1
|
Mou Y, Liu Z, Xu W, Zheng B, Ma M, Qin X, Zheng J, Ni R, Li H, Wang L, Bai Y, Fan J, Qi X, Zhang Q, Zhang P, Zhang D. Dual-source powered sea urchin-like nanomotors for intravesical photothermal therapy of bladder cancer. J Nanobiotechnology 2025; 23:355. [PMID: 40380217 PMCID: PMC12084915 DOI: 10.1186/s12951-025-03446-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 05/05/2025] [Indexed: 05/19/2025] Open
Abstract
Bladder cancer (BCa) ranks as the 9th most prevalent malignancy worldwide, featured by its high risk of recurrence. Intravesical therapy constitutes the most important modality to tackle BCa, but its efficiency is often compromised due to the dense physiological barriers in BCa, the instability of the catalytic environment, and the rapid clearance facilitated by periodic urination. Here, we present a dual-source powered sea urchin-like nanomotor, which feature a gold nanocore decorated with ultrasmall platinum nanoparticles and ureases, enable rapid propulsion through the catalytic conversion of abundant urea and hydrogen peroxide present in the bladder cavity and BCa microenvironment, respectively. Our dual-source powered Au-Pt@ur NPs nanoparticles translocated across the mucus barrier rapidly, deeply penetrated tumor and hence chemo-resected bladder tumors in all cases. These results hold substantial promise for the development of biocompatible nanomotors for improved BCa intravesical therapy.
Collapse
Affiliation(s)
- Yixuan Mou
- Urology & Nephrology Center, Department of Urology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Zhenghong Liu
- Urology & Nephrology Center, Department of Urology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Wentao Xu
- Cancer Center, Department of Interventional Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, People's Republic of China
| | - Bin Zheng
- Urology & Nephrology Center, Department of Urology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China
| | - Minghai Ma
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiaowen Qin
- Urology & Nephrology Center, Department of Urology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Department of Nutrition and Food Hygiene, The National Key Discipline, School of Public Health, Harbin Medical University, Harbin, 150081, P.R. China
| | - Jiajia Zheng
- Cancer Center, Department of Interventional Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, People's Republic of China
| | - Ran Ni
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310000, Zhejiang, China
| | - Haichang Li
- Urology & Nephrology Center, Department of Urology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Lei Wang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yuchen Bai
- Urology & Nephrology Center, Department of Urology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Jinhai Fan
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiaolong Qi
- Urology & Nephrology Center, Department of Urology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Qi Zhang
- Urology & Nephrology Center, Department of Urology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| | - Pu Zhang
- Urology & Nephrology Center, Department of Urology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| | - Dahong Zhang
- Urology & Nephrology Center, Department of Urology, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
2
|
Sun J, Chu R, Wu X, Yu Q, Xiao W, Ao H, Wang Y, Wu T, Ju H, Wu J, Lei J. Anti-biopassivated Reticular Micromotors for Bladder Cancer Therapy. J Am Chem Soc 2025. [PMID: 40378338 DOI: 10.1021/jacs.5c02949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
The limited lifespan of enzyme-powered micro/nanomotors (MNMs) hinders their biomedical applications due to the easy deactivation in tumor microenvironments. In this study, by taking advantage of hydrogen bond-rich metal-organic frameworks (MOFs), we design anti-biopassivated urease-powered MOF motors (Ur-MOFtors) with sustained motility for bladder cancer therapy. Such reticular Ur-MOFtors exhibited an exceptionally long locomotion lifespan exceeding 90 min in highly concentrated urea, which was an 18-fold enhancement compared with urease-adsorbed MOFs, resulting in excellent anti-biopassivation of MOFtors. The underlying molecular mechanism of persistent motion involves hydrogen bonding interaction between the MOF skeleton and the catalytic product, as identified by in situ infrared spectroscopy and density functional theory. Based on the preserved enzymatic activity comparable to native urease, the self-propulsion pathway of Ur-MOFtors is driven by ionic self-diffusiophoresis with the positive chemotactic motion toward urea. Benefiting from the persistent motion of Ur-MOFtors in physiological urea, a rapid bladder cancer therapy was achieved with few instillation sessions and short treatment cycles during intravesical administration. This hydrogen bond-rich framework presents a promising anti-biopassivated approach to overcoming the short lifespan and easy deactivation of enzymatic motors for advanced therapeutic robotics.
Collapse
Affiliation(s)
- Jun Sun
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Ran Chu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan 250021, China
| | - Xiaoqian Wu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Qian Yu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Wencheng Xiao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Hang Ao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yuru Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Taikang Wu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jie Wu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jianping Lei
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
3
|
Ruiz-González N, Sánchez-deAlcázar D, Esporrín-Ubieto D, Di Carlo V, Sánchez S. Hyaluronic Acid-Based Nanomotors: Crossing Mucosal Barriers to Tackle Antimicrobial Resistance. ACS APPLIED MATERIALS & INTERFACES 2025; 17:27988-27999. [PMID: 40298340 PMCID: PMC12086762 DOI: 10.1021/acsami.5c03636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Bacterial infections pose a significant global health challenge aggravated by the rise of antimicrobial resistance (AMR). Among the obstacles preventing effective treatment are biological barriers (BBs) within the body such as the mucus layer. These BBs trap antimicrobials, necessitating higher doses and ultimately accelerating AMR. Addressing this issue requires innovative therapeutic strategies capable of bypassing BBs to deliver drugs more effectively. Here, we present nanomotors (NMs) based on hyaluronic acid (HA)- and urease-nanogels (NGs) as a solution to navigate effectively in viscous media by catalyzing the decomposition of urea into ammonium and carbon dioxide. These HA-based nanomotors (HA-NMs) were loaded with chloramphenicol (CHL) antibiotic and demonstrated superior antimicrobial activity against Escherichia coli(E. coli) compared to mesoporous silica NMs (MSNP-NMs), a reference in the field of NMs. Moreover, using an in vitro transwell model we evaluated the ability of HA-NMs to penetrate mucin barriers, effectively reducing E. coli proliferation, whereas the free antibiotic did not reduce bacteria proliferation. The optical density reduction at 24 h was over ten times greater than with free CHL. These organic-based enzyme-powered NMs represent a significant advancement in drug delivery, offering a promising approach to combat AMR while addressing the challenges of crossing complex BBs.
Collapse
Affiliation(s)
- Noelia Ruiz-González
- The
Barcelona Institute of Science and Technology (BIST), Institute for Bioengineering of Catalonia (IBEC), Baldiri i Reixac 10-12, 08028 Barcelona, Spain
- Facultat
de Física, Universitat de Barcelona
(UB). C. Martí i Franquès, 1-11, 08028 Barcelona, Spain
| | - Daniel Sánchez-deAlcázar
- The
Barcelona Institute of Science and Technology (BIST), Institute for Bioengineering of Catalonia (IBEC), Baldiri i Reixac 10-12, 08028 Barcelona, Spain
| | - David Esporrín-Ubieto
- The
Barcelona Institute of Science and Technology (BIST), Institute for Bioengineering of Catalonia (IBEC), Baldiri i Reixac 10-12, 08028 Barcelona, Spain
| | - Valerio Di Carlo
- The
Barcelona Institute of Science and Technology (BIST), Institute for Bioengineering of Catalonia (IBEC), Baldiri i Reixac 10-12, 08028 Barcelona, Spain
| | - Samuel Sánchez
- The
Barcelona Institute of Science and Technology (BIST), Institute for Bioengineering of Catalonia (IBEC), Baldiri i Reixac 10-12, 08028 Barcelona, Spain
- Catalan
Institution for Research and Advanced Studies (ICREA), Passeig Lluís Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
4
|
Ju X, Javorková E, Michalička J, Pumera M. Single-Atom Colloidal Nanorobotics Enhanced Stem Cell Therapy for Corneal Injury Repair. ACS NANO 2025. [PMID: 40359418 DOI: 10.1021/acsnano.4c18874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Corneal repair using mesenchymal stem cell therapy faces challenges due to long-term cell survival issues. Here, we design cerium oxide with gold single-atom-based nanorobots (CeSAN-bots) for treating corneal damage in a synergistic combination with stem cells. Powered by glucose, CeSAN-bots exhibit enhanced diffusion and active motion due to the cascade reaction catalyzed by gold and cerium oxide. CeSAN-bots demonstrate a two-fold increase in cellular uptake efficiency into mesenchymal stem cells compared to passive uptake. CeSAN-bots possess intrinsic antioxidant and immunomodulatory properties, promoting corneal regeneration. Validation in a mouse corneal alkali burn model reveals an improvement in corneal clarity restoration when stem cells are incorporated with CeSAN-bots. This work presents a strategy for developing glucose-driven, enzyme-free, single-atom-based ultrasmall nanorobots with promising applications in targeted intracellular delivery in diverse biological environments.
Collapse
Affiliation(s)
- Xiaohui Ju
- Future Energy and Innovation Laboratory, Central European Institute of Technology, Brno University of Technology, Purkyňova 123, Brno 61200, Czech Republic
| | - Eliška Javorková
- Department of Toxicology and Molecular Epidemiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, Prague 14200, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, Prague 12844, Czech Republic
| | - Jan Michalička
- Central European Institute of Technology, Brno University of Technology, Purkyňova 123, Brno 61200, Czech Republic
| | - Martin Pumera
- Future Energy and Innovation Laboratory, Central European Institute of Technology, Brno University of Technology, Purkyňova 123, Brno 61200, Czech Republic
- Advanced Nanorobots & Multiscale Robotics Laboratory, Faculty of Electrical Engineering and Computer Science, VSB-Technical University of Ostrava, 17. listopadu 2172/15, Ostrava 70800, Czech Republic
- Department of Medical Research, China Medical University Hospital, China Medical University, No. 91 Hsueh-Shih Road, Taichung 40402, Taiwan
| |
Collapse
|
5
|
Eberhard E, Burger L, Pastrana CL, Seyed-Allaei H, Giunta G, Gerland U. Force Generation by Enhanced Diffusion in Enzyme-Loaded Vesicles. NANO LETTERS 2025; 25:5754-5761. [PMID: 40138661 PMCID: PMC11987064 DOI: 10.1021/acs.nanolett.5c00306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/14/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
The diffusion coefficient of some metabolic enzymes increases with the concentration of their cognate substrate, a phenomenon known as enhanced diffusion. In the presence of substrate gradients, enhanced diffusion induces enzymatic drift, resulting in a nonhomogeneous enzyme distribution. Here, we study the effects of enhanced diffusion on enzyme-loaded vesicles placed in external substrate gradients using a combination of computer simulations and analytical modeling. We observe that the spatially inhomogeneous enzyme profiles generated by enhanced diffusion result in a pressure gradient across the vesicle, which leads to macroscopically observable effects, namely deformation and self-propulsion of the vesicle. Our analytical model allows us to characterize the dependence of the velocity of propulsion on experimentally tunable parameters. The effects predicted by our work provide an avenue for further validation of enhanced diffusion, and might be leveraged for the design of novel synthetic cargo transporters, such as targeted drug delivery systems.
Collapse
Affiliation(s)
| | | | | | - Hamid Seyed-Allaei
- Physics of Complex Biosystems, Department
of Bioscience, School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Giovanni Giunta
- Physics of Complex Biosystems, Department
of Bioscience, School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Ulrich Gerland
- Physics of Complex Biosystems, Department
of Bioscience, School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| |
Collapse
|
6
|
Liu L, Li X, Chen Y, Gao J, Jiang Y, Ye Y, Wang P, Peng F, Tu Y. Gout management using uricase and sodium citrate hollow mesoporous nanomotors. Nat Commun 2025; 16:2339. [PMID: 40057522 PMCID: PMC11890746 DOI: 10.1038/s41467-025-56100-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 01/09/2025] [Indexed: 05/13/2025] Open
Abstract
Uricase (UOx)-based gout treatments are generally limited due in part to the accumulation of H2O2 in an arthrosis environment characterized by a sluggish metabolism. Here we develop a self-cascade nanomotor with high efficiency toward simultaneous uric acid (UA) degradation and H2O2 elimination on the basis of UOx and sodium citrate loading in amine functionalized hollow mesoporous silica nanoparticles (AHMSNs). Due to the inherent asymmetry of AHMSNs, the developed nanomotors can be actuated by ionic diffusiophoresis induced by the enzymatic UA degradation, thus enlarging the diffusion range within the joint cavity. H2O2 generated during UA decomposition can be simultaneously eliminated by the loaded sodium citrate, resulting in a considerable improvement in mammalian cell viability. In vivo studies display powerful therapeutic effect in gouty models using this self-cascade nanomotor system. These results present the potential of nanomotors as active therapeutic agents for gout, providing a perspective on the application of enzyme-driven micro/nanomotors.
Collapse
Affiliation(s)
- Lu Liu
- Pharmacy Department, The Fifth Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xiurong Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Yongxian Chen
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Junbin Gao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Yuejun Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Yicheng Ye
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Peng Wang
- Pharmacy Department, The Fifth Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China.
| | - Fei Peng
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, China.
| | - Yingfeng Tu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Li WW, Yu ZL, Jia J. Urease-powered micro/nanomotors: Current progress and challenges. J Pharm Anal 2025; 15:101095. [PMID: 40177066 PMCID: PMC11964642 DOI: 10.1016/j.jpha.2024.101095] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/26/2024] [Accepted: 09/02/2024] [Indexed: 04/05/2025] Open
Abstract
Enzyme-powered micro/nanomotors (MNMs) (EMNMs) use natural enzymes to facilitate the decomposition of fuels, including hydrogen peroxide (H2O2), glucose, triglycerides, and urea to provide power. EMNMs can achieve self-propulsion through the in situ utilization of biofuels without additional fuels, exhibiting excellent biocompatibility and significant potential for application in the biomedical field. Compared with H2O2, which may cause oxidative damage to the body, urea exhibits superior biosafety characteristics. Presently, urease-powered MNMs (UMNMs) have made notable progress in their applications in the biomedical field and have garnered considerable attention from researchers. In this review, we present the latest advancements in the biomedical field of UMNMs, primarily focusing on: 1) diverse materials used for constructing the fundamental framework of motors; 2) control of motor movement through the regulation of enzymatic reaction rates; and 3) research directions for the clinical application of motors, including in vivo imaging, biomarker detection, cancer treatment, optical therapy, overcoming biological barriers, antibacterial interventions, antithrombotic strategies, and gastric disease management. Despite showing immense potential in biomedical applications, there are still several challenges impeding its practical implementation, such as maintaining activity in the in vivo environment while accurately targeting specific sites to achieve the desired clinical therapeutic effects.
Collapse
Affiliation(s)
- Wen-Wen Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Zi-Li Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Jun Jia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
8
|
Qin YS, Yi J, Chen YJ, Zhang W, Tang SF. Recent Advances in Micro/Nanomotor for the Therapy and Diagnosis of Atherosclerosis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11443-11468. [PMID: 39648908 DOI: 10.1021/acsami.4c15165] [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: 12/10/2024]
Abstract
Atherosclerotic cardiovascular disease poses a significant global public health threat with a high incidence that can result in severe mortality and disability. The lack of targeted effects from traditional therapeutic drugs on atherosclerosis may cause damage to other organs and tissues, necessitating the need for a more focused approach to address this dilemma. Micro/nanomotors are self-propelled micro/nanoscale devices capable of converting external energy into autonomous movement, which offers advantages in enhancing penetration depth and retention while increasing contact area with abnormal sites, such as atherosclerotic plaque, inflammation, and thrombosis, within blood vessel walls. Recent studies have demonstrated the crucial role micro/nanomotors play in treating atherosclerotic cardiovascular disease. Hence, this review highlights the recent progress of micro/nanomotor technology in atherosclerotic cardiovascular disease, including the effective promotion of micro/nanomotors in the circulatory system, overcoming hemorheological barriers, targeting the atherosclerotic plaque microenvironment, and targeting intracellular drug delivery, to facilitate atherosclerotic plaque localization and therapy. Furthermore, we also describe the potential application of micro/nanomotors in the imaging of vulnerable plaque. Finally, we discuss key challenges and prospects for treating atherosclerotic cardiovascular disease while emphasizing the importance of designing individualized management strategies specific to its causes and microenvironmental factors.
Collapse
Affiliation(s)
- Yu-Sheng Qin
- Department of Laboratory Medicine, Liuzhou Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology (Liuzhou People's Hospital), Liuzhou People's Hospital, Liuzhou 545006, China
| | - Juan Yi
- Department of Laboratory Medicine, Liuzhou Traditional Chinese Medical Hospital, The Third Affiliated Hospital of Guangxi University of Chinese Medicine, Liuzhou 545006, China
| | - Yan-Jun Chen
- Department of Pathology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Wei Zhang
- Department of Radiology, Liuzhou People's Hospital, Liuzhou 545006, China
| | - Shi-Fu Tang
- Department of Laboratory Medicine, Liuzhou Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology (Liuzhou People's Hospital), Liuzhou People's Hospital, Liuzhou 545006, China
| |
Collapse
|
9
|
Ivanov NM, Slivkov AI, Huck WTS. A Urease-Based pH Photoswitch: A General Route to Light-to-pH Transduction. Angew Chem Int Ed Engl 2025; 64:e202415614. [PMID: 39263723 PMCID: PMC11735890 DOI: 10.1002/anie.202415614] [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: 08/15/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/13/2024]
Abstract
New approaches for the integration of chemical and physical stimuli to control the dynamics of artificial enzymatic reaction networks (ERNs) are needed. Here, we present a general approach to convert a light stimulus into a time-programmed pH response. We developed and characterized a panel of photoswitchable inhibitors of urease. Urease activity, now regulated by light via the photoinhibitors, leads to an increase in pH upon hydrolysis of urea into ammonia. Careful choice of characteristics of light, and concentrations of enzyme, substrate, and photoinhibitor, allowed us to control the timing of the pH transition. Furthermore, as all enzymes have an activity-pH profile, the urease photoinhibitor system can be used to regulate the activities of other enzymes in small reaction networks.
Collapse
Affiliation(s)
- Nikita M. Ivanov
- Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525AJNijmegenThe Netherlands
| | - Alexandar I. Slivkov
- Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525AJNijmegenThe Netherlands
| | - Wilhelm T. S. Huck
- Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525AJNijmegenThe Netherlands
| |
Collapse
|
10
|
Raj T, Roy S, Kumar A, Roy B, Mani E, Sudhakar S. Direct measurement of self-diffusiophoretic force generated by active colloids of different patch coverage using optical tweezers. J Colloid Interface Sci 2025; 677:986-996. [PMID: 39178677 DOI: 10.1016/j.jcis.2024.07.237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 08/26/2024]
Abstract
HYPOTHESIS Synthetic micro/nanomotors are gaining extensive attention for various biomedical applications (especially in drug delivery) due to their ability to mimic the motion of biological micro/nanoscale swimmers. The feasibility of these applications relies on tight control of propulsion speed, direction, and type of motion (translation, circular, etc.) along with the exerted self-propulsive force. We propose to exploit the variation of both self-propulsion speed and force of active colloids with different patch coverages (with and without supporting layer) for engineering diffusiophoretic micro/nanomotors. EXPERIMENTS The microswimmers were designed at various patch coverages (10°, 30°, and 90°) with (Ti/Pt) and without (Pt) an adhesion layer for the catalytic patch through glancing angle metal deposition (GLAD) technique. Mean-square displacement (MSD) analysis was performed to obtain the self-propulsion parameters like speed and angular speed. Using optical tweezers (OT), the self-propulsive force was measured from the force power spectral density. FINDINGS The findings of our experiments suggest the non-requirement of any adhesion layer preceding the catalyst deposition since the Pt 10° colloidal batch had the maximal self-propulsion speed (4.61±0.3μm/s) and force (345±57fN) for 5% w/v H2O2 fuel concentration. Moreover, the self-propulsion speed and force decreased with increasing patch size, contrary to theoretical estimates. Also, the self-propulsive force obtained from MSD is 2 to 4 times lower in magnitude than the OT based force values. We believe that the self-propelling motion of the micromotors is possibly hindered due to interactions with the surface of the quartz cuvette during the optical microscopic analysis. Further, the MSD is limited to the self-propulsive motion in two dimensions. On the other hand, OT based force measurement involve trapping the particles in the bulk of the solution entirely avoiding the particle-substrate interactions. Hence, OT based force measurements are better than the propulsion velocity based stokes drag force estimates. We believe that this study can lay the foundation in designing efficient micro/nanomotors for translational biomedical applications.
Collapse
Affiliation(s)
- Thilak Raj
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai 600036, India.
| | - Srestha Roy
- Department of Physics, Indian Institute of Technology Madras, Chennai 600036, India.
| | - Ashwin Kumar
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai 600036, India.
| | - Basudev Roy
- Department of Physics, Indian Institute of Technology Madras, Chennai 600036, India.
| | - Ethayaraja Mani
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai 600036, India; Centre for Soft and Biological Matter, Indian Institute of Technology Madras, Chennai 600036, India.
| | - Swathi Sudhakar
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai 600036, India; Centre for Soft and Biological Matter, Indian Institute of Technology Madras, Chennai 600036, India.
| |
Collapse
|
11
|
He T, Yang Y, Chen X. A Lifetime of Catalytic Micro-/Nanomotors. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 15:13. [PMID: 39791773 PMCID: PMC11723389 DOI: 10.3390/nano15010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
Microscopic and nanoscopic motors, often referred to as micro-/nanomotors, are autonomous devices capable of converting chemical energy from their surroundings into mechanical motion or forces necessary for propulsion. These devices draw inspiration from natural biomolecular motor proteins, and in recent years, synthetic micro-/nanomotors have attracted significant attention. Among these, catalytic micro-/nanomotors have emerged as a prominent area of research. Despite considerable progress in their design and functionality, several obstacles remain, especially regarding the development of biocompatible materials and fuels, the integration of intelligent control systems, and the translation of these motors into practical applications. Thus, a comprehensive understanding of the current advancements in catalytic micro-/nanomotors is critical. This review aims to provide an in-depth overview of their fabrication techniques, propulsion mechanisms, key influencing factors, control methodologies, and potential applications. Furthermore, we examine their physical and hydrodynamic properties in fluidic environments to optimize propulsion efficiency. Lastly, we evaluate their biosafety and biocompatibility to facilitate their use in biological systems. The review also addresses key challenges and proposes potential solutions to advance their practical deployment.
Collapse
Affiliation(s)
| | | | - Xuebo Chen
- School of Electronic and Information Engineering, University of Science and Technology Liaoning, Anshan 114051, China; (T.H.); (Y.Y.)
| |
Collapse
|
12
|
Hu JQ, Zhao R, Cui RF, Kou JL, Chen JX. Dynamics of Nanomotors Propelled by Enzyme Cascade Reactions. Int J Mol Sci 2024; 25:12586. [PMID: 39684298 PMCID: PMC11641574 DOI: 10.3390/ijms252312586] [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: 10/24/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Enzyme-powered nanomotors have attracted significant attention in materials science and biomedicine for their biocompatibility, versatility, and the use of biofuels in biological environments. Here, we employ a hybrid mesoscale method combining molecular dynamics and multi-particle collision dynamics (MD-MPC) to study the dynamics of nanomotors powered by enzyme reactions. Two cascade enzymes are constructed to be layered on the same surface of a Janus colloid, providing a confined space that greatly enhances reaction efficiency. Simulations indicate that such a configuration significantly improves the utilization of intermediate products and, consequently, increases the self-propulsion of the Janus motor. By presenting the gradient fields of substrates and products, as well as the hydrodynamics surrounding the motor, we explore the underlying mechanism behind the enhanced autonomous velocity. Additionally, we discuss the improvements in environmental safety of the modified motor, which may shed light on the fabrication of biocatalytic nano-machines in experiments.
Collapse
Affiliation(s)
- Jia-Qi Hu
- School of Physics, Hangzhou Normal University, Hangzhou 311121, China; (J.-Q.H.); (R.Z.)
| | - Rui Zhao
- School of Physics, Hangzhou Normal University, Hangzhou 311121, China; (J.-Q.H.); (R.Z.)
| | - Ru-Fei Cui
- Department of Physics, Hangzhou Dianzi University, Hangzhou 310027, China;
| | - Jian-Long Kou
- Institute of Condensed Matter Physics, Zhejiang Institute of Photoelectronics and Zhejiang Institute for Advanced Light Source, Zhejiang Normal University, Jinhua 321004, China;
| | - Jiang-Xing Chen
- School of Physics, Hangzhou Normal University, Hangzhou 311121, China; (J.-Q.H.); (R.Z.)
| |
Collapse
|
13
|
Choi H, Jeong SH, Simó C, Bakenecker A, Liop J, Lee HS, Kim TY, Kwak C, Koh GY, Sánchez S, Hahn SK. Urease-powered nanomotor containing STING agonist for bladder cancer immunotherapy. Nat Commun 2024; 15:9934. [PMID: 39548120 PMCID: PMC11568179 DOI: 10.1038/s41467-024-54293-z] [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: 03/23/2023] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
Most non-muscle invasive bladder cancers have been treated by transurethral resection and following intravesical injection of immunotherapeutic agents. However, the delivery efficiency of therapeutic agents into bladder wall is low due to frequent urination, which leads to the failure of treatment with side effects. Here, we report a urease-powered nanomotor containing the agonist of stimulator of interferon genes (STING) for the efficient activation of immune cells in the bladder wall. After characterization, we perform in vitro motion analysis and assess in vivo swarming behaviors of nanomotors. The intravesical instillation results in the effective penetration and retention of nanomotors in the bladder. In addition, we confirm the anti-tumor effect of nanomotor containing the STING agonist (94.2% of inhibition), with recruitment of CD8+ T cells (11.2-fold compared with PBS) and enhanced anti-tumor immune responses in bladder cancer model in female mice. Furthermore, we demonstrate the better anti-tumor effect of nanomotor containing the STING agonist than those of the gold standard Bacille Calmette-Guerin therapy and the anti-PD-1 inhibitor pembrolizumab in bladder cancer model. Taken together, the urease-powered nanomotor would provide a paradigm as a next-generation platform for bladder cancer immunotherapy.
Collapse
Affiliation(s)
- Hyunsik Choi
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- PHI BIOMED Co., Seocho-gu, Seoul, Korea
| | - Seung-Hwan Jeong
- Department of Urology, Seoul National University College of Medicine, Jongno-gu, Seoul, Korea
- Department of Urology, Seoul National University Hospital, Jongno-gu, Seoul, Korea
| | - Cristina Simó
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), San Sebastian, Guipúzcoa, Spain
| | - Anna Bakenecker
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Jordi Liop
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), San Sebastian, Guipúzcoa, Spain
| | - Hye Sun Lee
- Department of Urology, Seoul National University Hospital, Jongno-gu, Seoul, Korea
| | - Tae Yeon Kim
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, Korea
| | - Cheol Kwak
- Department of Urology, Seoul National University College of Medicine, Jongno-gu, Seoul, Korea.
- Department of Urology, Seoul National University Hospital, Jongno-gu, Seoul, Korea.
| | - Gou Young Koh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon, Korea.
| | - Samuel Sánchez
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avancats (ICREA), Passeig Lluís Companys 23, Barcelona, Spain.
| | - Sei Kwang Hahn
- PHI BIOMED Co., Seocho-gu, Seoul, Korea.
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, Korea.
| |
Collapse
|
14
|
Chen S, Peetroons X, Bakenecker AC, Lezcano F, Aranson IS, Sánchez S. Collective buoyancy-driven dynamics in swarming enzymatic nanomotors. Nat Commun 2024; 15:9315. [PMID: 39472587 PMCID: PMC11522643 DOI: 10.1038/s41467-024-53664-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 10/15/2024] [Indexed: 11/02/2024] Open
Abstract
Enzymatic nanomotors harvest kinetic energy through the catalysis of chemical fuels. When a drop containing nanomotors is placed in a fuel-rich environment, they assemble into ordered groups and exhibit intriguing collective behaviour akin to the bioconvection of aerobic microorganismal suspensions. This collective behaviour presents numerous advantages compared to individual nanomotors, including expanded coverage and prolonged propulsion duration. However, the physical mechanisms underlying the collective motion have yet to be fully elucidated. Our study investigates the formation of enzymatic swarms using experimental analysis and computational modelling. We show that the directional movement of enzymatic nanomotor swarms is due to their solutal buoyancy. We investigate various factors that impact the movement of nanomotor swarms, such as particle concentration, fuel concentration, fuel viscosity, and vertical confinement. We examine the effects of these factors on swarm self-organization to gain a deeper understanding. In addition, the urease catalysis reaction produces ammonia and carbon dioxide, accelerating the directional movement of active swarms in urea compared with passive ones in the same conditions. The numerical analysis agrees with the experimental findings. Our findings are crucial for the potential biomedical applications of enzymatic nanomotor swarms, ranging from enhanced diffusion in bio-fluids and targeted delivery to cancer therapy.
Collapse
Grants
- 866348 EC | EU Framework Programme for Research and Innovation H2020 | H2020 Priority Excellent Science | H2020 European Research Council (H2020 Excellent Science - European Research Council)
- 2021 SGR 01606 Departament d'Innovació, Universitats i Empresa, Generalitat de Catalunya (Department of Innovation, Education and Enterprise, Government of Catalonia)
- 2023 FI-1 00654 Departament d'Innovació, Universitats i Empresa, Generalitat de Catalunya (Department of Innovation, Education and Enterprise, Government of Catalonia)
- CEX2018-000789-S Ministry of Economy and Competitiveness | Agencia Estatal de Investigación (Spanish Agencia Estatal de Investigación)
- Departament d'Innovació, Universitats i Empresa, Generalitat de Catalunya (Department of Innovation, Education and Enterprise, Government of Catalonia)
- The research leading to these results has also received funding from the grants PID2021-128417OB-I00 and PDC2022-133753-I00 funded by MCIN/AEI/ 10.13039/501100011033 and, by “ERDF A way of making Europe” and European Union Next Generation EU, (Bots4BB and BOJOS projects).
Collapse
Affiliation(s)
- Shuqin Chen
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
- Faculty of Physics, University of Barcelona, Martí i Franquès 1, Barcelona, Spain
| | - Xander Peetroons
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - Anna C Bakenecker
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - Florencia Lezcano
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - Igor S Aranson
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA.
- Department of Chemistry, The Pennsylvania State University, University Park, PA, 16802, USA.
- Department of Mathematics, The Pennsylvania State University, University Park, PA, 16802, USA.
| | - Samuel Sánchez
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain.
- Catalan Institute for Research and Advanced Studies (ICREA), Passeig Lluís Companys 23, Barcelona, Spain.
| |
Collapse
|
15
|
Fraire JC, Prado-Morales C, Aldaz Sagredo A, Caelles AG, Lezcano F, Peetroons X, Bakenecker AC, Di Carlo V, Sánchez S. Swarms of Enzymatic Nanobots for Efficient Gene Delivery. ACS APPLIED MATERIALS & INTERFACES 2024; 16:47192-47205. [PMID: 39262054 PMCID: PMC11403613 DOI: 10.1021/acsami.4c08770] [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: 09/13/2024]
Abstract
This study investigates the synthesis and optimization of nanobots (NBs) loaded with pDNA using the layer-by-layer (LBL) method and explores the impact of their collective motion on the transfection efficiency. NBs consist of biocompatible and biodegradable poly(lactic-co-glycolic acid) (PLGA) nanoparticles and are powered by the urease enzyme, enabling autonomous movement and collective swarming behavior. In vitro experiments were conducted to validate the delivery efficiency of fluorescently labeled NBs, using two-dimensional (2D) and three-dimensional (3D) cell models: murine urothelial carcinoma cell line (MB49) and spheroids from human urothelial bladder cancer cells (RT4). Swarms of pDNA-loaded NBs showed enhancements of 2.2- to 2.6-fold in delivery efficiency and 6.8- to 8.1-fold in material delivered compared to inhibited particles (inhibited enzyme) and the absence of fuel in a 2D cell culture. Additionally, efficient intracellular delivery of pDNA was demonstrated in both cell models by quantifying and visualizing the expression of eGFP. Swarms of NBs exhibited a >5-fold enhancement in transfection efficiency compared to the absence of fuel in a 2D culture, even surpassing the Lipofectamine 3000 commercial transfection agent (cationic lipid-mediated transfection). Swarms also demonstrated up to a 3.2-fold enhancement in the amount of material delivered in 3D spheroids compared to the absence of fuel. The successful transfection of 2D and 3D cell cultures using swarms of LBL PLGA NBs holds great potential for nucleic acid delivery in the context of bladder treatments.
Collapse
Affiliation(s)
- Juan C Fraire
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona, Spain
| | - Carles Prado-Morales
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona, Spain
- Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Ana Aldaz Sagredo
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona, Spain
| | - Ainhoa G Caelles
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona, Spain
| | - Florencia Lezcano
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona, Spain
| | - Xander Peetroons
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona, Spain
| | - Anna C Bakenecker
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona, Spain
| | - Valerio Di Carlo
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona, Spain
| | - Samuel Sánchez
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona, Spain
- Catalan Institute for Research and Advanced Studies (ICREA), Passeig de Lluís Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
16
|
Lanz C, Künnecke N, Krysiak Y, Polarz S. Accelerating and breaking adaptive nano-colloids (<100 nm) into unsteady state operation via push-pull effects. NANOSCALE 2024; 16:15358-15365. [PMID: 39087938 DOI: 10.1039/d4nr01644k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
Unlike conventional colloids showing random mobility because of Brownian motion, active colloids contain nanomotors that translate chemical or physical triggers into directed movement. Whereas the acceleration of such particles works well, it is difficult to decelerate them by request. Compared to the existing literature on microscaled swimmers/robots, the main question of the current paper is whether nanoscaled colloids (<100 nm) can also be actively controlled despite the stronger relevance of rotational diffusion at such dimensions. We developed nanoparticles comprising two independent mechanisms for propulsion: a chemical engine associated with a Janus-type modification of organosilica nanoparticles and physical locomotion because of a superparamagnetic core inside these particles. Both triggers can be used independently to initiate the particles' directed and anisotropic movement. The magnetic forces can be tuned, most importantly concerning the angle defining the chemical acceleration. Superposition and a boost state are adopted for a parallel alignment. However, when the magnetic field acting on the particles is turned to an antiparallel orientation, a rapid deceleration can be observed, and the colloids halt.
Collapse
Affiliation(s)
- Cornelia Lanz
- Institute of Inorganic Chemistry, Leibniz Universität Hannover, Callinstrasse 9, D-30167 Hannover, Germany.
| | - Nele Künnecke
- Institute of Inorganic Chemistry, Leibniz Universität Hannover, Callinstrasse 9, D-30167 Hannover, Germany.
| | - Yaşar Krysiak
- Institute of Inorganic Chemistry, Leibniz Universität Hannover, Callinstrasse 9, D-30167 Hannover, Germany.
| | - Sebastian Polarz
- Institute of Inorganic Chemistry, Leibniz Universität Hannover, Callinstrasse 9, D-30167 Hannover, Germany.
| |
Collapse
|
17
|
Tang S, Tang D, Zhou H, Li Y, Zhou D, Peng X, Ren C, Su Y, Zhang S, Zheng H, Wan F, Yoo J, Han H, Ma X, Gao W, Wu S. Bacterial outer membrane vesicle nanorobot. Proc Natl Acad Sci U S A 2024; 121:e2403460121. [PMID: 39008666 PMCID: PMC11287275 DOI: 10.1073/pnas.2403460121] [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: 02/19/2024] [Accepted: 06/05/2024] [Indexed: 07/17/2024] Open
Abstract
Autonomous nanorobots represent an advanced tool for precision therapy to improve therapeutic efficacy. However, current nanorobotic designs primarily rely on inorganic materials with compromised biocompatibility and limited biological functions. Here, we introduce enzyme-powered bacterial outer membrane vesicle (OMV) nanorobots. The immobilized urease on the OMV membrane catalyzes the decomposition of bioavailable urea, generating effective propulsion for nanorobots. This OMV nanorobot preserves the unique features of OMVs, including intrinsic biocompatibility, immunogenicity, versatile surface bioengineering for desired biofunctionalities, capability of cargo loading and protection. We present OMV-based nanorobots designed for effective tumor therapy by leveraging the membrane properties of OMVs. These involve surface bioengineering of robotic body with cell-penetrating peptide for tumor targeting and penetration, which is further enhanced by active propulsion of nanorobots. Additionally, OMV nanorobots can effectively safeguard the loaded gene silencing tool, small interfering RNA (siRNA), from enzymatic degradation. Through systematic in vitro and in vivo studies using a rodent model, we demonstrate that these OMV nanorobots substantially enhanced siRNA delivery and immune stimulation, resulting in the utmost effectiveness in tumor suppression when juxtaposed with static groups, particularly evident in the orthotopic bladder tumor model. This OMV nanorobot opens an inspiring avenue to design advanced medical robots with expanded versatility and adaptability, broadening their operation scope in practical biomedical domains.
Collapse
Affiliation(s)
- Songsong Tang
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA91125
| | - Daitian Tang
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
- South China Hospital, Health Science Center, Shenzhen University, Shenzhen518116, People’s Republic of China
- Luohu Clinical Institute of Shantou University Medical College, Shantou University Medical College, Shantou515000, People’s Republic of China
| | - Houhong Zhou
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
- Department of General Surgery, Shenzhen Samii Medical Center, Shenzhen518118, People’s Republic of China
| | - Yangyang Li
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
| | - Dewang Zhou
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
| | - Xiqi Peng
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
- South China Hospital, Health Science Center, Shenzhen University, Shenzhen518116, People’s Republic of China
- Luohu Clinical Institute of Shantou University Medical College, Shantou University Medical College, Shantou515000, People’s Republic of China
| | - Chunyu Ren
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
| | - Yilin Su
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
- Luohu Clinical Institute of Shantou University Medical College, Shantou University Medical College, Shantou515000, People’s Republic of China
| | - Shaohua Zhang
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
- South China Hospital, Health Science Center, Shenzhen University, Shenzhen518116, People’s Republic of China
| | - Haoxiang Zheng
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
- South China Hospital, Health Science Center, Shenzhen University, Shenzhen518116, People’s Republic of China
| | - Fangchen Wan
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
| | - Jounghyun Yoo
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA91125
| | - Hong Han
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA91125
| | - Xiaotian Ma
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA91125
| | - Wei Gao
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA91125
| | - Song Wu
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen518000, People’s Republic of China
- South China Hospital, Health Science Center, Shenzhen University, Shenzhen518116, People’s Republic of China
- Luohu Clinical Institute of Shantou University Medical College, Shantou University Medical College, Shantou515000, People’s Republic of China
| |
Collapse
|
18
|
Dindo M, Bevilacqua A, Soligo G, Calabrese V, Monti A, Shen AQ, Rosti ME, Laurino P. Chemotactic Interactions Drive Migration of Membraneless Active Droplets. J Am Chem Soc 2024; 146:15965-15976. [PMID: 38620052 DOI: 10.1021/jacs.4c02823] [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: 04/17/2024]
Abstract
In nature, chemotactic interactions are ubiquitous and play a critical role in driving the collective behavior of living organisms. Reproducing these interactions in vitro is still a paramount challenge due to the complexity of mimicking and controlling cellular features, such as tangled metabolic networks, cytosolic macromolecular crowding, and cellular migration, on a microorganism size scale. Here, we generate enzymatically active cell-sized droplets able to move freely, and by following a chemical gradient, able to interact with the surrounding droplets in a collective manner. The enzyme within the droplets generates a pH gradient that extends outside the edge of the droplets. We discovered that the external pH gradient triggers droplet migration and controls its directionality, which is selectively toward the neighboring droplets. Hence, by changing the enzyme activity inside the droplet, we tuned the droplet migration speed. Furthermore, we showed that these cellular-like features can facilitate the reconstitution of a simple and linear protometabolic pathway and increase the final reaction product generation. Our work suggests that simple and stable membraneless droplets can reproduce complex biological phenomena, opening new perspectives as bioinspired materials and synthetic biology tools.
Collapse
Affiliation(s)
- Mirco Dindo
- Protein Engineering and Evolution Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0412, Japan
| | - Alessandro Bevilacqua
- Protein Engineering and Evolution Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0412, Japan
| | - Giovanni Soligo
- Complex Fluids and Flows Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0412, Japan
| | - Vincenzo Calabrese
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0412, Japan
| | - Alessandro Monti
- Complex Fluids and Flows Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0412, Japan
| | - Amy Q Shen
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0412, Japan
| | - Marco Edoardo Rosti
- Complex Fluids and Flows Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0412, Japan
| | - Paola Laurino
- Protein Engineering and Evolution Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa 904-0412, Japan
- Institute for Protein Research, Osaka University, Suita 565-0871, Japan
| |
Collapse
|
19
|
Tseng YC, Song J, Zhang J, Shandilya E, Sen A. Chemomechanical Communication between Liposomes Based on Enzyme Cascades. J Am Chem Soc 2024; 146:16097-16104. [PMID: 38805671 DOI: 10.1021/jacs.4c03415] [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: 05/30/2024]
Abstract
Communication between cells is crucial to the survival of both uni- and multicellular organisms. The primary mode of communication involves chemical cues. There is great current interest in mimicking this behavior in synthetic cells to understand the physical basis of intercellular communication and design collective functional behavior. Using liposomal cell mimics, we demonstrate how a chemical input can elicit a mechanical response (enhanced motility). We employed a single substrate to trigger enzyme cascade-induced control of the diffusion of up to three different liposome populations. Furthermore, substrate competition allows temporal control over enhanced diffusion. The use of enzyme cascades to propagate chemical signals provides a robust and efficient mechanism for diverse populations of protocells to coordinate their motion in response to signals from each other.
Collapse
Affiliation(s)
- Yu-Ching Tseng
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Jiaqi Song
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Jianhua Zhang
- College of Chemistry and Chemical Engineering, Wuhan Textile University, Wuhan 430200, China
| | - Ekta Shandilya
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Ayusman Sen
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
20
|
Chen T, Duan Y, Dai W, Guo W, Jing P, Ma S, Mao C, Wan M, Zhang S. Carbon monoxide-releasing nanomotors based on endogenous biochemical reactions for tumor therapy. J Colloid Interface Sci 2024; 663:396-404. [PMID: 38412725 DOI: 10.1016/j.jcis.2024.02.135] [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: 12/07/2023] [Revised: 02/02/2024] [Accepted: 02/17/2024] [Indexed: 02/29/2024]
Abstract
The lack of selective release ability in the tumor microenvironment and the limited efficacy of monotherapy are important factors that limit the current use of carbon monoxide (CO) donors for tumor therapy. Herein, inspired by endogenous biochemical reactions in vivo, one kind of CO-releasing nanomotor was designed for the multimodal synergistic treatment of tumor. Specifically, glucose oxidase (GOx) and 5-aminolevulinic acid (5-ALA) were co-modified onto metal-organic framework material (MIL-101) to obtain MIL-GOx-ALA nanomotors (M-G-A NMs), which exhibit excellent biocompatibility and degradation ability in tumor microenvironment. Subsequently, the released 5-ALA generates CO in the tumor microenvironment through an endogenous reaction and further acts on mitochondria to release large amounts of reactive oxygen species (ROS), which directly kill tumor cells. Furthermore, the produced ROS and the degradation products of M-G-A NMs can also provide the reaction substrate for the Fenton reaction, thereby enhancing chemodynamic therapy (CDT) and inducing apoptosis of tumor cells. Both in vitro and in vivo experimental data confirm the successful occurrence of the above process, and the combination of CO gas therapy/enhanced CDT can effectively inhibit tumor growth. This CDT-enhancing agent designed based on endogenous biochemical reactions has good prospects for tumor treatment application.
Collapse
Affiliation(s)
- Tiantian Chen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Yu Duan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Wenjun Dai
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Wenyan Guo
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Pengshen Jing
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Shenglin Ma
- Molecular Diagnostic Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou 310006, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China; Institute for Life and Health, Nanjing Drum Tower Hospital, Nanjing Normal University, Nanjing 210023, China.
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China; Institute for Life and Health, Nanjing Drum Tower Hospital, Nanjing Normal University, Nanjing 210023, China.
| | - Shirong Zhang
- Molecular Diagnostic Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou 310006, China.
| |
Collapse
|
21
|
Patiño Padial T, Del Grosso E, Gentile S, Baranda Pellejero L, Mestre R, Paffen LJMM, Sánchez S, Ricci F. Synthetic DNA-based Swimmers Driven by Enzyme Catalysis. J Am Chem Soc 2024; 146:12664-12671. [PMID: 38587543 DOI: 10.1021/jacs.4c02094] [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: 04/09/2024]
Abstract
Here, we report DNA-based synthetic nanostructures decorated with enzymes (hereafter referred to as DNA-enzyme swimmers) that self-propel by converting the enzymatic substrate to the product in solution. The DNA-enzyme swimmers are obtained from tubular DNA structures that self-assemble spontaneously by the hybridization of DNA tiles. We functionalize these DNA structures with two different enzymes, urease and catalase, and show that they exhibit concentration-dependent movement and enhanced diffusion upon addition of the enzymatic substrate (i.e., urea and H2O2). To demonstrate the programmability of such DNA-based swimmers, we also engineer DNA strands that displace the enzyme from the DNA scaffold, thus acting as molecular "brakes" on the DNA swimmers. These results serve as a first proof of principle for the development of synthetic DNA-based enzyme-powered swimmers that can self-propel in fluids.
Collapse
Affiliation(s)
- Tania Patiño Padial
- Department of Chemical Sciences and Technologies, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
- Biomedical Engineering Department, Institute for Complex Molecular Systems, Technische Universiteit Eindhoven, Het Kranenveld 14, 5612 AZ Eindhoven, The Netherlands
| | - Erica Del Grosso
- Department of Chemical Sciences and Technologies, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Serena Gentile
- Department of Chemical Sciences and Technologies, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Lorena Baranda Pellejero
- Department of Chemical Sciences and Technologies, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Rafael Mestre
- School of Electronics and Computer Science (ECS), University of Southampton, University Road, Southampton SO17 1BJ, U.K
| | - Lars J M M Paffen
- Biomedical Engineering Department, Institute for Complex Molecular Systems, Technische Universiteit Eindhoven, Het Kranenveld 14, 5612 AZ Eindhoven, The Netherlands
| | - Samuel Sánchez
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona, Spain
- Catalan Institute for Research and Advanced Studies (ICREA), Pg. Lluís Companys 23, Barcelona 08010, Spain
| | - Francesco Ricci
- Department of Chemical Sciences and Technologies, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| |
Collapse
|
22
|
Liu Y, Cui S, Ma W, Wu Y, Xin R, Bai Y, Chen Z, Xu J, Ge J. Direct Imaging of Protein Clusters in Metal-Organic Frameworks. J Am Chem Soc 2024; 146:12565-12576. [PMID: 38661569 DOI: 10.1021/jacs.4c01483] [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: 04/26/2024]
Abstract
Protein@metal-organic frameworks (P@MOFs) prepared by coprecipitation of protein, metal ions, and organic ligands represent an effective method for protein stabilization with a wide spectrum of applications. However, the formation mechanism of P@MOFs via the coprecipitation process and the reason why proteins can retain their biological activity in the frameworks with highly concentrated metal ions remain unsettled. Here, by a combined methodology of single molecule localization microscopy and clustering analysis, we discovered that in this process enzyme molecules form clusters with metal ions and organic ligands, contributing to both the nucleation and subsequent crystal growth. We proposed that the clusters played an important role in the retention of overall enzymatic activity by sacrificing protein molecules on the cluster surface. This work offers fresh perspectives on protein behaviors in the formation of P@MOFs, inspiring future endeavors in the design and development of artificial bionanocomposites with high biological activities.
Collapse
Affiliation(s)
- Yu Liu
- Key Lab for Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Shitong Cui
- Key Lab for Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Wenjun Ma
- The State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Yibo Wu
- Key Lab for Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Ruobing Xin
- Key Lab for Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Yunxiu Bai
- Key Lab for Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Zhuo Chen
- The State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Jianhong Xu
- The State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
| | - Jun Ge
- Key Lab for Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing 100084, China
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen 518055, China
| |
Collapse
|
23
|
Li Y, Liu J, Wu Y, He Q. Rotary F oF 1-ATP Synthase-Driven Flasklike Pentosan Colloidal Motors with ATP Synthesis and Storage. J Am Chem Soc 2024. [PMID: 38598314 DOI: 10.1021/jacs.4c00334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
We report the hierarchical assembly of a chloroplast-derived rotary FoF1-ATPase motor-propelled flasklike pentosan colloidal motor (FPCM) with the ability of the synthesis, storage, and triggered release of biological energy currency ATP. These streamlined and submicrometer-sized hollow flasklike pentosan colloidal motors are prepared by combining a soft-template-based hydrothermal polymerization with a vacuum infusion of chloroplast-derived proteoliposomes containing rotary FoF1-ATPase motors. The generation of proton motive force across the proteoliposomes by injecting an acidic buffer solution promotes the rotation of FoF1-ATPase motors to drive the self-propelled motion of FPCMs, accompanying the inner ATP synthesis and storage. These rotary FoF1-ATPase motor-powered FPCMs exhibit a chemotactic behavior by migrating from their neck opening to their round bottom along a proton gradient of the external environment (negative chemotaxis). Such rotary biomolecular motor-driven flasklike pentosan colloidal motors with ATP synthesis and on-demand release make them promising candidates for engineering novel intelligent nanocarriers to actively regulate cellular metabolism.
Collapse
Affiliation(s)
- Yue Li
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
| | - Jun Liu
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Wenzhou Institute, University of Chinese Academy of Sciences, 1 Jinlian Street, Wenzhou 325000, China
| | - Yingjie Wu
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
| | - Qiang He
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
| |
Collapse
|
24
|
Simó C, Serra-Casablancas M, Hortelao AC, Di Carlo V, Guallar-Garrido S, Plaza-García S, Rabanal RM, Ramos-Cabrer P, Yagüe B, Aguado L, Bardia L, Tosi S, Gómez-Vallejo V, Martín A, Patiño T, Julián E, Colombelli J, Llop J, Sánchez S. Urease-powered nanobots for radionuclide bladder cancer therapy. NATURE NANOTECHNOLOGY 2024; 19:554-564. [PMID: 38225356 PMCID: PMC11026160 DOI: 10.1038/s41565-023-01577-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 11/20/2023] [Indexed: 01/17/2024]
Abstract
Bladder cancer treatment via intravesical drug administration achieves reasonable survival rates but suffers from low therapeutic efficacy. To address the latter, self-propelled nanoparticles or nanobots have been proposed, taking advantage of their enhanced diffusion and mixing capabilities in urine when compared with conventional drugs or passive nanoparticles. However, the translational capabilities of nanobots in treating bladder cancer are underexplored. Here, we tested radiolabelled mesoporous silica-based urease-powered nanobots in an orthotopic mouse model of bladder cancer. In vivo and ex vivo results demonstrated enhanced nanobot accumulation at the tumour site, with an eightfold increase revealed by positron emission tomography in vivo. Label-free optical contrast based on polarization-dependent scattered light-sheet microscopy of cleared bladders confirmed tumour penetration by nanobots ex vivo. Treating tumour-bearing mice with intravesically administered radio-iodinated nanobots for radionuclide therapy resulted in a tumour size reduction of about 90%, positioning nanobots as efficient delivery nanosystems for bladder cancer therapy.
Collapse
Affiliation(s)
- Cristina Simó
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Spain
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St Louis, MO, USA
| | - Meritxell Serra-Casablancas
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Ana C Hortelao
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Valerio Di Carlo
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Sandra Guallar-Garrido
- Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Sandra Plaza-García
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Spain
| | - Rosa Maria Rabanal
- Unitat de Patologia Murina i Comparada, Department of Animal Medicine and Surgery, Veterinary Faculty, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Pedro Ramos-Cabrer
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Balbino Yagüe
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Spain
| | - Laura Aguado
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Spain
- Laboratory of Neuroimaging and Biomarkers of Inflammation, Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Lídia Bardia
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Sébastien Tosi
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Department of Biomedical Sciences, Faculty Of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vanessa Gómez-Vallejo
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Spain
| | - Abraham Martín
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Laboratory of Neuroimaging and Biomarkers of Inflammation, Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Tania Patiño
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
- Biomedical Engineering Department, Institute for Complex Molecular Systems, Technische Universiteit Eindhoven, Eindhoven, The Netherlands
| | - Esther Julián
- Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Julien Colombelli
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
| | - Jordi Llop
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Spain.
| | - Samuel Sánchez
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
25
|
Liu W, Nie H, Li H, Liu Y, Tian M, Wang S, Yang Y, Long W. Engineered platelet cell motors for boosted cancer radiosensitization. J Colloid Interface Sci 2024; 658:540-552. [PMID: 38128197 DOI: 10.1016/j.jcis.2023.12.091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/10/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Design of engineered cells to target and deliver nanodrugs to the hard-to-reach regions has become an exciting research area. However, the limited penetration and retention of cell-based carriers in tumor tissue restricted their therapeutic efficiency. Inspired by the enhanced delivery behavior of mobile micro/nanomotors, herein, urease-powered platelet cell motors (PLT@Au@Urease) capable of active locomotion, tumor targeting, and radiosensitizers delivery were designed for boosting radiosensitization. The engineered platelet cell motors were constructed by in situ synthesis and loading of radiosensitizers gold nanoparticles in platelets, and then conjugation with urease as the engine. Under physiological concentration of urea, thrust around PLT@Au@Urease motors can be generated via the biocatalytic reactions of urease, leading to rapid tumor cell targeting and enhanced cellular uptake of radiosensitizers. Encouragingly, in comparison with engineered PLT without propulsion capability (PLT@Au), the self-propelled PLT@Au@Urease motors could significantly increase intracellular ROS level and exacerbate nuclear DNA damage induced by γ-radiation, resulting in a remarkably high sensitization enhancement rate (1.89) than that of PLT@Au (1.08). In vivo experiments with 4 T1-bearing mice demonstrated that PLT@Au@Urease in combination with radiation therapy possessed good antitumor performance. Such an intelligent cell motor would provide a promising approach to enhance radiosensitization and broaden the applications of cell motor-based delivery systems.
Collapse
Affiliation(s)
- Wei Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Hongmei Nie
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - He Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Ya Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Maoye Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Shuhuai Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yuwei Yang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Wei Long
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| |
Collapse
|
26
|
Chen S, Prado-Morales C, Sánchez-deAlcázar D, Sánchez S. Enzymatic micro/nanomotors in biomedicine: from single motors to swarms. J Mater Chem B 2024; 12:2711-2719. [PMID: 38239179 DOI: 10.1039/d3tb02457a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Micro/nanomotors (MNMs) have evolved from single self-propelled entities to versatile systems capable of performing one or multiple biomedical tasks. When single MNMs self-assemble into coordinated swarms, either under external control or triggered by chemical reactions, they offer advantages that individual MNMs cannot achieve. These benefits include intelligent multitasking and adaptability to changes in the surrounding environment. Here, we provide our perspective on the evolution of MNMs, beginning with the development of enzymatic MNMs since the first theoretical model was proposed in 2005. These enzymatic MNMs hold immense promise in biomedicine due to their advantages in biocompatibility and fuel availability. Subsequently, we introduce the design and application of single motors in biomedicine, followed by the control of MNM swarms and their biomedical applications. In the end, we propose viable solutions for advancing the development of MNM swarms and anticipate valuable insights into the creation of more intelligent and controllable MNM swarms for biomedical applications.
Collapse
Affiliation(s)
- Shuqin Chen
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri I Reixac 10-12, 08028 Barcelona, Spain.
| | - Carles Prado-Morales
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri I Reixac 10-12, 08028 Barcelona, Spain.
| | - Daniel Sánchez-deAlcázar
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri I Reixac 10-12, 08028 Barcelona, Spain.
| | - Samuel Sánchez
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri I Reixac 10-12, 08028 Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Psg. Lluís Companys, 23, 08010, Barcelona, Spain
| |
Collapse
|
27
|
Ruiz-González N, Esporrín-Ubieto D, Hortelao AC, Fraire JC, Bakenecker AC, Guri-Canals M, Cugat R, Carrillo JM, Garcia-Batlletbó M, Laiz P, Patiño T, Sánchez S. Swarms of Enzyme-Powered Nanomotors Enhance the Diffusion of Macromolecules in Viscous Media. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309387. [PMID: 38200672 DOI: 10.1002/smll.202309387] [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: 10/17/2023] [Indexed: 01/12/2024]
Abstract
Over the past decades, the development of nanoparticles (NPs) to increase the efficiency of clinical treatments has been subject of intense research. Yet, most NPs have been reported to possess low efficacy as their actuation is hindered by biological barriers. For instance, synovial fluid (SF) present in the joints is mainly composed of hyaluronic acid (HA). These viscous media pose a challenge for many applications in nanomedicine, as passive NPs tend to become trapped in complex networks, which reduces their ability to reach the target location. This problem can be addressed by using active NPs (nanomotors, NMs) that are self-propelled by enzymatic reactions, although the development of enzyme-powered NMs, capable of navigating these viscous environments, remains a considerable challenge. Here, the synergistic effects of two NMs troops, namely hyaluronidase NMs (HyaNMs, Troop 1) and urease NMs (UrNMs, Troop 2) are demonstrated. Troop 1 interacts with the SF by reducing its viscosity, thus allowing Troop 2 to swim more easily through the SF. Through their collective motion, Troop 2 increases the diffusion of macromolecules. These results pave the way for more widespread use of enzyme-powered NMs, e.g., for treating joint injuries and improving therapeutic effectiveness compared with traditional methods.
Collapse
Affiliation(s)
- Noelia Ruiz-González
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - David Esporrín-Ubieto
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - Ana C Hortelao
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - Juan C Fraire
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - Anna C Bakenecker
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - Marta Guri-Canals
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
| | - Ramón Cugat
- Mutualidad de Futbolistas - Delegación Catalana, Federación Española de Fútbol, Barcelona, 08010, Spain
- Instituto Cugat, Hospital Quironsalud Barcelona, Spain, Fundación García Cugat, Barcelona, 08023, Spain
| | - José María Carrillo
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain. García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, 46115, Spain
| | | | - Patricia Laiz
- Instituto Cugat, Hospital Quironsalud Barcelona, Spain, Fundación García Cugat, Barcelona, 08023, Spain
| | - Tania Patiño
- Department of Biomedical Engineering, Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, 5612 AZ, The Netherlands
| | - Samuel Sánchez
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, 08028, Spain
- Institució Catalana de Recerca i Estudies Avancats (ICREA), Passeig Lluís Companys 23, Barcelona, 08010, Spain
| |
Collapse
|
28
|
Wu J, Jiao N, Lin D, Li N, Ma T, Tung S, Cheng W, Wu A, Liu L. Dual-Responsive Nanorobot-Based Marsupial Robotic System for Intracranial Cross-Scale Targeting Drug Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2306876. [PMID: 37899660 DOI: 10.1002/adma.202306876] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/27/2023] [Indexed: 10/31/2023]
Abstract
Nanorobots capable of active movement are an exciting technology for targeted therapeutic intervention. However, the extensive motion range and hindrance of the blood-brain barrier impeded their clinical translation in glioblastoma therapy. Here, a marsupial robotic system constructed by integrating chemical/magnetic hybrid nanorobots (child robots) with a miniature magnetic continuum robot (mother robot) for intracranial cross-scale targeting drug delivery is reported. For primary targeting on macroscale, the continuum robot enters the cranial cavity through a minimally invasive channel (e.g., Ommaya device) in the skull and transports the nanorobots to pathogenic regions. Upon circumventing the blood-brain barrier, the released nanorobots perform secondary targeting on microscale to further enhance the spatial resolution of drug delivery. In vitro experiments against primary glioblastoma cells derived from different patients are conducted for personalized treatment guidance. The operation feasibility within organisms is shown in ex vivo swine brain experiments. The biosafety of the treatment system is suggested in in vivo experiments. Owing to the hierarchical targeting method, the targeting rate, targeting accuracy, and treatment efficacy have improved greatly. The marsupial robotic system offers a novel intracranial local therapeutic strategy and constitutes a key milestone in the development of glioblastoma treatment platforms.
Collapse
Affiliation(s)
- Junfeng Wu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, 110016, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Niandong Jiao
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, 110016, China
| | - Daojing Lin
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, 110016, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Na Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, 110016, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tianyang Ma
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Steve Tung
- Department of Mechanical Engineering, University of Arkansas, Arkansas, 72701, USA
| | - Wen Cheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Anhua Wu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Lianqing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, 110016, China
| |
Collapse
|
29
|
Sapre A, Mandal NS, Somasundar A, Bhide A, Song J, Borhan A, Sen A. Enzyme Catalysis Causes Fluid Flow, Motility, and Directional Transport on Supported Lipid Bilayers. ACS APPLIED MATERIALS & INTERFACES 2024; 16:9380-9387. [PMID: 38319873 DOI: 10.1021/acsami.3c15383] [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: 02/08/2024]
Abstract
The dynamic interplay between the composition of lipid membranes and the behavior of membrane-bound enzymes is critical to the understanding of cellular function and viability, and the design of membrane-based biosensing platforms. While there is a significant body of knowledge about how lipid composition and dynamics affect membrane-bound enzymes, little is known about how enzyme catalysis influences the motility and lateral transport on lipid membranes. Using enzyme-attached lipids in supported bilayers (SLBs), we provide direct evidence of catalysis-induced fluid flow that underlies the observed motility on SLBs. Additionally, by using active enzyme patches, we demonstrate the directional transport of tracer particles on SLBs. As expected, enhancing the membrane viscosity by incorporating cholesterol into the bilayer suppresses the overall movement. These are the first steps in understanding diffusion and transport on lipid membranes due to active, out-of-equilibrium processes that are the hallmark of living systems. In general, our study demonstrates how active enzymes can be used to control diffusion and transport in confined 2-D environments.
Collapse
Affiliation(s)
- Aditya Sapre
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Niladri Sekhar Mandal
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Ambika Somasundar
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Ashlesha Bhide
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Jiaqi Song
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Ali Borhan
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Ayusman Sen
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
- Department of Chemistry, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
30
|
Patiño T, Llacer-Wintle J, Pujals S, Albertazzi L, Sánchez S. Unveiling protein corona formation around self-propelled enzyme nanomotors by nanoscopy. NANOSCALE 2024; 16:2904-2912. [PMID: 38054755 DOI: 10.1039/d3nr03749e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
The interaction of nanoparticles with biological media is a topic of general interest for drug delivery systems and among those for active nanoparticles, also called nanomotors. Herein, we report the use of super resolution microscopy, in particular, stochastic optical reconstruction microscopy (STORM), to characterize the formation of a protein corona around active enzyme-powered nanomotors. First, we characterized the distribution and number of enzymes on nano-sized particles and characterized their motion capabilities. Then, we incubated the nanomotors with fluorescently labelled serum proteins. Interestingly, we observed a significant decrease of protein corona formation (20%) and different composition, which was studied by proteomic analysis. Moreover, motion was not hindered, as nanomotors displayed enhanced diffusion regardless of the protein corona. Elucidating how active particles interact with biological media and maintain their self-propulsion after protein corona formation will pave the way for the use of these systems in complex biological fluids in biomedicine.
Collapse
Affiliation(s)
- Tania Patiño
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028, Barcelona, Catalonia, Spain.
- Biomedical Engineering Department, Institute for Complex Molecular Systems. Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Joaquin Llacer-Wintle
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028, Barcelona, Catalonia, Spain.
| | - Sílvia Pujals
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028, Barcelona, Catalonia, Spain.
| | - Lorenzo Albertazzi
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028, Barcelona, Catalonia, Spain.
- Biomedical Engineering Department, Institute for Complex Molecular Systems. Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Samuel Sánchez
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028, Barcelona, Catalonia, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Pg. Lluís Companys 23, Barcelona, 08010, Spain
| |
Collapse
|
31
|
Sun J, Wu J, Ju H. Effects of Size and Asymmetry on Catalase-Powered Silica Micro/nanomotors. Chem Asian J 2024; 19:e202300900. [PMID: 37990785 DOI: 10.1002/asia.202300900] [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: 10/12/2023] [Revised: 11/06/2023] [Accepted: 11/20/2023] [Indexed: 11/23/2023]
Abstract
Enzyme-powered micro/nanomotors that can autonomously move in biological environment are attractive in the fields of biology and biomedicine. The fabrication of enzyme-powered micro/nanomotors normally focuses on constructing Janus structures of micro/nanomaterials, based on the intuition that the Janus coating of enzymes can generate driving force from asymmetric catalytic reactions. Here, in the fabrication of catalase-powered silica micro/nanomotors (C-MNMs), an archetypical model of enzyme-powered micro/nanomotors, we find the silica size rather than asymmetric coating of catalase determines the motion ability of C-MNMs. The effects of size and asymmetry have been investigated by a series of C-MNMs at various sizes (0.5, 2, 5 and 10 μm) and asymmetric levels (full-, half- and most-coated with catalase). The motion performance indicates that 500 nm and 2 μm C-MNMs show obvious increases (varying from 134% to 618%) of diffusion coefficient, but C-MNMs bigger than 5 μm have no self-propulsion behaviour at all, regardless of asymmetric levels. In addition, although asymmetry facilitates enhanced diffusion of C-MNMs, only 2 μm C-MNMs are sensitive to asymmetric level. This work elucidates the primary and secondary roles of size and asymmetry in the preparation of C-MNMs, paving the way to fabricate enzyme-powered micro/nanomotors with high motion performance in future.
Collapse
Affiliation(s)
- Jun Sun
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 xianlin Road, Nanjing, 210023, P. R. China
| | - Jie Wu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 xianlin Road, Nanjing, 210023, P. R. China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 xianlin Road, Nanjing, 210023, P. R. China
| |
Collapse
|
32
|
Zhou X, Ma E, Zhang Y, Xing Y, Xu W, Chen L, Zhou H, Zhang X, Jiang C, Xu K, Wang H, Zheng S. NIR-Actuated Targeted Janus Nanomotors Remodel Immunosuppressive Tumor Microenvironment for Augmented Cancer Immunotherapy. Adv Healthc Mater 2024; 13:e2302272. [PMID: 37824087 DOI: 10.1002/adhm.202302272] [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: 07/17/2023] [Revised: 10/09/2023] [Indexed: 10/13/2023]
Abstract
Tumor-associated macrophages (TAMs) always display immunosuppressive M2 phenotype in the tumor microenvironment to facilitate tumor growth, invasion, and metastasis. Ibrutinib (IBR), a novel irreversible Bruton's tyrosine kinase (BTK) inhibitor, has been employed to repolarize the BTK-overexpressed TAMs from M2 to M1 phenotype to remodel the immunosuppressive tumor microenvironment. However, the poor solubility of IBR extremely hinders its bioavailability, which results in low tumor accumulation and TAMs uptake in vivo. Herein, NIR laser-actuated Janus nanomotors are proposed for the effective and deep delivery of IBR to TAMs in solid tumor for targeted immunotherapy. Under NIR irradiation, the Janus nanomotors exhibit efficient photothermal conversion to produce powerful propulsion via self-thermophoresis with a speed of 12.15 µm s-1 . Combined with the salic acid targeting and IBR loading, the nanomotors significantly boost their binding and uptake efficacy by M2-like macrophages during the active motion, which highly facilitate the reprogramming of M2 to M1 macrophages in vitro. Furtherly, the autonomous motion also validly improves in vivo accumulation and penetration depth in tumors to alter the M1/M2 polarization balance and activate T cells. Overall, the synthesized IC@MSA JNMs would provide a promising strategy for the efficient delivery of immunological agents toward targeted cancer immunotherapy.
Collapse
Affiliation(s)
- Xiaoyu Zhou
- School of Medical Imaging, Xuzhou Medical University, 221006, Xuzhou, China
- Department of Radiology, Chengdu Fifth People's Hospital, Chengdu, 611130, China
| | - Enhui Ma
- School of Chemical Engineering and Technology, China University of Mining and Technology, 221116, Xuzhou, China
| | - Yingying Zhang
- School of Medical Imaging, Xuzhou Medical University, 221006, Xuzhou, China
| | - Yujuan Xing
- School of Medical Imaging, Xuzhou Medical University, 221006, Xuzhou, China
| | - Wenbei Xu
- School of Medical Imaging, Xuzhou Medical University, 221006, Xuzhou, China
| | - Liang Chen
- School of Medical Imaging, Xuzhou Medical University, 221006, Xuzhou, China
| | - Hong Zhou
- School of Medical Imaging, Xuzhou Medical University, 221006, Xuzhou, China
| | - Xinran Zhang
- School of Medical Imaging, Xuzhou Medical University, 221006, Xuzhou, China
| | - Canran Jiang
- School of Medical Imaging, Xuzhou Medical University, 221006, Xuzhou, China
| | - Kai Xu
- School of Medical Imaging, Xuzhou Medical University, 221006, Xuzhou, China
- Department of Radiology, Affiliated Hospital of Xuzhou Medical University, 221004, Xuzhou, China
| | - Hong Wang
- School of Chemical Engineering and Technology, China University of Mining and Technology, 221116, Xuzhou, China
| | - Shaohui Zheng
- School of Medical Imaging, Xuzhou Medical University, 221006, Xuzhou, China
- Department of Radiology, Affiliated Hospital of Xuzhou Medical University, 221004, Xuzhou, China
| |
Collapse
|
33
|
Archer RJ, Ebbens SJ. Symmetrical Catalytic Colloids Display Janus-Like Active Brownian Particle Motion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303154. [PMID: 37870200 PMCID: PMC10667803 DOI: 10.1002/advs.202303154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/30/2023] [Indexed: 10/24/2023]
Abstract
Catalytic Janus colloids, with one hemi-sphere covered by a hydrogen peroxide reduction catalyst such as platinum, represent one of the most experimentally explored examples of self-motile active colloid systems. This paper comparatively investigates the motile behavior of symmetrical catalytic colloids produced by a solution-based metal salt reduction process. Despite the significant differences in the distribution of catalytic activity, this study finds that the motion produced by symmetrical colloids is equivalent to that previously reported for Janus colloids. It also shows that introducing a Janus structure to the symmetrical colloids via masking does not significantly modify their motion. These findings could indicate that very subtle variations in surface reactivity can be sufficient to produce Janus-like active Brownian particle-type motion, or that a symmetry-breaking phenomena is present. The study will consequently motivate fresh theoretical attention and also demonstrate a straightforward route to access large quantities of motile active colloids, which are expected to show subtly different phenomenology compared to those with Janus structures.
Collapse
Affiliation(s)
- Richard J. Archer
- Molecular Robotics LaboratoryDepartment of RoboticsGraduate School of EngineeringTohoku UniversitySendai980‐8579Japan
| | - Stephen J. Ebbens
- Department of Chemical and Biological EngineeringUniversity of SheffieldMappin StreetSheffieldS1 3JDUK
| |
Collapse
|
34
|
O'Callaghan JA, Lee D, Hammer DA. Asymmetry-Enhanced Motion of Urease-Powered Micromotors from Double Emulsion-Templated Microcapsules. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37902731 DOI: 10.1021/acsami.3c10222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
Autonomous motion of enzyme-powered motors has important implications for drug delivery, cell-cell communication, and protocell engineering. Although many of these systems are inspired by the motion of biological cells, most of them lack key structural features, like micrometer-sized boundaries and aqueous compartments, and rely on bubble propulsion to generation motion. In this study, we use droplet microfluidics to generate large populations of cell-sized microcapsules with poly(lactic-co-glycolic acid) shells and functionalize their surfaces with the enzyme urease to drive their motion. We adjust the number of surface functional groups for urease conjugation by preparing microcapsules with two different surfactants, poly(vinyl alcohol) (PVA) and poly(ethylene-alt-maleic anhydride) (PEMA). We also tune the surface roughness of the microcapsules by varying the concentration of silica nanoparticles in the droplet middle phase. We find that PEMA plays a crucial role in increasing the grafting density of urease on the surface of smooth microcapsules, leading to active motion in the presence of urea. In addition, rough microcapsules prepared with PEMA and loaded with comparable amounts of urease move up to three times faster than their smooth counterparts, which we believe is due to an asymmetric distribution of urease on the surface, giving rise to a preferred direction of motion. Taken together, these results provide new insights into the role that various stabilizing agents play in the induction of motion by enzymatic motors prepared from microfluidics, which is a potentially powerful tool for future preparation of motile protocells in biomedicine.
Collapse
Affiliation(s)
- Jessica Ann O'Callaghan
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Daeyeon Lee
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Daniel A Hammer
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
35
|
Shrivastava A, Du Y, Adepu HK, Li R, Madhvacharyula AS, Swett AA, Choi JH. Motility of Synthetic Cells from Engineered Lipids. ACS Synth Biol 2023; 12:2789-2801. [PMID: 37729546 DOI: 10.1021/acssynbio.3c00271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Synthetic cells are artificial systems that resemble natural cells. Significant efforts have been made over the years to construct synthetic protocells that can mimic biological mechanisms and perform various complex processes. These include compartmentalization, metabolism, energy supply, communication, and gene reproduction. Cell motility is also of great importance, as nature uses elegant mechanisms for intracellular trafficking, immune response, and embryogenesis. In this review, we discuss the motility of synthetic cells made from lipid vesicles and relevant molecular mechanisms. Synthetic cell motion may be classified into surface-based or solution-based depending on whether it involves interactions with surfaces or movement in fluids. Collective migration behaviors have also been demonstrated. The swarm motion requires additional mechanisms for intercellular signaling and directional motility that enable communication and coordination among the synthetic vesicles. In addition, intracellular trafficking for molecular transport has been reconstituted in minimal cells with the help of DNA nanotechnology. These efforts demonstrate synthetic cells that can move, detect, respond, and interact. We envision that new developments in protocell motility will enhance our understanding of biological processes and be instrumental in bioengineering and therapeutic applications.
Collapse
Affiliation(s)
- Aishwary Shrivastava
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, Indiana 47907, United States
| | - Yancheng Du
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, Indiana 47907, United States
| | - Harshith K Adepu
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, Indiana 47907, United States
| | - Ruixin Li
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, Indiana 47907, United States
| | - Anirudh S Madhvacharyula
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, Indiana 47907, United States
| | - Alexander A Swett
- School of Mechanical Engineering, Purdue University, Neil Armstrong Hall of Engineering, 701 W. Stadium Avenue, West Lafayette, Indiana 47907, United States
| | - Jong Hyun Choi
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, Indiana 47907, United States
| |
Collapse
|
36
|
Rheem HB, Choi H, Yang S, Han S, Rhee SY, Jeong H, Lee KB, Lee Y, Kim IS, Lee H, Choi IS. Fugetaxis of Cell-in-Catalytic-Coat Nanobiohybrids in Glucose Gradients. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301431. [PMID: 37282761 DOI: 10.1002/smll.202301431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/09/2023] [Indexed: 06/08/2023]
Abstract
Manipulation and control of cell chemotaxis remain an underexplored territory despite vast potential in various fields, such as cytotherapeutics, sensors, and even cell robots. Herein is achieved the chemical control over chemotactic movement and direction of Jurkat T cells, as a representative model, by the construction of cell-in-catalytic-coat structures in single-cell nanoencapsulation. Armed with the catalytic power of glucose oxidase (GOx) in the artificial coat, the nanobiohybrid cytostructures, denoted as Jurkat[Lipo_GOx] , exhibit controllable, redirected chemotactic movement in response to d-glucose gradients, in the opposite direction to the positive-chemotaxis direction of naïve, uncoated Jurkat cells in the same gradients. The chemically endowed, reaction-based fugetaxis of Jurkat[Lipo_GOx] operates orthogonally and complementarily to the endogenous, binding/recognition-based chemotaxis that remains intact after the formation of a GOx coat. For instance, the chemotactic velocity of Jurkat[Lipo_GOx] can be adjusted by varying the combination of d-glucose and natural chemokines (CXCL12 and CCL19) in the gradient. This work offers an innovative chemical tool for bioaugmenting living cells at the single-cell level through the use of catalytic cell-in-coat structures.
Collapse
Affiliation(s)
- Hyeong Bin Rheem
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| | - Hyunwoo Choi
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| | - Seoin Yang
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| | - Sol Han
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| | - Su Yeon Rhee
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| | - Hyeongseop Jeong
- Division of Scientific Instrumentation & Management, Korea Basic Science Institute (KBSI), Cheongju, 28119, South Korea
| | - Kyung-Bok Lee
- Division of Scientific Instrumentation & Management, Korea Basic Science Institute (KBSI), Cheongju, 28119, South Korea
| | - Yeji Lee
- Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
- Chemical & Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea
| | - In-San Kim
- Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
- Chemical & Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea
| | - Hojae Lee
- Department of Chemistry, Hallym University, Chuncheon, 24252, South Korea
| | - Insung S Choi
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon, 34141, South Korea
| |
Collapse
|
37
|
Ghosh C, Ghosh S, Chatterjee A, Bera P, Mampallil D, Ghosh P, Das D. Dual enzyme-powered chemotactic cross β amyloid based functional nanomotors. Nat Commun 2023; 14:5903. [PMID: 37737223 PMCID: PMC10516904 DOI: 10.1038/s41467-023-41301-x] [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: 04/25/2022] [Accepted: 08/30/2023] [Indexed: 09/23/2023] Open
Abstract
Nanomotor chassis constructed from biological precursors and powered by biocatalytic transformations can offer important applications in the future, specifically in emergent biomedical techniques. Herein, cross β amyloid peptide-based nanomotors (amylobots) were prepared from short amyloid peptides. Owing to their remarkable binding capabilities, these soft constructs are able to host dedicated enzymes to catalyze orthogonal substrates for motility and navigation. Urease helps in powering the self-diffusiophoretic motion, while cytochrome C helps in providing navigation control. Supported by the simulation model, the design principle demonstrates the utilization of two distinct transport behaviours for two different types of enzymes, firstly enhanced diffusivity of urease with increasing fuel (urea) concentration and secondly, chemotactic motility of cytochrome C towards its substrate (pyrogallol). Dual catalytic engines allow the amylobots to be utilized for enhanced catalysis in organic solvent and can thus complement the technological applications of enzymes.
Collapse
Affiliation(s)
- Chandranath Ghosh
- Department of Chemical Sciences and Centre for Advanced Functional Materials, Indian Institute of Science Education and Research (IISER), Kolkata, Mohanpur, 741246, India
| | - Souvik Ghosh
- Department of Chemical Sciences and Centre for Advanced Functional Materials, Indian Institute of Science Education and Research (IISER), Kolkata, Mohanpur, 741246, India
| | - Ayan Chatterjee
- Department of Chemical Sciences and Centre for Advanced Functional Materials, Indian Institute of Science Education and Research (IISER), Kolkata, Mohanpur, 741246, India
| | - Palash Bera
- Tata Institute of Fundamental Research (TIFR), Hyderabad, Telangana, 500046, India
| | - Dileep Mampallil
- Department of Physics, Indian Institute of Science Education and Research (IISER) Tirupati, Mangalam, Andhra Pradesh, 517507, India
| | - Pushpita Ghosh
- School of Chemistry, Indian Institute of Science Education and Research (IISER), Thiruvananthapuram, Kerala, 695551, India
| | - Dibyendu Das
- Department of Chemical Sciences and Centre for Advanced Functional Materials, Indian Institute of Science Education and Research (IISER), Kolkata, Mohanpur, 741246, India.
| |
Collapse
|
38
|
Luo T, Yang H, Wang R, Pu Y, Cai Z, Zhao Y, Bi Q, Lu J, Jin R, Nie Y, Shuai X. Bifunctional Cascading Nanozymes Based on Carbon Dots Promotes Photodynamic Therapy by Regulating Hypoxia and Glycolysis. ACS NANO 2023; 17:16715-16730. [PMID: 37594768 DOI: 10.1021/acsnano.3c03169] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
Photodynamic therapy (PDT) still faces great challenges with suitable photosensitizers, oxygen supply, and reactive oxygen species (ROS) accumulation, especially in the tumor microenvironment, feathering hypoxia, and high glucose metabolism. Herein, a carbon dots (CDs)-based bifunctional nanosystem (MnZ@Au), acting as photosensitizer and nanozyme with cascading glucose oxidase (GOx)- and catalase (CAT)-like reactivity, was developed for improving hypoxia and regulating glucose metabolism to enhance PDT. The MnZ@Au was constructed using Mn-doped CDs (Mn-CDs) as a core and zeolitic imidazolate framework-8 (ZIF-8) as a shell to form a hybrid (MnZ), followed by anchoring ultrasmall Au nanoparticles (AuNPs) onto the surface of MnZ through the ion exchange and in situ reduction methods. MnZ@Au catalyzed glucose consumption and oxygen generation by cascading GOx- and CAT-like nanozyme reactions, which was further enhanced by its own photothermal properties. In vitro and in vivo studies also confirmed that MnZ@Au greatly improved CDs penetration, promoted ROS accumulation, and enhanced PDT efficacy, leading to efficient tumor growth inhibition in the breast tumor model. Besides, MnZ@Au enabled photoacoustic (PA) imaging to provide a mapping of Mn-CDs distribution and oxygen saturation, showing the real-time catalytic process of MnZ@Au in vivo. 18F-Fluorodeoxyglucose positron emission tomography (18F-FDG PET) imaging also validated the decreased glucose uptake in tumors treated by MnZ@Au. Therefore, the integrated design provided a promising strategy to utilize and regulate the tumor microenvironment, promote penetration, enhance PDT, and finally prevent tumor deterioration.
Collapse
Affiliation(s)
- Tianying Luo
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
| | - Huan Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
| | - Ruohan Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
| | - Yiyao Pu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
| | - Zhongyuan Cai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
| | - Yangyang Zhao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
| | - Qunjie Bi
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
| | - Jiao Lu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
| | - Yu Nie
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
39
|
Tian H, Ou J, Wang Y, Sun J, Gao J, Ye Y, Zhang R, Chen B, Wang F, Huang W, Li H, Liu L, Shao C, Xu Z, Peng F, Tu Y. Bladder microenvironment actuated proteomotors with ammonia amplification for enhanced cancer treatment. Acta Pharm Sin B 2023; 13:3862-3875. [PMID: 37719374 PMCID: PMC10501867 DOI: 10.1016/j.apsb.2023.02.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/17/2023] [Accepted: 02/17/2023] [Indexed: 03/05/2023] Open
Abstract
Enzyme-driven micro/nanomotors consuming in situ chemical fuels have attracted lots of attention for biomedical applications. However, motor systems composed by organism-derived organics that maximize the therapeutic efficacy of enzymatic products remain challenging. Herein, swimming proteomotors based on biocompatible urease and human serum albumin are constructed for enhanced antitumor therapy via active motion and ammonia amplification. By decomposing urea into carbon dioxide and ammonia, the designed proteomotors are endowed with self-propulsive capability, which leads to improved internalization and enhanced penetration in vitro. As a glutamine synthetase inhibitor, the loaded l-methionine sulfoximine further prevents the conversion of toxic ammonia into non-toxic glutamine in both tumor and stromal cells, resulting in local ammonia amplification. After intravesical instillation, the proteomotors achieve longer bladder retention and thus significantly inhibit the growth of orthotopic bladder tumor in vivo without adverse effects. We envision that the as-developed swimming proteomotors with amplification of the product toxicity may be a potential platform for active cancer treatment.
Collapse
Affiliation(s)
- Hao Tian
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Juanfeng Ou
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yong Wang
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Jia Sun
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Junbin Gao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yicheng Ye
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ruotian Zhang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Bin Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Fei Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Weichang Huang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Huaan Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Lu Liu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Chuxiao Shao
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, Central Laboratory of Lishui People's Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Zhili Xu
- Department of Ultrasound, Institute of Ultrasound in Musculoskeletal Sports Medicine, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Fei Peng
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yingfeng Tu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| |
Collapse
|
40
|
Huang W, Wen L, Tian H, Jiang J, Liu M, Ye Y, Gao J, Zhang R, Wang F, Li H, Shen L, Peng F, Tu Y. Self-Propelled Proteomotors with Active Cell-Free mtDNA Clearance for Enhanced Therapy of Sepsis-Associated Acute Lung Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301635. [PMID: 37518854 PMCID: PMC10520684 DOI: 10.1002/advs.202301635] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/10/2023] [Indexed: 08/01/2023]
Abstract
Acute lung injury (ALI) is a frequent and serious complication of sepsis with limited therapeutic options. Gaining insights into the inflammatory dysregulation that causes sepsis-associated ALI can help develop new therapeutic strategies. Herein, the crucial role of cell-free mitochondrial DNA (cf-mtDNA) in the regulation of alveolar macrophage activation during sepsis-associated ALI is identified. Most importantly, a biocompatible hybrid protein nanomotor (NM) composed of recombinant deoxyribonuclease I (DNase-I) and human serum albumin (HSA) via glutaraldehyde-mediated crosslinking is prepared to obtain an inhalable nanotherapeutic platform targeting pulmonary cf-mtDNA clearance. The synthesized DNase-I/HSA NMs are endowed with self-propulsive capability and demonstrate superior performances in stability, DNA hydrolysis, and biosafety. Pulmonary delivery of DNase-I/HSA NMs effectively eliminates cf-mtDNAs in the lungs, and also improves sepsis survival by attenuating pulmonary inflammation and lung injury. Therefore, pulmonary cf-mtDNA clearance strategy using DNase-I/HSA NMs is considered to be an attractive approach for sepsis-associated ALI.
Collapse
Affiliation(s)
- Weichang Huang
- Department of Critical Care MedicineDongguan Institute of Respiratory and Critical Care MedicineAffiliated Dongguan HospitalSouthern Medical UniversityDongguan523059China
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Lihong Wen
- Department of Plastic SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Hao Tian
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Jiamiao Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Meihuan Liu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Yicheng Ye
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Junbin Gao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Ruotian Zhang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Fei Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Huaan Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Lihan Shen
- Department of Critical Care MedicineDongguan Institute of Respiratory and Critical Care MedicineAffiliated Dongguan HospitalSouthern Medical UniversityDongguan523059China
| | - Fei Peng
- School of Materials Science and EngineeringSun Yat‐Sen UniversityGuangzhou510275China
| | - Yingfeng Tu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| |
Collapse
|
41
|
Liu S, Xu D, Chen J, Peng N, Ma T, Liang F. Nanozymatic magnetic nanomotors for enhancing photothermal therapy and targeting intracellular SERS sensing. NANOSCALE 2023; 15:12944-12953. [PMID: 37486742 DOI: 10.1039/d3nr02739b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Self-propelled micro/nanomotors (MNMs) have emerged as promising tools for biomedical applications owing to their active and controllable movement, which is achieved by converting energy derived from chemical reactions or external physical fields into mechanical forces. However, it remains a challenge to develop all-in-one MNMs that integrate multiple bio-friendly engines and biomedical functions. In this study, we present a nanozymatic magnetic nanomotor capable of self-propulsion, driven by its intrinsic engines, and possessing inherent biomedical functions. The nanomotors with a core-island structure are fabricated by a general scalable chemistry synthesis approach. The core of the nanomotors is magnetic Fe3O4 nanoparticles, while the surrounding islands consist of Au nanostars. Such components naturally equip the nanomotors with the dual engine of the magnetic core and gold nanozyme. In addition, the localized surface plasmon resonance (LSPR) effect of the Au nanostar imparts the nanomotors with favourable photothermal conversion and surface-enhanced Raman scattering (SERS) properties. The nanomotors exhibit glucose concentration-dependent motion behavior of enhanced diffusion, leading to improved endocytosis for enhanced photothermal treatment. When exposed to a magnetic field, the nanomotors demonstrate both directional locomotion towards target cells and up-and-down oscillatory movement, enabling the efficient gathering of intracellular analytes for SERS sensing. To conclude, the as-prepared nanomotors represent an active and controllable nanoplatform with a simple structure and are naturally equipped with dual engines and dual biomedical functions, providing new perspectives to the development of all-in-one biomedical MNMs.
Collapse
Affiliation(s)
- Shimi Liu
- The State Key Laboratory of Refractories and Metallurgy, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China.
- Improve-WUST Joint Laboratory of Advanced Technology for Point-of-Care Testing and Precision Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Dandan Xu
- The State Key Laboratory of Refractories and Metallurgy, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China.
- Improve-WUST Joint Laboratory of Advanced Technology for Point-of-Care Testing and Precision Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Junling Chen
- The State Key Laboratory of Refractories and Metallurgy, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China.
- Improve-WUST Joint Laboratory of Advanced Technology for Point-of-Care Testing and Precision Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Na Peng
- The State Key Laboratory of Refractories and Metallurgy, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China.
| | - Tao Ma
- The State Key Laboratory of Refractories and Metallurgy, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China.
- Improve-WUST Joint Laboratory of Advanced Technology for Point-of-Care Testing and Precision Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Feng Liang
- The State Key Laboratory of Refractories and Metallurgy, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China.
- Improve-WUST Joint Laboratory of Advanced Technology for Point-of-Care Testing and Precision Medicine, Wuhan University of Science and Technology, Wuhan 430081, China
| |
Collapse
|
42
|
Fraire JC, Guix M, Hortelao AC, Ruiz-González N, Bakenecker AC, Ramezani P, Hinnekens C, Sauvage F, De Smedt SC, Braeckmans K, Sánchez S. Light-Triggered Mechanical Disruption of Extracellular Barriers by Swarms of Enzyme-Powered Nanomotors for Enhanced Delivery. ACS NANO 2023; 17:7180-7193. [PMID: 37058432 PMCID: PMC10134497 DOI: 10.1021/acsnano.2c09380] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Targeted drug delivery depends on the ability of nanocarriers to reach the target site, which requires the penetration of different biological barriers. Penetration is usually low and slow because of passive diffusion and steric hindrance. Nanomotors (NMs) have been suggested as the next generation of nanocarriers in drug delivery due to their autonomous motion and associated mixing hydrodynamics, especially when acting collectively as a swarm. Here, we explore the concept of enzyme-powered NMs designed as such that they can exert disruptive mechanical forces upon laser irradiation. The urease-powered motion and swarm behavior improve translational movement compared to passive diffusion of state-of-the-art nanocarriers, while optically triggered vapor nanobubbles can destroy biological barriers and reduce steric hindrance. We show that these motors, named Swarm 1, collectively displace through a microchannel blocked with type 1 collagen protein fibers (barrier model), accumulate onto the fibers, and disrupt them completely upon laser irradiation. We evaluate the disruption of the microenvironment induced by these NMs (Swarm 1) by quantifying the efficiency by which a second type of fluorescent NMs (Swarm 2) can move through the cleared microchannel and be taken up by HeLa cells at the other side of the channel. Experiments showed that the delivery efficiency of Swarm 2 NMs in a clean path was increased 12-fold in the presence of urea as fuel compared to when no fuel was added. When the path was blocked with the collagen fibers, delivery efficiency dropped considerably and only depicted a 10-fold enhancement after pretreatment of the collagen-filled channel with Swarm 1 NMs and laser irradiation. The synergistic effect of active motion (chemically propelled) and mechanical disruption (light-triggered nanobubbles) of a biological barrier represents a clear advantage for the improvement of therapies which currently fail due to inadequate passage of drug delivery carriers through biological barriers.
Collapse
Affiliation(s)
- Juan C. Fraire
- Institute
for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona Spain
- Laboratory
for General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical
Sciences, Ghent University, 9000 Ghent, Belgium
| | - Maria Guix
- Institute
for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona Spain
- Departament
de Ciéncia dels Materials i Química Física,
Institut de Química Teòrica i Computacional Barcelona, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Ana C. Hortelao
- Institute
for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona Spain
| | - Noelia Ruiz-González
- Institute
for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona Spain
| | - Anna C. Bakenecker
- Institute
for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona Spain
| | - Pouria Ramezani
- Laboratory
for General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical
Sciences, Ghent University, 9000 Ghent, Belgium
| | - Charlotte Hinnekens
- Laboratory
for General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical
Sciences, Ghent University, 9000 Ghent, Belgium
| | - Félix Sauvage
- Laboratory
for General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical
Sciences, Ghent University, 9000 Ghent, Belgium
| | - Stefaan C. De Smedt
- Laboratory
for General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical
Sciences, Ghent University, 9000 Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory
for General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical
Sciences, Ghent University, 9000 Ghent, Belgium
| | - Samuel Sánchez
- Institute
for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028 Barcelona Spain
- Catalan
Institute for Research and Advanced Studies (ICREA), Passeig de Lluís Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
43
|
Jin H, Cui J, Zhan W. Enzymatic Janus Liposome Micromotors. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:4198-4206. [PMID: 36893357 DOI: 10.1021/acs.langmuir.3c00335] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
A liposome-based micromotor system that utilizes regional enzymatic conversion and gas generation to achieve directional motion in water is presented. Constituted mainly of a low-melting lipid and a high-melting lipid together with cholesterol, these liposomes maintain stable Janus configuration at room temperature as a result of lipid liquid-liquid phase separation. Local placement of enzymes such as horseradish peroxidase is realized via affinity binding between avidin and biotin, the latter as a lipid conjugate sorted specifically into one domain of these Janus liposomes as a minor component. In the presence of the substrate, hydrogen peroxide, these enzyme-decorated Janus liposomes undergo directional motion, yielding velocities exceeding thermal diffusion by three folds in some cases. Experimental details on liposome size control, motor assembly, and substrate distribution are presented; effects of key experimental factors on liposome motion, such as substrate concentration and liposome Janus ratio, are also examined. This work thus provides a viable approach to building asymmetrical lipid-assembled, enzyme-attached colloids and, in addition, stresses the importance of asymmetry in achieving particle directional motion.
Collapse
Affiliation(s)
- Hui Jin
- Department of Chemistry and Biochemistry, Auburn University, Auburn, Alabama 36849, United States
| | - Jinyan Cui
- Department of Chemistry and Biochemistry, Auburn University, Auburn, Alabama 36849, United States
| | - Wei Zhan
- Department of Chemistry and Biochemistry, Auburn University, Auburn, Alabama 36849, United States
| |
Collapse
|
44
|
Meng J, Zhang P, Liu Q, Ran P, Xie S, Wei J, Li X. Pyroelectric Janus nanomotors for synergistic electrodynamic-photothermal-antibiotic therapies of bacterial infections. Acta Biomater 2023; 162:20-31. [PMID: 36931421 DOI: 10.1016/j.actbio.2023.03.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/25/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
Antibacterial electrotherapy is currently activated by external electric field or self-powered generators, but usually needs complicated power management circuits. Herein, near-infrared illumination (NIR) of pyroelectric nanoparticles (NPs) produces a built-in electric field to address the effectiveness and safety concerns in the antibacterial treatment. Janus tBT@PDA NPs were obtained by capping polydopamine (PDA) on tetragonal BaTiO3 (tBT) NPs through defining the polymerization time, followed by ciprofloxacin (CIP) loading on the PDA caps to fabricate Janus tBT@PDA-Cip NPs. NIR illumination of PDA caps creates temperature variations on tBT NPs to generate photothermal and pyroelectric effects. Finite element simulation reveals a pyroelectric potential of over 1 V and sufficient reactive oxygen species (ROS) are produced to exhibit pyroelectric dynamic therapy (PEDT). The elevated temperature on one side of the Janus NPs produces thermophoretic force to drive NP motion, which enhances interactions with bacteria and overcomes limitations in the short action distance and lifespan of ROS. The pyroelectric field accelerates CIP release through weakening the π-π stacking and electrostatic interaction with PDA and also interrupts membrane potentials of bacteria to enhance CIP invasion into bacteria. The synergistic antibacterial effect of pyroelectric tBT@PDA-Cip NPs causes the fully recovery of S. aureus-infected skin wounds and regeneration of intact epidermis, blood vessels and hair follicles, while no obvious pathological change or inflammatory lesion is detected in the major organs. Thus, the pyroelectric Janus nanomotors demonstrate synergistic PEDT/photothermal/antibiotic effects to enhance antibacterial efficacy while avoiding the necessity of excessive heat, ROS and antibiotic doses. STATEMENT OF SIGNIFICANCE: Antibacterial treatment is challenged by antibiotics-derived side effects and the evolution of resistant strains. Phototherapy is commonly associated with excessive heat and oxidative stress, and their combinations with other agents are especially encouraged to strengthen antibacterial efficacy while alleviating the associated side effects. Electric field is another activator to generate antibacterial abilities, but usually requires complicated power management and bulk electrodes, making it inconvenient in a biological setup. To address these challenges, we propose a strategy to generate microelectric field on nanoparticles themselves and achieve synergistic electrodynamic-photothermal-antibiotic therapies. The pyroelectric effect weakens interactions between nanoparticles and antibiotics to accelerate drug release, and the built-in pyroelectric field increases membrane fluidity to enhance bacterial uptake of antibiotics.
Collapse
Affiliation(s)
- Jie Meng
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, PR China; Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Peng Zhang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Qingjie Liu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Pan Ran
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Shuang Xie
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, PR China; Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Junwu Wei
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, PR China; Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Xiaohong Li
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, PR China; Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China.
| |
Collapse
|
45
|
Fusi AD, Li Y, Llopis‐Lorente A, Patiño T, van Hest JCM, Abdelmohsen LKEA. Achieving Control in Micro-/Nanomotor Mobility. Angew Chem Int Ed Engl 2023; 62:e202214754. [PMID: 36413146 PMCID: PMC10107182 DOI: 10.1002/anie.202214754] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/23/2022]
Abstract
Unprecedented opportunities exist for the generation of advanced nanotechnologies based on synthetic micro/nanomotors (MNMs), such as active transport of medical agents or the removal of pollutants. In this regard, great efforts have been dedicated toward controlling MNM motion (e.g., speed, directionality). This was generally performed by precise engineering and optimizing of the motors' chassis, engine, powering mode (i.e., chemical or physical), and mechanism of motion. Recently, new insights have emerged to control motors mobility, mainly by the inclusion of different modes that drive propulsion. With high degree of synchronization, these modes work providing the required level of control. In this Minireview, we discuss the diverse factors that impact motion; these include MNM morphology, modes of mobility, and how control over motion was achieved. Moreover, we highlight the main limitations that need to be overcome so that such motion control can be translated into real applications.
Collapse
Affiliation(s)
- Alexander D. Fusi
- Departments of Chemical Engineering and Chemistry, and Biomedical EngineeringInstitute for Complex Molecular SystemsTechnische Universiteit EindhovenHet Kranenveld 145612AZ EindhovenThe Netherlands
| | - Yudong Li
- Departments of Chemical Engineering and Chemistry, and Biomedical EngineeringInstitute for Complex Molecular SystemsTechnische Universiteit EindhovenHet Kranenveld 145612AZ EindhovenThe Netherlands
| | - A. Llopis‐Lorente
- Departments of Chemical Engineering and Chemistry, and Biomedical EngineeringInstitute for Complex Molecular SystemsTechnische Universiteit EindhovenHet Kranenveld 145612AZ EindhovenThe Netherlands
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN)Institute of Molecular Recognition and Technological Development (IDM)Universitat Politècnica de ValènciaCamino de Vera s/n46022ValenciaSpain
| | - Tania Patiño
- Departments of Chemical Engineering and Chemistry, and Biomedical EngineeringInstitute for Complex Molecular SystemsTechnische Universiteit EindhovenHet Kranenveld 145612AZ EindhovenThe Netherlands
| | - Jan C. M. van Hest
- Departments of Chemical Engineering and Chemistry, and Biomedical EngineeringInstitute for Complex Molecular SystemsTechnische Universiteit EindhovenHet Kranenveld 145612AZ EindhovenThe Netherlands
| | - Loai K. E. A. Abdelmohsen
- Departments of Chemical Engineering and Chemistry, and Biomedical EngineeringInstitute for Complex Molecular SystemsTechnische Universiteit EindhovenHet Kranenveld 145612AZ EindhovenThe Netherlands
| |
Collapse
|
46
|
Wang Y, Shen J, Handschuh-Wang S, Qiu M, Du S, Wang B. Microrobots for Targeted Delivery and Therapy in Digestive System. ACS NANO 2023; 17:27-50. [PMID: 36534488 DOI: 10.1021/acsnano.2c04716] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Untethered miniature robots enable targeted delivery and therapy deep inside the gastrointestinal tract in a minimally invasive manner. By combining actuation systems and imaging tools, significant progress has been made toward the development of functional microrobots. These robots can be actuated by external fields and fuels while featuring real-time tracking feedback toward certain regions and can perform the therapeutic process by rational exertion of the local environment of the gastrointestinal tract (e.g., pH, enzyme). Compared with conventional surgical tools, such as endoscopic devices and catheters, miniature robots feature minimally invasive diagnosis and treatment, multifunctionality, high safety and adaptivity, embodied intelligence, and easy access to tortuous and narrow lumens. In addition, the active motion of microrobots enhances local penetration and retention of drugs in tissues compared to common passive oral drug delivery. Based on the dissimilar microenvironments in the various sections of the gastrointestinal tract, this review introduces the advances of miniature robots for minimally invasive targeted delivery and therapy of diseases along the gastrointestinal tract. The imaging modalities for the tracking and their application scenarios are also discussed. We finally evaluate the challenges and barriers that retard their applications and hint on future research directions in this field.
Collapse
Affiliation(s)
- Yun Wang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen518055, P.R. China
| | - Jie Shen
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen518036, P.R. China
| | - Stephan Handschuh-Wang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen518055, P.R. China
| | - Ming Qiu
- Department of Neurosurgery, South China Hospital of Shenzhen University, Shenzhen518111, P.R. China
| | - Shiwei Du
- Department of Neurosurgery, South China Hospital of Shenzhen University, Shenzhen518111, P.R. China
| | - Ben Wang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen518055, P.R. China
| |
Collapse
|
47
|
Zhang D, Gorochowski TE, Marucci L, Lee HT, Gil B, Li B, Hauert S, Yeatman E. Advanced medical micro-robotics for early diagnosis and therapeutic interventions. Front Robot AI 2023; 9:1086043. [PMID: 36704240 PMCID: PMC9871318 DOI: 10.3389/frobt.2022.1086043] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/15/2022] [Indexed: 01/12/2023] Open
Abstract
Recent technological advances in micro-robotics have demonstrated their immense potential for biomedical applications. Emerging micro-robots have versatile sensing systems, flexible locomotion and dexterous manipulation capabilities that can significantly contribute to the healthcare system. Despite the appreciated and tangible benefits of medical micro-robotics, many challenges still remain. Here, we review the major challenges, current trends and significant achievements for developing versatile and intelligent micro-robotics with a focus on applications in early diagnosis and therapeutic interventions. We also consider some recent emerging micro-robotic technologies that employ synthetic biology to support a new generation of living micro-robots. We expect to inspire future development of micro-robots toward clinical translation by identifying the roadblocks that need to be overcome.
Collapse
Affiliation(s)
- Dandan Zhang
- Department of Engineering Mathematics, University of Bristol, Bristol, United Kingdom
- Bristol Robotics Laboratory, Bristol, United Kingdom
| | - Thomas E. Gorochowski
- School of Biological Sciences, University of Bristol, Bristol, United Kingdom
- BrisEngBio, University of Bristol, Bristol, United Kingdom
| | - Lucia Marucci
- Department of Engineering Mathematics, University of Bristol, Bristol, United Kingdom
- School of Biological Sciences, University of Bristol, Bristol, United Kingdom
- BrisEngBio, University of Bristol, Bristol, United Kingdom
| | - Hyun-Taek Lee
- Department of Mechanical Engineering, Inha University, Incheon, South Korea
| | - Bruno Gil
- Department of Electrical and Electronic Engineering, Imperial College London, London, United Kingdom
| | - Bing Li
- The Institute for Materials Discovery, University College London, London, United Kingdom
- Department of Brain Science, Imperial College London, London, United Kingdom
- Care Research & Technology Centre, UK Dementia Research Institute, Imperial College London, London, United Kingdom
| | - Sabine Hauert
- Department of Engineering Mathematics, University of Bristol, Bristol, United Kingdom
- Bristol Robotics Laboratory, Bristol, United Kingdom
- BrisEngBio, University of Bristol, Bristol, United Kingdom
| | - Eric Yeatman
- Department of Electrical and Electronic Engineering, Imperial College London, London, United Kingdom
| |
Collapse
|
48
|
Li T, Liu Z, Hu J, Chen L, Chen T, Tang Q, Yu B, Zhao B, Mao C, Wan M. A Universal Chemotactic Targeted Delivery Strategy for Inflammatory Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2206654. [PMID: 36122571 DOI: 10.1002/adma.202206654] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/13/2022] [Indexed: 06/15/2023]
Abstract
Above 50% of deaths can be attributed to chronic inflammatory diseases; thus, the construction of drug delivery systems based on effective interaction of inflammatory factors with chemotactic nanoparticles is meaningful. Herein, a zwitterion-based artificial chemotactic nanomotor is proposed for universal precise targeting strategy in vivo, where the high level of reactive oxygen species (ROS) and inducible nitric oxide synthase (iNOS) in inflammatory sites are used as a chemoattractant. Multidimensional static models, dynamic models, and in vivo models are established to evaluate chemotactic performance. The results show that the upregulated ROS and iNOS can induce the chemotaxis of nanomotors to diseased tissues in inflammation-related disease models. Further, mesoscale hydrodynamics simulations are performed to explain the chemotactic behavior of the nanomotors. Such a chemotactic delivery strategy is expected to improve delivery efficiency and may be applicable to a variety of inflammatory diseases.
Collapse
Affiliation(s)
- Ting Li
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Zhiyong Liu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Jinglei Hu
- Kuang Yaming Honors School, Nanjing University, Nanjing, 210023, China
| | - Lin Chen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Tiantian Chen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Qianqian Tang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Bixia Yu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Bo Zhao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| |
Collapse
|
49
|
Xu L, Zhang K, Ma X, Li Y, Jin Y, Liang C, Wang Y, Duan W, Zhang H, Zhang Z, Shi J, Liu J, Wang Y, Li W. Boosting cisplatin chemotherapy by nanomotor-enhanced tumor penetration and DNA adducts formation. J Nanobiotechnology 2022; 20:429. [PMID: 36175999 PMCID: PMC9523964 DOI: 10.1186/s12951-022-01622-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
Despite many nano-based strategies devoted to delivering cisplatin for tumor therapy, its clinical benefits are compromised by poor tissue penetration and limited DNA adducts formation of the drug. Herein, a cisplatin loading nanomotor based janus structured Ag-polymer is developed for cisplatin delivery of deeper tissue and increased DNA adducts formation. The nanomotor displayed a self‐propelled tumor penetration fueled by hydrogen peroxide (H2O2) in tumor tissues, which is catalytically decomposed into a large amount of oxygen bubbles by Ag nanoparticles (NPs). Notably, cisplatin could elevate the intracellular H2O2 level through cascade reactions, further promote the degradation of Ag NPs accompanied with the Ag+ release, which could downregulate intracellular Cl− through the formation of AgCl precipitate, thereby enhancing cisplatin dechlorination and Pt–DNA formation. Moreover, polymer can also inhibit the activity of ALKBH2 (a Fe2+-dependent DNA repair enzyme) by chelating intracellular Fe2+ to increase the proportion of irreparable Pt–DNA cross-links. It is found that deep tissue penetration, as well as the increased formation and maintenance of Pt–DNA adducts induced by the nanomotor afford 80% of tumor growth inhibition with negligible toxicity. This work provides an important perspective of resolving chemotherapeutic barriers for boosting cisplatin therapy.
Collapse
Affiliation(s)
- Lihua Xu
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Center for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xing Ma
- School of Materials Science and Engineering & Flexible Printed Electronic Technology Center, Harbin Institute of Technology (Shenzhen), Shenzhen, 518055, China
| | - Yingying Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yajie Jin
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Chenglin Liang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yong Wang
- School of Materials Science and Engineering & Flexible Printed Electronic Technology Center, Harbin Institute of Technology (Shenzhen), Shenzhen, 518055, China
| | - Wendi Duan
- School of Materials Science and Engineering & Flexible Printed Electronic Technology Center, Harbin Institute of Technology (Shenzhen), Shenzhen, 518055, China
| | - Hongling Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Junjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yunlong Wang
- Henan Bioengineering Research Center, Zhengzhou, 450000, Henan, China.
| | - Wentao Li
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
50
|
Abstract
Micro-/nanorobots (MNRs) can be autonomously propelled on demand in complex biological environments and thus may bring revolutionary changes to biomedicines. Fluorescence has been widely used in real-time imaging, chemo-/biosensing, and photo-(chemo-) therapy. The integration of MNRs with fluorescence generates fluorescent MNRs with unique advantages of optical trackability, on-the-fly environmental sensitivity, and targeting chemo-/photon-induced cytotoxicity. This review provides an up-to-date overview of fluorescent MNRs. After the highlighted elucidation about MNRs of various propulsion mechanisms and the introductory information on fluorescence with emphasis on the fluorescent mechanisms and materials, we systematically illustrate the design and preparation strategies to integrate MNRs with fluorescent substances and their biomedical applications in imaging-guided drug delivery, intelligent on-the-fly sensing and photo-(chemo-) therapy. In the end, we summarize the main challenges and provide an outlook on the future directions of fluorescent MNRs. This work is expected to attract and inspire researchers from different communities to advance the creation and practical application of fluorescent MNRs on a broad horizon.
Collapse
Affiliation(s)
- Manyi Yang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, P. R. China
| | - Xia Guo
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, P. R. China
| | - Fangzhi Mou
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, P. R. China
| | - Jianguo Guan
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, P. R. China
| |
Collapse
|