1
|
Bernardelli C, Piergiovanni M, Bianchi E, Carlo-Stella C, Costantino ML, Casagrande G. A Microfluidic Approach for Intracellular Delivery into Red Blood Cells: A Deeper Understanding of the Role of Chemical/Rheological Properties of the Cellular Suspension. Ann Biomed Eng 2025; 53:1128-1137. [PMID: 39971860 PMCID: PMC12006216 DOI: 10.1007/s10439-025-03678-2] [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: 09/20/2024] [Accepted: 01/07/2025] [Indexed: 02/21/2025]
Abstract
Red Blood Cells (RBCs) are a promising drug delivery system candidate for many drugs. Using autologous cells helps to overcome biocompatibility issues, while microfluidics allows accurate control of the intracellular delivery of molecules through fluidic shear stress. With the ultimate goal of exploiting this delivery technique for clinical applications, we investigate how the chemical/rheological characteristics of the suspension and the properties of the RBCs in different animals influence the delivery mechanism. As regard the suspension of RBC, we study the effects induced by the hematocrit and by the presence of proteins such as albumin (Bovine Serum Albumin-BSA). Regarding the cellular properties of RBCs, we aim to investigate the exportability of the technique to the RBC of the most used animal models and identify the most suitable one. The presence of BSA implies a more significant variability of the intracellular delivery. However, 70 ÷ 94% of the cells have successfully encapsulated the probe molecule. Regarding the effect of hematocrit, however, the implementation of the experiment is more challenging due to the increase in viscosity and the easier sedimentation at low flow rates. Evaluation of intracellular delivery in the RBCs of various animal samples has instead led to the proposal of the mouse as the most suitable model for preclinical studies on this particular delivery approach.
Collapse
Affiliation(s)
- Clara Bernardelli
- Laboratory of Biological Structure Mechanics LaBS, Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milano, Italy
- MERYLO' srl, Varedo, MB, Italy
| | - Monica Piergiovanni
- Laboratory of Biological Structure Mechanics LaBS, Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milano, Italy
- MERYLO' srl, Varedo, MB, Italy
| | - Elena Bianchi
- Laboratory of Biological Structure Mechanics LaBS, Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milano, Italy.
- MERYLO' srl, Varedo, MB, Italy.
| | | | - Maria Laura Costantino
- Laboratory of Biological Structure Mechanics LaBS, Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milano, Italy
| | - Giustina Casagrande
- Laboratory of Biological Structure Mechanics LaBS, Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milano, Italy
- MERYLO' srl, Varedo, MB, Italy
- Humanitas University, Milano, Italy
| |
Collapse
|
2
|
Yang S, Zhang X, Wang Y, Liu J, Wang L, Liao Y, Yang Y, Dai T, Yin X, Li S, Han L, Zhu J, Feng H. Mannose-modified erythrocyte membrane-coated Chuanmingshen violaceum polysaccharide PLGA nanoparticles to improve immune responses in mice. Int Immunopharmacol 2025; 152:114450. [PMID: 40080925 DOI: 10.1016/j.intimp.2025.114450] [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: 11/27/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/15/2025]
Abstract
This study developed a poly(lactic-co-glycolic acid) (PLGA) biomimetic nanoparticle (Man-RBC-CVPP) containing Chuanmingshen violaceum polysaccharide (CVP) and coated with a macrophage-targeting mannose receptor (DSPE-PEG-Man) modified RBCM. In vitro experiments demonstrated that Man-RBC-CVPP enhances antigen uptake and immune responses in RAW264.7 cells and can induce an immune response in mouse macrophages by activating the TLR4-mediated NF-κB signaling pathway. In vivo experiments showed that Man-RBC-CVPP promotes the activation of splenic dendritic cells (DCs) by increasing the expression of major histocompatibility complex class II (MHCII), CD80+, and CD86+. Further, it improves the maturation of splenic lymphocytes, increasing the expression of CD4+ and CD8+. It also upregulates the secretion of cytokines, raises serum levels of the specific antibody IgG, and slows the release of OVA at the injection site. In summary, Man-RBC-CVPP can effectively enhance both cellular and humoral immune responses and provide controlled, long-term antigen release.
Collapse
Affiliation(s)
- Shuyao Yang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan 610041, PR China
| | - Xinnan Zhang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan 610041, PR China
| | - Yao Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan 610041, PR China
| | - Jie Liu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan 610041, PR China
| | - Lu Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan 610041, PR China
| | - Yi Liao
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan 610041, PR China
| | - Yanwen Yang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan 610041, PR China
| | - Tao Dai
- Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan 610041, PR China; College of Veterinary Medicine, Southwest University, Rongchang, Chongqing 402460, PR China
| | - Xuemei Yin
- Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan 610041, PR China; College of Pharmacy, Southwest Minzu University, Chengdu, Sichuan 610041, PR China
| | - Shanshan Li
- Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan 610041, PR China; College of Pharmacy, Southwest Minzu University, Chengdu, Sichuan 610041, PR China
| | - Lu Han
- Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan 610041, PR China; College of Pharmacy, Southwest Minzu University, Chengdu, Sichuan 610041, PR China
| | - Jiangjiang Zhu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan 610041, PR China.
| | - Haibo Feng
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
3
|
Della Pelle G, Markelc B, Bozic T, Šribar J, Krizaj I, Zagar Soderznik K, Hudoklin S, Kreft ME, Urbančič I, Kisovec M, Podobnik M, Kostevšek N. Red Blood Cell Membrane Vesicles for siRNA Delivery: A Biocompatible Carrier With Passive Tumor Targeting and Prolonged Plasma Residency. Int J Nanomedicine 2025; 20:3269-3301. [PMID: 40109366 PMCID: PMC11921803 DOI: 10.2147/ijn.s504644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/04/2025] [Indexed: 03/22/2025] Open
Abstract
Background Despite many advances in gene therapy, the delivery of small interfering RNAs is still challenging. Erythrocytes are the most abundant cells in the human body, and their membrane possesses unique features. From them, erythrocytes membrane vesicles can be generated, employable as nano drug delivery system with prolonged blood residence and high biocompatibility. Methods Human erythrocyte ghosts were extruded in the presence of siRNA, and the objects were termed EMVs (erythrocyte membrane vesicles). An ultracentrifugation-based method was applied to select only the densest EMVs, ie, those containing siRNA. We evaluated their activity in vitro in B16F10 cells expressing fluorescent tdTomato and in vivo in B16F10 tumor-bearing mice after a single injection. Results The EMVs had a negative zeta potential, a particle size of 170 nm and excellent colloidal stability after one month of storage. With 0.3 nM siRNA, more than 75% gene knockdown was achieved in vitro, and 80% was achieved in vivo, at 2 days PI at 2.5 mg/kg. EMVs mostly accumulate around blood vessels in the lungs, brain and tumor. tdTomato fluorescence steadily decreased in tumor areas with higher EMVs concentration, which indicates efficient gene knockdown. Approximately 2% of the initial dose of EMVs was still present in the plasma after 2 days. Conclusion The entire production process of the purified siRNA-EMVs took approximately 4 hours. The erythrocyte marker CD47 offered protection against macrophage recognition in the spleen and in the blood. The excellent biocompatibility and pharmacokinetic properties of these materials make them promising platforms for future improvements, ie, active targeting and codelivery with conventional chemotherapeutics.
Collapse
Affiliation(s)
- Giulia Della Pelle
- Department for Nanostructured Materials, Jožef Stefan Institute, Ljubljana, 1000, Slovenia
- Jožef Stefan International Postgraduate School, Ljubljana, 1000, Slovenia
| | - Bostjan Markelc
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Tim Bozic
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Jernej Šribar
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, 1000, Slovenia
| | - Igor Krizaj
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, 1000, Slovenia
| | | | - Samo Hudoklin
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, 1000, Slovenia
| | - Mateja Erdani Kreft
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, 1000, Slovenia
| | - Iztok Urbančič
- Laboratory of Biophysics, Condensed Matter Physics Department, Jožef Stefan Institute, Ljubljana, 1000, Slovenia
| | - Matic Kisovec
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, 1000, Slovenia
| | - Marjetka Podobnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, 1000, Slovenia
| | - Nina Kostevšek
- Department for Nanostructured Materials, Jožef Stefan Institute, Ljubljana, 1000, Slovenia
| |
Collapse
|
4
|
Miranda M, Brandsma E, Robben L, Van Dender H, van Alphen FPJ, Fijnvandraat K, van den Biggelaar M, Lacroix-Desmazes S, van Bruggen R, Voorberg J. Exploring red blood cells as an antigen delivery system to modulate the immune response towards FVIII in hemophilia A. J Thromb Haemost 2025; 23:836-848. [PMID: 39617188 DOI: 10.1016/j.jtha.2024.11.012] [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: 08/15/2024] [Revised: 10/23/2024] [Accepted: 11/14/2024] [Indexed: 03/10/2025]
Abstract
BACKGROUND The main complication in hemophilia A treatment is the development of inhibitory antibodies against factor (F)VIII. Immune tolerance induction, the gold standard for eradicating anti-FVIII antibodies, is efficient in only 60% to 80% of cases. This underscores the need for more efficient induction of tolerance in patients with hemophilia A with FVIII inhibitors. OBJECTIVES In this study, we explored whether red blood cells (RBCs) can be utilized as antigen delivery system to modulate the immune response against FVIII. METHODS Two promiscuously HLA-DR-presented peptides derived from the A2 and C1 domains of FVIII were fused to the TAT cell-penetrating peptide and incubated with RBCs. RESULTS Biotinylated TAT-A2 and TAT-C1 peptides were found to interact with RBCs as shown by flow cytometry and imaging flow cytometry. Moreover, macrophages efficiently phagocytosed TAT-FVIII peptide-treated RBCs. Using mass spectrometry-based immunopeptidomics we established that TAT-FVIII peptides were presented on major histocompatibility complex class II of macrophages that phagocytosed TAT peptide-pulsed RBCs. Specifically, the TAT-A2 peptide exhibited efficient processing and presentation on HLA-DR molecules. Importantly, incubation of TAT-C1 peptide-treated RBCs-loaded macrophages with a FVIII-specific T-cell hybridoma led to a significant increase in IL-2 production, suggesting functional presentation of TAT-C1-derived peptides by macrophages. CONCLUSION Our findings indicate that RBCs can serve as effective vehicle for the delivery of FVIII-derived peptides to antigen-presenting cells. The successful display of T-cell epitopes on antigen-presenting cell using ex vivo-loaded RBC may be potentially utilized to modulate pathogenic immune responses such as observed in a subset of patients with hemophilia A.
Collapse
Affiliation(s)
- Mariarosaria Miranda
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Eelke Brandsma
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Lotte Robben
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Helena Van Dender
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Floris P J van Alphen
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Karin Fijnvandraat
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands; Department of Pediatric Hematology, Amsterdam University Medical Center Location, University of Amsterdam, Amsterdam, The Netherlands
| | - Maartje van den Biggelaar
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Sebastien Lacroix-Desmazes
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Robin van Bruggen
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Jan Voorberg
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands.
| |
Collapse
|
5
|
Qu Y, Wei R, Cheng S, Liu X, Zhao W, Zhao C. Hirudin-Like Noninvasive Polyurethane Solid Anticoagulant Fabricated by One-Pot In Situ Solution Polymerization and Phase Conversion Methods. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11782-11794. [PMID: 39937629 DOI: 10.1021/acsami.4c20757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2025]
Abstract
In the process of blood purification, the use of anticoagulants is necessary to prevent treatment interruptions due to blood clotting. Currently, traditional liquid anticoagulant medications, which enter the bloodstream, may lead to safety concerns, such as internal bleeding and thrombocytopenia. To address these issues, we prepared solid anticoagulant microspheres based on polyurethane (PU) through one-pot in situ solution polymerization and phase conversion methods. The microspheres prolong the activated partial thromboplastin time (APTT) and thrombin time (TT) by 30-40 s and demonstrate good blood compatibility. Interestingly, we find that polyurethane in the microspheres has the ability to selectively bind thrombin, like the anticoagulant mechanism of hirudin. The strong binding between PU and thrombin is confirmed through microscale thermography (MST) (the dissociation constant Kd is about 71 μM). Furthermore, the anticoagulant microspheres, in combination with other studies of anticoagulation mechanisms, are found to inhibit multiple steps in the coagulation cascade. More importantly, the noninvasive microspheres do not release PU into the bloodstream, ensuring the safety of anticoagulation therapy and suggesting significant potential for application in the field of blood purification.
Collapse
Affiliation(s)
- Yang Qu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China
| | - Ran Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Shengjun Cheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Xianda Liu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Weifeng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Changsheng Zhao
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
- Med-X Center for Materials, Sichuan University, Chengdu 610041, China
| |
Collapse
|
6
|
Weerarathna IN, Kumar P, Dzoagbe HY, Kiwanuka L. Advancements in Micro/Nanorobots in Medicine: Design, Actuation, and Transformative Application. ACS OMEGA 2025; 10:5214-5250. [PMID: 39989765 PMCID: PMC11840590 DOI: 10.1021/acsomega.4c09806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/23/2025] [Accepted: 01/29/2025] [Indexed: 02/25/2025]
Abstract
In light of the ongoing technological transformation, embracing advancements that foster shared benefits is essential. Nanorobots, a breakthrough within nanotechnology, have demonstrated significant potential in fields such as medicine, where diagnostic and therapeutic applications are the primary focus areas. This review provides a comprehensive overview of nanotechnology, robots, and their evolving role in medical applications, particularly highlighting the use of nanorobots. Various design strategies and operational principles, including sensors, actuators, and nanocontrollers, are discussed based on prior research. Key nanorobot medical applications include biomedical imaging, biosensing, minimally invasive surgery, and targeted drug delivery, each utilizing advanced actuation technologies to enhance precision. The paper further examines recent progress in micro/nanorobot actuation and addresses important considerations for the future, including biocompatibility, control, navigation, delivery, targeting, safety, and ethical implications. This review offers a holistic perspective on how nanorobots can reshape medical practices, paving the way for precision medicine and improved patient outcomes.
Collapse
Affiliation(s)
- Induni Nayodhara Weerarathna
- Department
of Biomedical Sciences, Datta Meghe Institute
of Higher Education and Research (Deemed to be University), Wardha, Maharashtra-442001, India
| | - Praveen Kumar
- Department
of Computer Science and Medical Engineering, Datta Meghe Institute of Higher Education and Research (Deemed to
be University), Wardha, Maharashtra-442001, India
| | - Hellen Yayra Dzoagbe
- Datta
Meghe College of Pharmacy, Datta Meghe Institute of Higher Education
and Research, (Deemed to be University), Wardha, Maharashtra-442001, India
| | - Lydia Kiwanuka
- Department
of Medical Radiology and Imaging Technology, Datta Meghe Institute of Higher Education and Research (Deemed to
be University), Wardha, Maharashtra-442001, India
| |
Collapse
|
7
|
Acharya B, McGlade CA, Yin H, Kawano T, Haar L, Mackman N, Sellers RS, Tan X, Bhatt AP, Lawrence DS, Vickerman BM. Photothrombolytics: A light-driven technology for the targeted lysis of thrombi. J Control Release 2025; 378:281-293. [PMID: 39615753 PMCID: PMC11830540 DOI: 10.1016/j.jconrel.2024.11.059] [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: 07/02/2024] [Revised: 11/04/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024]
Abstract
Occlusive blood clots remain a significant global health challenge and result in emergencies that are main causes of death and disability worldwide. Thrombolytic agents (including tissue plasminogen activator, tPA) are the only pharmacological means to dissolve blood clots. However, these drugs have modest efficacy and severe safety concerns persist. We have developed light-responsive tPA-loaded red blood cells (tPA-RBCsPhoto) to target thrombolytic activity at the site of a blood clot. Herein, we describe the use of light to control the release of tPA from engineered RBCs and the subsequent degradation of a blood clot ex vivo. Furthermore, we have employed this technology to restore blood flow to an occluded mouse artery in vivo using a targeted dose that is 25 times lower than conventional systemic tPA treatment.
Collapse
Affiliation(s)
- Basanta Acharya
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Caylie A McGlade
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Haifeng Yin
- McAllister Heart Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Tomohiro Kawano
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Lauren Haar
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Nigel Mackman
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Rani S Sellers
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Xianming Tan
- Department of Biostatistics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Aadra P Bhatt
- Division of Gastroenterology & Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - David S Lawrence
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Brianna M Vickerman
- Eshelman Innovation, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| |
Collapse
|
8
|
Zhang J, Liu J, Ding J, Yu H, Li Z, Chen Y, Lin Y, Niu Y, Lu L, Jin X, Zheng Y. Tris(2-chloroethyl) Phosphate Leads to Unbalanced Circulating Erythrocyte in Mice by Activating both Medullary and Extramedullary Erythropoiesis. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:199-211. [PMID: 39743774 DOI: 10.1021/acs.est.4c09436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Tris(2-chloroethyl) phosphate (TCEP), a prevalent organophosphorus flame retardant, has been identified in various environmental matrices and human blood samples, provoking alarm regarding its hematological toxicity, a subject that has not been thoroughly investigated. Red blood cells (RBCs), or erythrocytes, are the predominant cell type in peripheral blood and are crucial for the maintenance of physiological health. This investigation employed oral gavage to examine the effects of TCEP exposure on erythrocyte counts in mice and to clarify the underlying mechanisms. The results demonstrated a marked increase in circulating RBC counts post-TCEP exposure, concomitantly heightening the risk of polycythemia vera (PV). TCEP exposure stimulated erythropoiesis across all stages of medullary development, including the differentiation of hematopoietic stem cells into erythroid progenitors, the progression of erythrocyte development, and the maturation of erythrocyte. Moreover, TCEP potentiated extramedullary erythropoiesis in the spleen and liver. Subsequent bioinformatics analysis implied that TCEP-induced erythropoiesis was attributed to p53 downregulation. Thus, these findings indicate that TCEP disrupts erythrocyte-mediated hematological homeostasis through the enhancement of both medullary and extramedullary erythropoiesis, leading to the alteration of hematological equilibrium.
Collapse
Affiliation(s)
- Jingxu Zhang
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Jing Liu
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Jian Ding
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Hongyan Yu
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Ziyuan Li
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yidi Chen
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yongfeng Lin
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yong Niu
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Lin Lu
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xiaoting Jin
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| |
Collapse
|
9
|
Wang Y, Liu Y, Su X, Niu L, Li N, Xu C, Sun Z, Guo H, Shen S, Yu M. Non-pathogenic Trojan horse Nissle1917 triggers mitophagy through PINK1/Parkin pathway to discourage colon cancer. Mater Today Bio 2024; 29:101273. [PMID: 39415764 PMCID: PMC11480251 DOI: 10.1016/j.mtbio.2024.101273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
Bacteria-mediated antitumor therapy has gained widespread attention for its innate tumor-targeting capability and excellent immune activation properties. Nevertheless, the clinical approval of bacterial therapies remains elusive primarily due to the formidable challenge of balancing safety with enhancing in vivo efficacy. In this study, leveraging the probiotic Escherichia coli Nissle1917 (EcN) emerges as a promising approach for colon cancer therapy, offering a high level of safety attributed to its lack of virulence factors and its tumor-targeting potential owing to its obligate anaerobic nature. Specifically, we delineate the erythrocyte (RBC) membrane-camouflaged EcN, termed as Trojan horse EcN@RBC, which triggers apoptosis in tumor cells by mitigating mitochondrial membrane potential (MMP) and subsequently activating the PINK1/Parkin pathway associated with mitophagy. Concurrently, the decline in MMP induced by mitophagy disrupts the mitochondrial permeability transition pore (MPTP), leading to the release of Cytochrome C and subsequent apoptosis induction. Moreover, synergistic effects were observed through the combination of the autophagy activator rapamycin, bolstering the antitumor efficacy in vivo. These findings offer novel insights into probiotic-mediated antitumor mechanisms and underscore the therapeutic potential of EcN@RBC for colon cancer patients.
Collapse
Affiliation(s)
- Yang Wang
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Yao Liu
- Clinical Oncology Center, Shanghai Municipal Hospital of TCM, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Xiaomin Su
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Lili Niu
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Nannan Li
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Ce Xu
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Zanya Sun
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Huishu Guo
- Central Laboratory, First Affiliated Hospital, Institute (college) of Integrative Medicine, Dalian Medical University, Dalian, 116011, China
| | - Shun Shen
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Minghua Yu
- Fudan University Clinical Research Center for Cell-based Immunotherapy & Department of Oncology, Fudan University Pudong Medical Center, Shanghai, 201399, China
| |
Collapse
|
10
|
Dubey A, Raju F, Lobo CL, Gs R, Hebbar S, Shetty A, Kumar P, El-Zahaby SA. Formulation and Characterization of RBCS Coated Carboplatin Loaded Nano-Liposomal Formulation for Managing Breast Cancer. Drug Dev Res 2024; 85:e70019. [PMID: 39558836 DOI: 10.1002/ddr.70019] [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: 08/11/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/20/2024]
Abstract
Cell membrane-coated Nano-Liposomes (CM-NLPs) offer a promising approach that combines the advantages of both host cells and synthetic nano-liposomes (NLPs). This technique involves coating liposomes with red blood cell (RBC) membranes to enhance their functionality. In this study, novel carboplatin-loaded NLPs (CP-NLPs) were formulated using phospholipids (Soya Phosphatidyl Choline) and cholesterol through the thin-film hydration method, and optimized using a 32 full factorial design. The optimized CP-NLPs were coated with RBC membranes, resulting in the formulation "CP-RBCs-NLPs." These were characterized for particle size, zeta potential, entrapment efficiency, transmission electron microscopy (TEM), differential scanning calorimetry (DSC), protein content, in vitro drug release, cell viability, and stability. The optimized CP-RBCs-NLPs exhibited a particle size of 103.6 nm, with zeta potential values of -27.3 mV indicating good stability. The entrapment efficiency was approximately 56%, and the drug release profile showed sustained release for up to 8 h. Cytotoxicity studies in human triple-negative breast cancer (MDA-MB468) cell lines demonstrated that CP-RBCs-NLPs effectively delivered the drug into target cells, facilitating cell death due to their bilayer structure similar to cell membranes. Overall, CP-RBCs-NLPs outperformed both carboplatin-loaded conventional NLPs (CP-CNLPs) and carboplatin-conventional solution (CP-CNS), making it a superior formulation for drug delivery.
Collapse
Affiliation(s)
- Akhilesh Dubey
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, Nitte (Deemed to be University), Mangaluru, Karnataka, India
| | - Faby Raju
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, Nitte (Deemed to be University), Mangaluru, Karnataka, India
| | - Cynthia Lizzie Lobo
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, Nitte (Deemed to be University), Mangaluru, Karnataka, India
| | - Ravi Gs
- Formulation and Development, Viatris R&D Centre, Bengaluru, India
| | - Srinivas Hebbar
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Amitha Shetty
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, Nitte (Deemed to be University), Mangaluru, Karnataka, India
| | - Pankaj Kumar
- Department of Pharmaceutical Chemistry, NGSM Institute of Pharmaceutical Sciences, Nitte (Deemed to be University), Mangaluru, Karnataka, India
| | - Sally A El-Zahaby
- Department of Pharmaceutics and Industrial Pharmacy, PharmD Program, Egypt-Japan University of Science and Technology (E-JUST), Alexandria, Egypt
| |
Collapse
|
11
|
Hadi Barhaghtalab R, Tanimowo Aiyelabegan H, Maleki H, Mirzavi F, Gholizadeh Navashenaq J, Abdi F, Ghaffari F, Vakili-Ghartavol R. Recent advances with erythrocytes as therapeutics carriers. Int J Pharm 2024; 665:124658. [PMID: 39236775 DOI: 10.1016/j.ijpharm.2024.124658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/24/2024] [Accepted: 08/31/2024] [Indexed: 09/07/2024]
Abstract
Erythrocytes have gained popularity as a natural option for in vivo drug delivery due to their advantages, which include lengthy circulation times, biocompatibility, and biodegradability. Consequently, the drug's pharmacokinetics and pharmacodynamics in red blood cells can be considerably up the dosage. Here, we provide an overview of the erythrocyte membrane's structure and discuss the characteristics of erythrocytes that influence their suitability as carrier systems. We also cover current developments in the erythrocyte-based nanocarrier, which could be used for both active and passive targeting of disease tissues, particularly those of the reticuloendothelial system (RES) and cancer tissues. We also go over the most recent discoveries about the in vivo and in vitro uses of erythrocytes for medicinal and diagnostic purposes. Moreover, the clinical relevance of erythrocytes is discussed in order to improve comprehension and enable the potential use of erythrocyte carriers in the management of various disorders.
Collapse
Affiliation(s)
| | | | - Hassan Maleki
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farshad Mirzavi
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Fereshteh Abdi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Noncommunicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran
| | - Faezeh Ghaffari
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roghayyeh Vakili-Ghartavol
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
12
|
Wu X, Wang F, Yang X, Gong Y, Niu T, Chu B, Qu Y, Qian Z. Advances in Drug Delivery Systems for the Treatment of Acute Myeloid Leukemia. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403409. [PMID: 38934349 DOI: 10.1002/smll.202403409] [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: 04/27/2024] [Revised: 06/06/2024] [Indexed: 06/28/2024]
Abstract
Acute myeloid leukemia (AML) is a common and catastrophic hematological neoplasm with high mortality rates. Conventional therapies, including chemotherapy, hematopoietic stem cell transplantation (HSCT), immune therapy, and targeted agents, have unsatisfactory outcomes for AML patients due to drug toxicity, off-target effects, drug resistance, drug side effects, and AML relapse and refractoriness. These intrinsic limitations of current treatments have promoted the development and application of nanomedicine for more effective and safer leukemia therapy. In this review, the classification of nanoparticles applied in AML therapy, including liposomes, polymersomes, micelles, dendrimers, and inorganic nanoparticles, is reviewed. In addition, various strategies for enhancing therapeutic targetability in nanomedicine, including the use of conjugating ligands, biomimetic-nanotechnology, and bone marrow targeting, which indicates the potential to reverse drug resistance, are discussed. The application of nanomedicine for assisting immunotherapy is also involved. Finally, the advantages and possible challenges of nanomedicine for the transition from the preclinical phase to the clinical phase are discussed.
Collapse
Affiliation(s)
- Xia Wu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Fangfang Wang
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Xijing Yang
- The Experimental Animal Center of West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Yuping Gong
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Ting Niu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Bingyang Chu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Ying Qu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Zhiyong Qian
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| |
Collapse
|
13
|
Misra R, Sanjana Sharath N. Red blood cells based nanotheranostics: A smart biomimetic approach for fighting against cancer. Int J Pharm 2024; 661:124401. [PMID: 38986966 DOI: 10.1016/j.ijpharm.2024.124401] [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: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
The technique of engineering drug delivery vehicles continues to develop, which bring enhancements in working more efficiently and minimizing side effects to make it more effective and safer. The intense capability of therapeutic agents to remain undamaged in a harsh extracellular environment is helpful to the success of drug development efforts. With this in mind, alterations of biopharmaceuticals with enhanced stability and decreased immunogenicity have been an increasingly active focus of such efforts. Red blood cells (RBCs), also known as erythrocytes have undergone extensive scrutiny as potential vehicles for drug delivery due to their remarkable attributes over the years of research. These include intrinsic biocompatibility, minimal immunogenicity, flexibility, and prolonged systemic circulation. Throughout the course of investigation, a diverse array of drug delivery platforms based on RBCs has emerged. These encompass genetically engineered RBCs, non-genetically modified RBCs, and RBC membrane-coated nanoparticles, each devised to cater to a range of biomedical objectives. Given their prevalence in the circulatory system, RBCs have gained significant attention for their potential to serve as biomimetic coatings for artificial nanocarriers. By virtue of their surface emulation capabilities and customizable core materials, nanocarriers mimicking these RBCs, hold considerable promise across a spectrum of applications, spanning drug delivery, imaging, phototherapy, immunomodulation, sensing, and detection. These multifaceted functionalities underscore the considerable therapeutic and diagnostic potential across various diseases. Our proposed review provides the synthesis of recent strides in the theranostic utilization of erythrocytes in the context of cancer. It also delves into the principal challenges and prospects intrinsic to this realm of research. The focal point of this review pertains to accentuating the significance of erythrocyte-based theranostic systems in combating cancer. Furthermore, it precisely records the latest and the most specific methodologies for tailoring the attributes of these biomimetic nanoscale formulations, attenuating various discoveries for the treatment and management of cancer.
Collapse
Affiliation(s)
- Ranjita Misra
- Department of Biotechnology, Centre for Research in Pure and Applied Sciences, School of Sciences, Jain (Deemed-to-be University), JC Road, Bengaluru 560027, Karnataka, India.
| | - Naomi Sanjana Sharath
- Department of Biotechnology, Centre for Research in Pure and Applied Sciences, School of Sciences, Jain (Deemed-to-be University), JC Road, Bengaluru 560027, Karnataka, India
| |
Collapse
|
14
|
Ban Z, Sun M, Ji H, Ning Q, Cheng C, Shi T, He M, Chen X, Lu H, He X, Guo C, He Y, Shao D, He Y. Immunogenicity-masking delivery of uricase against hyperuricemia and gout. J Control Release 2024; 372:862-873. [PMID: 38906421 DOI: 10.1016/j.jconrel.2024.06.042] [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: 02/19/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
Improving the activity of uricase and lowering its immunogenicity remain significant challenges in the enzyme replacement management of hyperuricemia and related inflammatory diseases. Herein, an immunogenicity-masking strategy based on engineered red blood cells (RBCs) was developed for effective uricase delivery against both hyperuricemia and gout. The dynamic membrane of RBCs enabled high resistance to protease inactivation and hydrogen peroxide accumulation. Benefiting from these advantages, a single infusion of RBC-loaded uricase (Uri@RBC) performed prolonged blood circulation and sustained hyperuricemia management. Importantly, RBCs masked the immunogenicity of uricase, leading to the maintenance of UA-lowering performance after repeated infusion through reduced antibody-mediated macrophage clearance. In an acute gout model, Uri@RBC profoundly alleviated joint edema and inflammation with minimal systemic toxicity. This study supports the employment of immunogenicity-masking tools for efficient and safe enzyme delivery, and this strategy may be leveraged to improve the usefulness of enzyme replacement therapies for managing a wide range of inflammatory diseases.
Collapse
Affiliation(s)
- Zhenglan Ban
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, PR China
| | - Madi Sun
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, PR China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, PR China
| | - Huihong Ji
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, PR China
| | - Quanxin Ning
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, PR China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, PR China
| | - Chuanxu Cheng
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, PR China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, PR China
| | - Tongfei Shi
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, PR China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, PR China
| | - Minghao He
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, PR China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, PR China
| | - Xuenian Chen
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, PR China
| | - Huanfen Lu
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, PR China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, PR China
| | - Xuan He
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, PR China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, PR China
| | - Chenyang Guo
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, PR China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, PR China
| | - Yan He
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, PR China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, PR China.
| | - Dan Shao
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, PR China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, PR China.
| | - Yi He
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, PR China.
| |
Collapse
|
15
|
Islam F, Lewis MR, Craig JD, Leyendecker PM, Deans TL. Advancing in vivo reprogramming with synthetic biology. Curr Opin Biotechnol 2024; 87:103109. [PMID: 38520824 PMCID: PMC11162311 DOI: 10.1016/j.copbio.2024.103109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 03/25/2024]
Abstract
Reprogramming cells will play a fundamental role in shaping the future of cell therapies by developing new strategies to engineer cells for improved performance and higher-order physiological functions. Approaches in synthetic biology harness cells' natural ability to sense diverse signals, integrate environmental inputs to make decisions, and execute complex behaviors based on the health of the organism or tissue. In this review, we highlight strategies in synthetic biology to reprogram cells, and discuss how recent approaches in the delivery of modified mRNA have created new opportunities to alter cell function in vivo. Finally, we discuss how combining concepts from synthetic biology and the delivery of mRNA in vivo could provide a platform for innovation to advance in vivo cellular reprogramming.
Collapse
Affiliation(s)
- Farhana Islam
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Mitchell R Lewis
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - James D Craig
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Peyton M Leyendecker
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Tara L Deans
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
16
|
Anastasiadi AT, Arvaniti VZ, Hudson KE, Kriebardis AG, Stathopoulos C, D’Alessandro A, Spitalnik SL, Tzounakas VL. Exploring unconventional attributes of red blood cells and their potential applications in biomedicine. Protein Cell 2024; 15:315-330. [PMID: 38270470 PMCID: PMC11074998 DOI: 10.1093/procel/pwae001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/08/2024] [Indexed: 01/26/2024] Open
Affiliation(s)
- Alkmini T Anastasiadi
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Vasiliki-Zoi Arvaniti
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Krystalyn E Hudson
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, NY 10032, USA
| | - Anastasios G Kriebardis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Caring Sciences, University of West Attica (UniWA), 12243 Egaleo, Greece
| | | | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, 13001 Aurora, CO, USA
| | - Steven L Spitalnik
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York City, NY 10032, USA
| | - Vassilis L Tzounakas
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece
| |
Collapse
|
17
|
Berikkhanova K, Taigulov E, Bokebaev Z, Kusainov A, Tanysheva G, Yedrissov A, Seredin G, Baltabayeva T, Zhumadilov Z. Drug-loaded erythrocytes: Modern approaches for advanced drug delivery for clinical use. Heliyon 2024; 10:e23451. [PMID: 38192824 PMCID: PMC10772586 DOI: 10.1016/j.heliyon.2023.e23451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/04/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024] Open
Abstract
Scientific organizations worldwide are striving to create drug delivery systems that provide a high local concentration of a drug in pathological tissue without side effects on healthy organs in the body. Important physiological properties of red blood cells (RBCs), such as frequent renewal ability, good oxygen carrying ability, unique shape and membrane flexibility, allow them to be used as natural carriers of drugs in the body. Erythrocyte carriers derived from autologous blood are even more promising drug delivery systems due to their immunogenic compatibility, safety, natural uniqueness, simple preparation, biodegradability and convenience of use in clinical practice. This review is focused on the achievements in the clinical application of targeted drug delivery systems based on osmotic methods of loading RBCs, with an emphasis on advancements in their industrial production. This article describes the basic methods used for encapsulating drugs into erythrocytes, key strategic approaches to the clinical use of drug-loaded erythrocytes obtained by hypotonic hemolysis. Moreover, clinical trials of erythrocyte carriers for the targeted delivery are discussed. This article explores the recent advancements and engineering approaches employed in the encapsulation of erythrocytes through hypotonic hemolysis methods, as well as the most promising inventions in this field. There is currently a shortage of reviews focused on the automation of drug loading into RBCs; therefore, our work fills this gap. Finally, further prospects for the development of engineering and technological solutions for the automatic production of drug-loaded RBCs were studied. Automated devices have the potential to provide the widespread production of RBC-encapsulated therapeutic drugs and optimize the process of targeted drug delivery in the body. Furthermore, they can expedite the widespread introduction of this innovative treatment method into clinical practice, thereby significantly expanding the effectiveness of treatment in both surgery and all areas of medicine.
Collapse
Affiliation(s)
- Kulzhan Berikkhanova
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay Batyr 53, Astana, 010000, Kazakhstan
| | - Erlan Taigulov
- University Medical Center, Nazarbayev University, Astana, 010000, Kazakhstan
- Astana Medical University, Astana, 010000, Kazakhstan
| | - Zhanybek Bokebaev
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay Batyr 53, Astana, 010000, Kazakhstan
- Astana Medical University, Astana, 010000, Kazakhstan
| | - Aidar Kusainov
- Semey State Medical University, Semey, 071400, Kazakhstan
| | | | - Azamat Yedrissov
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay Batyr 53, Astana, 010000, Kazakhstan
| | - German Seredin
- Center for Life Sciences, National Laboratory Astana, Nazarbayev University, Kabanbay Batyr 53, Astana, 010000, Kazakhstan
| | - Tolkyn Baltabayeva
- Scientific-Production Center of Transfusiology, Astana, 010000, Kazakhstan
| | - Zhaxybay Zhumadilov
- Departament of Surgery, School of Medicine, Nazarbayev University, Kabanbay Batyr 53, Astana, 010000, Kazakhstan
| |
Collapse
|
18
|
Kotańska M, Wojtaszek K, Kubacka M, Bednarski M, Nicosia N, Wojnicki M. The Influence of Caramel Carbon Quantum Dots and Caramel on Platelet Aggregation, Protein Glycation and Lipid Peroxidation. Antioxidants (Basel) 2023; 13:13. [PMID: 38275633 PMCID: PMC10812612 DOI: 10.3390/antiox13010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/05/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
Caramel, defined as a coloring agent and as an antioxidant, is used in several kinds of food products and is consumed by many people in different amounts. In our research we showed that the caramelization of sucrose under special conditions leads to the formation of carbon quantum dots (CQDs). So, it makes sense that humans also consume this type of CQDs, and it is theoretically possible for these particles to affect the body. Despite an increasing number of studies describing different types of CQDs, their biosafety is still not clearly understood. In our in vitro research, we examined the effects on platelet aggregation, protein glycation and lipid peroxidation of CQDs and caramel formed from a 20% sucrose solution. In vitro aggregation tests were conducted using freshly collected whole rat blood in a multiplate platelet function analyzer and measurer of electric impedance. The cytotoxic effect of the tested solutions on blood platelets was evaluated based on the release of lactate dehydrogenase. The formation of glycated bovine serum albumin was measured as fluorescence intensity and fructosamine level. The reducing power of the solutions was determined in adipose tissue, and their effect on lipid peroxidation in adipose tissue in vitro was also assessed. By measuring the intensity of hemolysis after incubation in solutions with red blood cell, we assessed their influence on the integration of the red blood cell membrane. All tests were performed in comparison with glucose and fructose and other frequently used sweeteners, such as erythritol and xylitol. Our study showed that caramel and CQDs formed from caramel may influence the glycation process and integrity of the red blood cell membrane, but unlike glucose and fructose, they decrease lipid peroxidation and may reduce Fe (III). Additionally, it is unlikely that they affect platelet aggregation. Compared to glucose and fructose, they may be safer for patients with metabolic disorders; however, further research is needed on the safety and biological activity of caramel and CQD.
Collapse
Affiliation(s)
- Magdalena Kotańska
- Department of Pharmacological Screening, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland;
| | - Konrad Wojtaszek
- Faculty of Non-Ferrous Metals, AGH University of Krakow, Mickiewicza Ave., 30-059 Krakow, Poland;
| | - Monika Kubacka
- Department of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland;
| | - Marek Bednarski
- Department of Pharmacological Screening, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland;
| | - Noemi Nicosia
- PhD Program in Neuroscience, Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
| | - Marek Wojnicki
- Faculty of Non-Ferrous Metals, AGH University of Krakow, Mickiewicza Ave., 30-059 Krakow, Poland;
| |
Collapse
|
19
|
Petrovic SM, Barbinta-Patrascu ME. Organic and Biogenic Nanocarriers as Bio-Friendly Systems for Bioactive Compounds' Delivery: State-of-the Art and Challenges. MATERIALS (BASEL, SWITZERLAND) 2023; 16:7550. [PMID: 38138692 PMCID: PMC10744464 DOI: 10.3390/ma16247550] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023]
Abstract
"Green" strategies to build up novel organic nanocarriers with bioperformance are modern trends in nanotechnology. In this way, the valorization of bio-wastes and the use of living systems to develop multifunctional organic and biogenic nanocarriers (OBNs) have revolutionized the nanotechnological and biomedical fields. This paper is a comprehensive review related to OBNs for bioactives' delivery, providing an overview of the reports on the past two decades. In the first part, several classes of bioactive compounds and their therapeutic role are briefly presented. A broad section is dedicated to the main categories of organic and biogenic nanocarriers. The major challenges regarding the eco-design and the fate of OBNs are suggested to overcome some toxicity-related drawbacks. Future directions and opportunities, and finding "green" solutions for solving the problems related to nanocarriers, are outlined in the final of this paper. We believe that through this review, we will capture the attention of the readers and will open new perspectives for new solutions/ideas for the discovery of more efficient and "green" ways in developing novel bioperformant nanocarriers for transporting bioactive agents.
Collapse
Affiliation(s)
- Sanja M. Petrovic
- Department of Chemical Technologies, Faculty of Technology, University of Nis, Bulevar Oslobodjenja 124, 1600 Leskovac, Serbia;
| | - Marcela-Elisabeta Barbinta-Patrascu
- Department of Electricity, Solid-State Physics and Biophysics, Faculty of Physics, University of Bucharest, 405 Atomistilor Street, P.O. Box MG-11, 077125 Măgurele, Romania
| |
Collapse
|
20
|
De Castro F, Stefàno E, Fanizzi FP, Di Corato R, Abdalla P, Luchetti F, Nasoni MG, Rinaldi R, Magnani M, Benedetti M, Antonelli A. Compatibility of Nucleobases Containing Pt(II) Complexes with Red Blood Cells for Possible Drug Delivery Applications. Molecules 2023; 28:6760. [PMID: 37836603 PMCID: PMC10574024 DOI: 10.3390/molecules28196760] [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: 09/05/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
The therapeutic advantages of some platinum complexes as major anticancer chemotherapeutic agents and of nucleoside analogue-based compounds as essential antiviral/antitumor drugs are widely recognized. Red blood cells (RBCs) offer a potential new strategy for the targeted release of therapeutic agents due to their biocompatibility, which can protect loaded drugs from inactivation in the blood, thus improving biodistribution. In this study, we evaluated the feasibility of loading model nucleobase-containing Pt(II) complexes into human RBCs that were highly stabilized by four N-donors and susceptible to further modification for possible antitumor/antiviral applications. Specifically, platinum-based nucleoside derivatives [PtII(dien)(N7-Guo)]2+, [PtII(dien)(N7-dGuo)]2+, and [PtII(dien)(N7-dGTP)] (dien = diethylenetriamine; Guo = guanosine; dGuo = 2'-deoxy-guanosine; dGTP = 5'-(2'-deoxy)-guanosine-triphosphate) were investigated. These Pt(II) complexes were demonstrated to be stable species suitable for incorporation into RBCs. This result opens avenues for the possible incorporation of other metalated nucleobases analogues, with potential antitumor and/or antiviral activity, into RBCs.
Collapse
Affiliation(s)
- Federica De Castro
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Monteroni, 73100 Lecce, Italy; (F.D.C.); (E.S.)
| | - Erika Stefàno
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Monteroni, 73100 Lecce, Italy; (F.D.C.); (E.S.)
| | - Francesco Paolo Fanizzi
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Monteroni, 73100 Lecce, Italy; (F.D.C.); (E.S.)
| | - Riccardo Di Corato
- Center for Biomolecular Nanotechnologies, Istituto Italiano di Tecnologia, 73010 Arnesano, Italy;
- Institute for Microelectronics and Microsystems (IMM), CNR, Via Monteroni, 73100 Lecce, Italy;
| | - Pasant Abdalla
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino Carlo Bo, Via Saffi 2, 61029 Urbino, Italy; (P.A.); (F.L.); (M.G.N.); (M.M.)
| | - Francesca Luchetti
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino Carlo Bo, Via Saffi 2, 61029 Urbino, Italy; (P.A.); (F.L.); (M.G.N.); (M.M.)
| | - Maria Gemma Nasoni
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino Carlo Bo, Via Saffi 2, 61029 Urbino, Italy; (P.A.); (F.L.); (M.G.N.); (M.M.)
| | - Rosaria Rinaldi
- Institute for Microelectronics and Microsystems (IMM), CNR, Via Monteroni, 73100 Lecce, Italy;
- Mathematics and Physics “E. De Giorgi” Department, University of Salento, Via Monteroni, 73100 Lecce, Italy
| | - Mauro Magnani
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino Carlo Bo, Via Saffi 2, 61029 Urbino, Italy; (P.A.); (F.L.); (M.G.N.); (M.M.)
| | - Michele Benedetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Monteroni, 73100 Lecce, Italy; (F.D.C.); (E.S.)
| | - Antonella Antonelli
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino Carlo Bo, Via Saffi 2, 61029 Urbino, Italy; (P.A.); (F.L.); (M.G.N.); (M.M.)
| |
Collapse
|