain length and PEG amount in LNPs were key factors to consider in successful RNA interference therapy for solid tumors.
Collapse
Affiliation(s)
- Dongyu Chen
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouve College of Health Sciences, Northeastern University, Boston, MA 02115, USA.
| | - Neha Parayath
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouve College of Health Sciences, Northeastern University, Boston, MA 02115, USA.
| | | | - Weimin Wang
- Dicerna Pharmaceuticals, Cambridge, MA 02140, USA
| | - Mansoor Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouve College of Health Sciences, Northeastern University, Boston, MA 02115, USA. and Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
31
|
Au JLS, Lu Z, Abbiati RA, Wientjes MG. Systemic Bioequivalence Is Unlikely to Equal Target Site Bioequivalence for Nanotechnology Oncologic Products. AAPS J 2019; 21:24. [PMID: 30710324 PMCID: PMC6432930 DOI: 10.1208/s12248-019-0296-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/09/2019] [Indexed: 11/30/2022] Open
Abstract
Approval of generic drugs by the US Food and Drug Administration (FDA) requires the product to be pharmaceutically equivalent to the reference listed drug (RLD) and demonstrate bioequivalence (BE) in effectiveness when administered to patients under the conditions in the RLD product labeling. Effectiveness is determined by drug exposure at the target sites. However, since such measurement is usually unavailable, systemic exposure is assumed to equal target site exposure and systemic BE to equal target site BE. This assumption, while it often applies to small molecule drug products that are readily dissolved in biological fluids and systemically absorbed, is unlikely to apply to nanotechnology products (NP) that exist as heterogeneous systems and are subjected to dimension- and material-dependent changes. This commentary provides an overview of the intersecting and spatial-dependent processes and variables governing the delivery and residence of oncologic NP in solid tumors. In order to provide a quantitative perspective of the collective effects of these processes, we used quantitative systems pharmacology (QSP) multi-scale modeling to capture the physicochemical and biological events on several scales (whole-body, organ/suborgan, cell/subcellular, spatial locations, time). QSP is an emerging field that entails using modeling and computation to facilitate drug development; an analogous approach (i.e., model-informed drug development) is advocated by to FDA. The QSP model-based simulations illustrated that small changes in NP attributes (e.g., size variations during manufacturing, interactions with proteins in biological milieu) could lead to disproportionately large differences in target site exposure, rending systemic BE unlikely to equal target site BE.
Collapse
Affiliation(s)
- Jessie L-S Au
- Institute of Quantitative Systems Pharmacology, 1815 Aston Avenue, suite 107, Carlsbad, California, 92008, USA.
- Optimum Therapeutics LLC, Carlsbad, California, 92008, USA.
- Department of Pharmaceutical Sciences, University of Oklahoma, Oklahoma City, Oklahoma, 73117, USA.
- College of Pharmacy, Taipei Medical University, Taipei, Taiwan, Republic of China.
| | - Ze Lu
- Institute of Quantitative Systems Pharmacology, 1815 Aston Avenue, suite 107, Carlsbad, California, 92008, USA
- Optimum Therapeutics LLC, Carlsbad, California, 92008, USA
| | - Roberto A Abbiati
- Institute of Quantitative Systems Pharmacology, 1815 Aston Avenue, suite 107, Carlsbad, California, 92008, USA
- Department of Pharmaceutical Sciences, University of Oklahoma, Oklahoma City, Oklahoma, 73117, USA
| | - M Guillaume Wientjes
- Institute of Quantitative Systems Pharmacology, 1815 Aston Avenue, suite 107, Carlsbad, California, 92008, USA
- Optimum Therapeutics LLC, Carlsbad, California, 92008, USA
| |
Collapse
|
32
|
Zhou X, Hao Y, Yuan L, Pradhan S, Shrestha K, Pradhan O, Liu H, Li W. Nano-formulations for transdermal drug delivery: A review. CHINESE CHEM LETT 2018. [DOI: 10.1016/j.cclet.2018.10.037] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
33
|
Abstract
The field of nanotechnology has grown exponentially during the last few decades, due in part to the use of nanoparticles in many manufacturing processes, as well as their potential as clinical agents for treatment of diseases and for drug delivery. This has created several new avenues by which humans can be exposed to nanoparticles. Unfortunately, investigations assessing the toxicological impacts of nanoparticles (i.e. nanotoxicity), as well as their possible risks to human health and the environment, have not kept pace with the rapid rise in their use. This has created a gap-in-knowledge and a substantial need for more research. Studies are needed to help complete our understanding of the mechanisms of toxicity of nanoparticles, as well as the mechanisms mediating their distribution and accumulation in cells and tissues and their elimination from the body. This review summarizes our knowledge on nanoparticles, including their various applications, routes of exposure, their potential toxicity and risks to human health.
Collapse
|
34
|
Dong X, Liang J, Yang A, Wang C, Kong D, Lv F. In Vivo Imaging Tracking and Immune Responses to Nanovaccines Involving Combined Antigen Nanoparticles with a Programmed Delivery. ACS APPLIED MATERIALS & INTERFACES 2018; 10:21861-21875. [PMID: 29901978 DOI: 10.1021/acsami.8b04867] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Combined nanovaccine can generate robust and persistent antigen-specific immune responses. A combined nanovaccine was developed based on antigen-loaded genipin-cross-linked-polyethyleneimine-antigen nanoparticles and in vivo multispectral fluorescence imaging tracked the antigen delivery of combined nanovaccine. The inner layer antigen nanoparticles carried abundant antigens by self-cross-linking for persistent immune response, whereas the outer antigen on the surface of antigen nanoparticles provided the initial antigen exposure. The delivery of combined nanovaccine was tracked dynamically and objectively by the separation of inner genipin cross-linked antigen nanoparticle and the outer fluorescent antigen. The immune responses of the combined nanovaccine were evaluated including antigen-specific CD4+ and CD8+ T-cell responses, IgG antibody level, immunological memory, and CD8+ cytotoxic T lymphocyte responses. The results indicated that the inner and outer antigens of combined vaccine can be tracked in real time with a programmed delivery by the dual fluorescence imaging. The programmed delivery of the inner and outer antigens induced strong immune responses with a combination of a quick delivery and a persistent delivery. With adequate antigen exposure, the dendritic cells were effectively activated and matured, and following T cells were further activated for immune response. Compared with a single nanoparticle formulation, the combined nanovaccine exactly elicited a stronger antigen-specific immune response.
Collapse
Affiliation(s)
- Xia Dong
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering , Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , P. R. China
| | - Jie Liang
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering , Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , P. R. China
| | - Afeng Yang
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering , Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , P. R. China
| | - Chun Wang
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering , Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , P. R. China
- Department of Biomedical Engineering , University of Minnesota , Minneapolis , Minnesota 55455 , United States
| | - Deling Kong
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering , Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , P. R. China
| | - Feng Lv
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering , Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , P. R. China
| |
Collapse
|
35
|
Feng Q, Liu Y, Huang J, Chen K, Huang J, Xiao K. Uptake, distribution, clearance, and toxicity of iron oxide nanoparticles with different sizes and coatings. Sci Rep 2018; 8:2082. [PMID: 29391477 PMCID: PMC5794763 DOI: 10.1038/s41598-018-19628-z] [Citation(s) in RCA: 383] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 01/05/2018] [Indexed: 02/05/2023] Open
Abstract
Iron oxide nanoparticles (IONPs) have been increasingly used in biomedical applications, but the comprehensive understanding of their interactions with biological systems is relatively limited. In this study, we systematically investigated the in vitro cell uptake, cytotoxicity, in vivo distribution, clearance and toxicity of commercially available and well-characterized IONPs with different sizes and coatings. Polyethylenimine (PEI)-coated IONPs exhibited significantly higher uptake than PEGylated ones in both macrophages and cancer cells, and caused severe cytotoxicity through multiple mechanisms such as ROS production and apoptosis. 10 nm PEGylated IONPs showed higher cellular uptake than 30 nm ones, and were slightly cytotoxic only at high concentrations. Interestingly, PEGylated IONPs but not PEI-coated IONPs were able to induce autophagy, which may play a protective role against the cytotoxicity of IONPs. Biodistribution studies demonstrated that all the IONPs tended to distribute in the liver and spleen, and the biodegradation and clearance of PEGylated IONPs in these tissues were relatively slow (>2 weeks). Among them, 10 nm PEGylated IONPs achieved the highest tumor uptake. No obvious toxicity was found for PEGylated IONPs in BALB/c mice, whereas PEI-coated IONPs exhibited dose-dependent lethal toxicity. Therefore, it is crucial to consider the size and coating properties of IONPs in their applications.
Collapse
Affiliation(s)
- Qiyi Feng
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yanping Liu
- Safety Evaluation Center, Sichuan Institute for Food and Drug Control, Chengdu, China
| | - Jian Huang
- Department of Thoracic Surgery, Wuxi People's Hospital of Nanjing Medical University, Wuxi, China.,Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ke Chen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jinxing Huang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Xiao
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China. .,Laboratory of Non-Human Primate Disease Model research, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
36
|
Chen D, Ganesh S, Wang W, Amiji M. Plasma protein adsorption and biological identity of systemically administered nanoparticles. Nanomedicine (Lond) 2017; 12:2113-2135. [DOI: 10.2217/nnm-2017-0178] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although a variety of nanoparticles (NPs) have been used for drug delivery applications, their surfaces are immediately covered by plasma protein corona upon systemic administration. As a result, the adsorbed proteins create a unique biological identity of the NPs that lead to unpredictable performance. The protein corona on NPs could also impede active targeting, induce off-target effects, trigger particle clearance and even provoke toxicity. This article reviews the fundamentals of NP–plasma protein interaction, the consequences of the interactions, and provides insights into the correlations of protein corona with biodistribution and cellular delivery. We hope that this review will trigger additional questions and possible solutions that lead to more favorable developments in NP-based targeted delivery systems.
Collapse
Affiliation(s)
- Dongyu Chen
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA 02115, USA
| | - Shanthi Ganesh
- Department of Pre-Clinical Oncology, Dicerna Pharmaceuticals, Inc., Cambridge, MA 02140, USA
| | - Weimin Wang
- Department of Chemistry and Formulation, Dicerna Pharmaceuticals, Inc., Cambridge, MA 02140, USA
| | - Mansoor Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
37
|
Mackeyev Y, Mark C, Kumar N, Serda RE. The influence of cell and nanoparticle properties on heating and cell death in a radiofrequency field. Acta Biomater 2017; 53:619-630. [PMID: 28179157 DOI: 10.1016/j.actbio.2017.02.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 01/27/2017] [Accepted: 02/02/2017] [Indexed: 02/08/2023]
Abstract
The use of non-invasive radiofrequency (RF) energy to induce mild thermal and non-thermal effects in cancer tissue is under study as an adjuvant to chemo, radio or immuno therapy. This study examines cell specific sensitivities to RF exposure and the potential of nanoparticles to elevate heating rates or enhance biological effects. Increases in the heating rate of water in an RF field operating at 13.56MHz (0.004-0.028°C/s) were positively correlated with concentration of hybrid nanoparticles (1-10mg/ml) consisting of water soluble malonodiserinolamide [60]fullerene (C60-ser) conjugated to the surface of mesoporous silica nanoparticles (SiO2-C60). The heating rate of highly conductive cell culture media (0.024°C/s) was similar to that of the highest concentration of nanoparticles in water, with no significant increase due to addition of nanoparticles at relevant doses (<100μg/ml). With respect to cell viability, anionic (SiO2 and SiO2-C60) or neutral (C60) nanoparticles did not influence RF-induced cell death, however, cationic nanoparticles (4-100μg/ml) caused dose-dependent increases in RF-induced cell death (24-42% compared to RF only). The effect of cell type, size and immortalization on sensitivity of cells to RF fields was examined in endothelial (HUVEC and HMVEC), fibroblast (primary dermal and L939) and cancer cells (HeLa and 4T1). While the state of cellular immortalization itself did not consistently influence the rate of RF-induced cell death compared to normal cell counter parts, cell size (ranging from 7 to 30μm) negatively correlated with cell sensitivity to RF (21-97% cell death following 6min irradiation). In summary, while nanoparticles do not alter the heating rate of biologically-relevant solutions, they can increase RF-induced cell death based on intrinsic cytotoxicity; and cells with smaller radii, and thereby greater surface membrane, are more susceptible to cell damage in an RF field than larger cells. STATEMENT OF SIGNIFICANCE The ability of nanoparticles to either direct heating or increase susceptibility of cancer cells to radiofrequency (RF) energy remains controversial, as is the impact of cell attributes on susceptibility of cells to RF-induced cell death. This manuscript examines the impact of nanoparticle charge, size, and cellular localization on RF-induced cell death and the influence of nanoparticles on the heating rates of water and biologically-relevant media. Susceptibility of immortalized or primary cells to RF energy and the impact of cell size are also examined. The ability to selectively modulate RF heating rates in specific biological locations or in specific cell populations would enhance the therapeutic potential of RF therapy.
Collapse
|