51
|
Goswami R, Arya RK, Sharma S, Dutta B, Stamov DR, Zhu X, Rahaman SO. Mechanosensing by TRPV4 mediates stiffness-induced foreign body response and giant cell formation. Sci Signal 2021; 14:eabd4077. [PMID: 34726952 PMCID: PMC9976933 DOI: 10.1126/scisignal.abd4077] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Implantation of biomaterials or devices into soft tissue often leads to the development of the foreign body response (FBR), an inflammatory condition that can cause implant failure, tissue injury, and death of the patient. Macrophages accumulate and fuse to generate destructive foreign body giant cells (FBGCs) at the tissue-implant interface, leading to the development of fibrous scar tissue around the implant that is generated by myofibroblasts. We previously showed that the FBR in vivo and FBGC formation in vitro require transient receptor potential vanilloid 4 (TRPV4), a mechanosensitive ion channel. Here, we report that TRPV4 was required specifically for the FBR induced by implant stiffness independently of biochemical cues and for intracellular stiffening that promotes FBGC formation in vitro. TRPV4 deficiency reduced collagen deposition and the accumulation of macrophages, FBGCs, and myofibroblasts at stiff, but not soft, implants in vivo and inhibited macrophage-induced differentiation of wild-type fibroblasts into myofibroblasts in vitro. Atomic force microscopy demonstrated that TRPV4 was required for implant-adjacent tissue stiffening in vivo and for cytoskeletal remodeling and intracellular stiffening induced by fusogenic cytokines in vitro. Together, these data suggest a mechanism whereby a reciprocal functional interaction between TRPV4 and substrate stiffness leads to cytoskeletal remodeling and cellular force generation to promote FBGC formation during the FBR.
Collapse
Affiliation(s)
- Rishov Goswami
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - Rakesh K. Arya
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - Shweta Sharma
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - Bidisha Dutta
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - Dimitar R. Stamov
- JPK BioAFM Business, Nano Surfaces Division, Bruker Nano GmbH, Am Studio 2D, 12489 Berlin, Germany
| | - Xiaoping Zhu
- Department of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA
| | - Shaik O. Rahaman
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA.,Corresponding author.:
| |
Collapse
|
52
|
Teunissen AJP, Burnett ME, Prévot G, Klein ED, Bivona D, Mulder WJM. Embracing nanomaterials' interactions with the innate immune system. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1719. [PMID: 33847441 PMCID: PMC8511354 DOI: 10.1002/wnan.1719] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/12/2021] [Accepted: 03/21/2021] [Indexed: 12/17/2022]
Abstract
Immunotherapy has firmly established itself as a compelling avenue for treating disease. Although many clinically approved immunotherapeutics engage the adaptive immune system, therapeutically targeting the innate immune system remains much less explored. Nanomedicine offers a compelling opportunity for innate immune system engagement, as many nanomaterials inherently interact with myeloid cells (e.g., monocytes, macrophages, neutrophils, and dendritic cells) or can be functionalized to target their cell-surface receptors. Here, we provide a perspective on exploiting nanomaterials for innate immune system regulation. We focus on specific nanomaterial design parameters, including size, form, rigidity, charge, and surface decoration. Furthermore, we examine the potential of high-throughput screening and machine learning, while also providing recommendations for advancing the field. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Abraham J. P. Teunissen
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Marianne E. Burnett
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Geoffrey Prévot
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Emma D. Klein
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Daniel Bivona
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Willem J. M. Mulder
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
- Laboratory of Chemical Biology, Department of Biochemical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
53
|
Yao D, Qiao F, Song C, Lv Y. Matrix stiffness regulates bone repair by modulating 12-lipoxygenase-mediated early inflammation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112359. [PMID: 34474906 DOI: 10.1016/j.msec.2021.112359] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/04/2021] [Accepted: 08/04/2021] [Indexed: 12/21/2022]
Abstract
Lipid metabolism in macrophages has been increasingly emphasized in exerting an anti-inflammatory effect and accelerating fracture healing. 12-lipoxygenase (12-LOX) is expressed in several cell types, including macrophages, and oxidizes polyunsaturated fatty acids (PUFAs) to generate both pro- and anti-inflammatory lipid mediators, of which the n-3 PUFAs play an important part in tissue homeostasis/fibrosis. Although mechanical factor regulates the lipid metabolic axis of inflammatory cells, specifically matrix stiffness influences macrophages metabolic responses, little is known about how matrix stiffness affects the 12-LOX-mediated early inflammation in bone repair. In the present study, demineralized bone matrix (DBM) scaffolds with different matrix stiffness were constructed by controlling the duration of decalcification (0 h (control), 1 h (high), 12 h (medium), and 5 d (low)) to repair the defected rat skull. The expression of inflammatory cytokines and macrophages polarization were analyzed. The lipid metabolites and lipid mediators' biosynthesis by matrix stiffness-regulated were further detected. The results showed that the low matrix stiffness could polarize macrophages into an anti-inflammatory phenotype, promote the expression of anti-inflammatory cytokines and specialized pro-resolving lipid mediators (SPMs) biosynthesis beneficial for the osteogenesis of mesenchymal stem cells (MSCs). After treated with ML355, the expression of anti-inflammatory cytokines/proteins and SPMs biosynthesis in macrophages cultured on low-matrix stiffness scaffolds were repressed, and there were almost no statistical differences among all groups. Findings from this study support that matrix stiffness regulates bone repair by modulating 12-LOX-mediated early inflammation, which suggest a direct mechanical impact of matrix stiffness on macrophages lipid metabolism and provide a new insight into the clinical application of SPMs for bone regeneration.
Collapse
Affiliation(s)
- Dongdong Yao
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Fangyu Qiao
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Chenchen Song
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Yonggang Lv
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China.
| |
Collapse
|
54
|
Zhang M, Gao S, Yang D, Fang Y, Lin X, Jin X, Liu Y, Liu X, Su K, Shi K. Influencing factors and strategies of enhancing nanoparticles into tumors in vivo. Acta Pharm Sin B 2021; 11:2265-2285. [PMID: 34522587 PMCID: PMC8424218 DOI: 10.1016/j.apsb.2021.03.033] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/05/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023] Open
Abstract
The administration of nanoparticles (NPs) first faces the challenges of evading renal filtration and clearance of reticuloendothelial system (RES). After that, NPs infiltrate through the expanded endothelial space and penetrated the dense stroma of tumor microenvironment to tumor cells. As long as possible to prolong the time of NPs remaining in tumor tissue, NPs release active agent and induce pharmacological action. This review provides a comprehensive summary of the physical and chemical properties of NPs and the influence of various biological factors in tumor microenvironment, and discusses how to improve the final efficacy through adjusting the characteristics and structure of NPs. Perspectives and future directions are also provided.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Kai Shi
- Corresponding author. Tel./fax: +86 24 43520557.
| |
Collapse
|
55
|
The Elucidation of the Molecular Mechanism of the Extrusion Process. MATERIALS 2021; 14:ma14154278. [PMID: 34361472 PMCID: PMC8348501 DOI: 10.3390/ma14154278] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/27/2021] [Accepted: 07/27/2021] [Indexed: 02/02/2023]
Abstract
Extrusion is a popular method for producing homogenous population of unilamellar liposomes. The technique relies on forcing a lipid suspension through cylindrical pores in a polycarbonate membrane. The quantification of the extrusion and/or recalibration processes make possible the acquisition of experimental data, which can be correlated with the mechanical properties of the lipid bilayer. In this work, the force needed for the extrusion process was correlated with the mechanical properties of a lipid bilayer derived from other experiments. Measurements were performed using a home-made dedicated device capable of maintaining a stable volumetric flux of a liposome suspension through well-defined pores and to continuously measure the extrusion force. Based on the obtained results, the correlation between the lipid bilayer bending rigidity and extrusion force was derived. Specifically, it was found that the bending rigidity of liposomes formed from well-defined lipid mixtures agrees with data obtained by others using flicker-noise spectroscopy or micromanipulation. The other issue addressed in the presented studies was the identification of molecular mechanisms leading to the formation of unilamellar vesicles in the extrusion process. Finally, it was demonstrated that during the extrusion, lipids are not exchanged between vesicles, i.e., vesicles can divide but no membrane fusion or lipid exchange between bilayers was detected.
Collapse
|
56
|
Vasse GF, Nizamoglu M, Heijink IH, Schlepütz M, van Rijn P, Thomas MJ, Burgess JK, Melgert BN. Macrophage-stroma interactions in fibrosis: biochemical, biophysical, and cellular perspectives. J Pathol 2021; 254:344-357. [PMID: 33506963 PMCID: PMC8252758 DOI: 10.1002/path.5632] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/18/2020] [Accepted: 01/08/2021] [Indexed: 12/16/2022]
Abstract
Fibrosis results from aberrant wound healing and is characterized by an accumulation of extracellular matrix, impairing the function of an affected organ. Increased deposition of extracellular matrix proteins, disruption of matrix degradation, but also abnormal post-translational modifications alter the biochemical composition and biophysical properties of the tissue microenvironment - the stroma. Macrophages are known to play an important role in wound healing and tissue repair, but the direct influence of fibrotic stroma on macrophage behaviour is still an under-investigated element in the pathogenesis of fibrosis. In this review, the current knowledge on interactions between macrophages and (fibrotic) stroma will be discussed from biochemical, biophysical, and cellular perspectives. Furthermore, we provide future perspectives with regard to how macrophage-stroma interactions can be examined further to ultimately facilitate more specific targeting of these interactions in the treatment of fibrosis. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Gwenda F Vasse
- University of Groningen, University Medical Center GroningenBiomedical Engineering Department‐FB40GroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials ScienceGroningenThe Netherlands
- University of Groningen, Department of Molecular PharmacologyGroningen Research Institute for PharmacyGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
| | - Mehmet Nizamoglu
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of Pathology and Medical BiologyGroningenThe Netherlands
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of Pathology and Medical BiologyGroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of PulmonologyGroningenThe Netherlands
| | - Marco Schlepütz
- Immunology & Respiratory Diseases ResearchBoehringer Ingelheim Pharma GmbH & Co KGBiberach an der RissGermany
| | - Patrick van Rijn
- University of Groningen, University Medical Center GroningenBiomedical Engineering Department‐FB40GroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials ScienceGroningenThe Netherlands
| | - Matthew J Thomas
- Immunology & Respiratory Diseases ResearchBoehringer Ingelheim Pharma GmbH & Co KGBiberach an der RissGermany
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials ScienceGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of Pathology and Medical BiologyGroningenThe Netherlands
| | - Barbro N Melgert
- University of Groningen, Department of Molecular PharmacologyGroningen Research Institute for PharmacyGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
| |
Collapse
|
57
|
Freeman S, Grinstein S. Promoters and Antagonists of Phagocytosis: A Plastic and Tunable Response. Annu Rev Cell Dev Biol 2021; 37:89-114. [PMID: 34152790 DOI: 10.1146/annurev-cellbio-120219-055903] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recent observations indicate that, rather than being an all-or-none response, phagocytosis is finely tuned by a host of developmental and environmental factors. The expression of key phagocytic determinants is regulated via transcriptional and epigenetic means that confer memory on the process. Membrane traffic, the cytoskeleton, and inside-out signaling control the activation of phagocytic receptors and their ability to access their targets. An exquisite extra layer of complexity is introduced by the coexistence of distinct "eat-me" and "don't-eat-me" signals on targets and of corresponding "eat" and "don't-eat" receptors on the phagocyte surface. Moreover, assorted physical barriers constitute "don't-come-close-to-me" hurdles that obstruct the engagement of ligands by receptors. The expression, mobility, and accessibility of all these determinants can be modulated, conferring extreme plasticity on phagocytosis and providing attractive targets for therapeutic intervention in cancer, atherosclerosis, and dementia. Expected final online publication date for the Annual Review of Cell and Developmental Biology, Volume 37 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Spencer Freeman
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario M5G0A4, Canada; , .,Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Sergio Grinstein
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario M5G0A4, Canada; , .,Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
58
|
Supercritical CO2 assisted preparation of chitosan-based nano-in-microparticles with potential for efficient pulmonary drug delivery. J CO2 UTIL 2021. [DOI: 10.1016/j.jcou.2021.101486] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
59
|
Li J, Jiang X, Li H, Gelinsky M, Gu Z. Tailoring Materials for Modulation of Macrophage Fate. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2004172. [PMID: 33565154 PMCID: PMC9245340 DOI: 10.1002/adma.202004172] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/31/2020] [Indexed: 05/03/2023]
Abstract
Human immune system acts as a pivotal role in the tissue homeostasis and disease progression. Immunomodulatory biomaterials that can manipulate innate immunity and adaptive immunity hold great promise for a broad range of prophylactic and therapeutic purposes. This review is focused on the design strategies and principles of immunomodulatory biomaterials from the standpoint of materials science to regulate macrophage fate, such as activation, polarization, adhesion, migration, proliferation, and secretion. It offers a comprehensive survey and discussion on the tunability of material designs regarding physical, chemical, biological, and dynamic cues for modulating macrophage immune response. The range of such tailorable cues encompasses surface properties, surface topography, materials mechanics, materials composition, and materials dynamics. The representative immunoengineering applications selected herein demonstrate how macrophage-immunomodulating biomaterials are being exploited for cancer immunotherapy, infection immunotherapy, tissue regeneration, inflammation resolution, and vaccination. A perspective on the future research directions of immunoregulatory biomaterials is also provided.
Collapse
Affiliation(s)
- Jinhua Li
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Xinquan Jiang
- Department of Prosthodontics, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Hongjun Li
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| |
Collapse
|
60
|
Shahab S, Kasra M, Dolatshahi-Pirouz A. Design and construction of a novel measurement device for mechanical characterization of hydrogels: A case study. PLoS One 2021; 16:e0247727. [PMID: 33630967 PMCID: PMC7906418 DOI: 10.1371/journal.pone.0247727] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/12/2021] [Indexed: 11/19/2022] Open
Abstract
Natural biopolymer-based hydrogels especially agarose and collagen gels, considering their biocompatibility with cells and their capacity to mimic biological tissues, have widely been used for in-vitro experiments and tissue engineering applications in recent years; nevertheless their mechanical properties are not always optimal for these purposes. Regarding the importance of the mechanical properties of hydrogels, many mechanical characterization studies have been carried out for such biopolymers. In this work, we have focused on understanding the mechanical role of agarose and collagen concentration on the hydrogel strength and elastic behavior. In this direction, Amirkabir Magnetic Bead Rheometry (AMBR) characterization device equipped with an optimized electromagnet, was designed and constructed for the measurement of hydrogel mechanical properties. The operation of AMBR set-up is based on applying a magnetic field to actuate magnetic beads in contact with the gel surface in order to actuate the gel itself. In simple terms the magnetic beads leads give rise to mechanical shear stress on the gel surface when under magnetic influence and together with the associated bead-gel displacement it is possible to calculate the hydrogel shear modulus. Agarose and Collagen gels with respectively 0.2-0.6 wt % and 0.2-0.5 wt % percent concentrations were prepared for mechanical characterization in terms of their shear modulus. The shear modulus values for the different percent concentrations of the agarose gel were obtained in the range 250-650 Pa, indicating the shear modulus increases by increasing in the agar gel concentration. In addition to this, the values of shear modulus for the collagen gel increase as function of concentration in the range 240-520 Pa in accordance with an approximately linear relationship between collagen concentration and gel strength.
Collapse
Affiliation(s)
- Shayan Shahab
- Tissue Engineering Laboratory, Biomedical Engineering Faculty, Amirkabir University of Technology-Tehran Polytechnic, Tehran, Iran
| | - Mehran Kasra
- Tissue Engineering Laboratory, Biomedical Engineering Faculty, Amirkabir University of Technology-Tehran Polytechnic, Tehran, Iran
| | - Alireza Dolatshahi-Pirouz
- Department of Health Technology, Institute of Biotherapeutic Engineering and Drug Targeting, Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Kgs Lyngby, Denmark
| |
Collapse
|
61
|
Daliri K, Pfannkuche K, Garipcan B. Effects of physicochemical properties of polyacrylamide (PAA) and (polydimethylsiloxane) PDMS on cardiac cell behavior. SOFT MATTER 2021; 17:1156-1172. [PMID: 33427281 DOI: 10.1039/d0sm01986k] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
In vitro cell culture is commonly applied in laboratories around the world. Cultured cells are either of primary origin or established cell lines. Such transformed cell lines are increasingly replaced by pluripotent stem cell derived organotypic cells with more physiological properties. The quality of the culture conditions and matrix environment is of considerable importance in this regard. In fact, mechanical cues of the extracellular matrix have substantial effects on the cellular physiology. This is especially true if contractile cells such as cardiomyocytes are cultured. Therefore, elastic biomaterials have been introduced as scaffolds in 2D and 3D culture models for different cell types, cardiac cells among them. In this review, key aspects of cell-matrix interaction are highlighted with focus on cardiomyocytes and chemical properties as well as strengths and potential pitfalls in using two commonly applied polymers for soft matrix engineering, polyacrylamide (PAA) and polydimethylsiloxane (PDMS) are discussed.
Collapse
Affiliation(s)
- Karim Daliri
- Institute for Neurophysiology, University of Cologne, Medical Faculty, Robert Koch Str. 39, 50931 Cologne, Germany.
| | - Kurt Pfannkuche
- Institute for Neurophysiology, University of Cologne, Medical Faculty, Robert Koch Str. 39, 50931 Cologne, Germany. and Department for Pediatric Cardiology, University Hospital Cologne, Cologne, Germany and Marga-and-Walter-Boll Laboratory for Cardiac Tissue Engineering, University of Cologne, Germany and Center for Molecular Medicine, University of Cologne, Germany
| | - Bora Garipcan
- Institute of Biomedical Engineering, Bogazici University, Cengelkoy, 34684, Istanbul, Turkey.
| |
Collapse
|
62
|
Identification of subtype specific biomarkers of clear cell renal cell carcinoma using random forest and greedy algorithm. Biosystems 2021; 204:104372. [PMID: 33582210 DOI: 10.1016/j.biosystems.2021.104372] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 12/25/2022]
Abstract
Suitable biomarkers can be good indicator for cancer subtype. To find biomarkers that can accurately distinguish clear cell renal cell carcinoma (ccRCC) subtypes, we first determined ccRCC subtypes based on the expression of mRNA, miRNA and lncRNA, named clear cell type 1 (ccluster1) and 2 (ccluster2), using three unsupervised clustering algorithms. Besides being associated with the expression pattern derived from the single type of RNA, the differences between subtypes are relevant to the interactions between RNAs. Then, based on ceRNA network, the optimal combination features are selected using random forest and greedy algorithm. Further, in survival-related sub-ceRNA, competing gene pairs centering on miR-106a, miR-192, miR-193b, miR-454, miR-32, miR-98, miR-143, miR-145, miR-204, miR-424 and miR-1271 can also well identify ccluster1 and ccluster2 with prediction accuracy over 92%. These subtype-specific features potentially enhance the accuracy with which machine learning methods predict specific ccRCC subtypes. Simultaneously, the changes of miR-106 and OIP5-AS1 affect cell proliferation and the prognosis of ccluster1. The changes of miR-145 and FAM13A-AS1 in ccluster2 have an effect on cell invasion, apoptosis, migration and metabolism function. Here miR-192 displays a unique characteristic in both subtypes. Two subtypes also display notable differences in diverse pathways. Tumors belonging to ccluster1 are characterized by Fc gamma R-mediated phagocytosis pathway that affects tissue remodeling and repair, whereas those belonging to ccluster2 are characterized by EGFR tyrosine kinase inhibitor resistance pathway that participates in regulation of cell homeostasis. In conclusion, identifying these gene pairs can shed light on therapeutic mechanisms of ccRCC subtypes.
Collapse
|
63
|
Hyun JY, Lee CH, Lee H, Jang WD, Shin I. Bacterial Lectin-Targeting Glycoconjugates for Selective Elimination of Pathogenic Bacteria. ACS Macro Lett 2020; 9:1429-1432. [PMID: 35653658 DOI: 10.1021/acsmacrolett.0c00454] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Herein we report a strategy to eradicate pathogenic bacteria selectively, which utilizes bacterial lectin-targeting glycoconjugates that contain an epitope or a photosensitizer to promote antibody-dependent cellular cytotoxicity (ADCC) or photodynamic therapy (PDT), respectively. Our results show that death promoted by using the designed synthetic glycoconjugates coupled with ADCC or PDT takes place selectively in pathogenic bacteria expressing lectins on their surfaces.
Collapse
Affiliation(s)
- Ji Young Hyun
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
- Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Chang-Hee Lee
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Hosoowi Lee
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Woo-Dong Jang
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Injae Shin
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
64
|
Han BJ, Murphy JD, Qin S, Ye J, Uccello TP, Garrett-Larsen J, Belt BA, Prieto PA, Egilmez NK, Lord EM, Linehan DC, Mills BN, Gerber SA. Microspheres Encapsulating Immunotherapy Agents Target the Tumor-Draining Lymph Node in Pancreatic Ductal Adenocarcinoma. Immunol Invest 2020; 49:808-823. [PMID: 32498585 PMCID: PMC7904097 DOI: 10.1080/08820139.2020.1765795] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION The tumor-draining lymph node (TDLN) plays a role in tumor immunity. Intratumorally administered microspheres (MS) that encapsulate immunomodulatory agents have emerged as a treatment strategy capable of causing profound changes in the tumor microenvironment (TME) and eliciting potent antitumor effects. We hypothesized that local delivery of MS to the TME may also drain to and therefore target the TDLN to initiate antitumor immune responses. METHODS Fluorescent MS were injected into orthotopically implanted murine pancreatic tumors, and tissues were examined by whole-mount microscopy and imaging flow cytometry. The role of the TDLN was investigated for mice treated with intratumoral interleukin-12 (IL-12)-encapsulated MS in combination with stereotactic body radiotherapy (SBRT) by cytokine profile and TDLN ablation. RESULTS Fluorescent AF-594 MS delivered intratumorally were detected in the tumor, peritumoral lymphatics, and the TDLN 2 h after injection. Phagocytic cells were observed with internalized fluorescent MS. SBRT + IL-12 MS-induced upregulation of Th1 and antitumor factors IL-12, IFN-γ, CXCL10, and granzyme B in the TDLN, and excision of the TDLN partially abrogated treatment efficacy. CONCLUSIONS Our results demonstrate that intratumorally administered MS not only target the TME, but also drain to the TDLN. Furthermore, MS encapsulated with a potent antitumor cytokine, IL-12, induce an antitumor cytokine profile in the TDLN, which is essential for treatment efficacy.
Collapse
Affiliation(s)
- Booyeon J. Han
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Joseph D. Murphy
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Shuyang Qin
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jian Ye
- Department of Surgery, University of Rochester Medical Center, Rochester, NY, USA
| | - Taylor P. Uccello
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jesse Garrett-Larsen
- Department of Surgery, University of Rochester Medical Center, Rochester, NY, USA
| | - Brian A. Belt
- Department of Surgery, University of Rochester Medical Center, Rochester, NY, USA
| | - Peter A. Prieto
- Department of Surgery, University of Rochester Medical Center, Rochester, NY, USA
| | - Nejat K. Egilmez
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Edith M. Lord
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - David C. Linehan
- Department of Surgery, University of Rochester Medical Center, Rochester, NY, USA
| | - Bradley N. Mills
- Department of Surgery, University of Rochester Medical Center, Rochester, NY, USA
| | - Scott A. Gerber
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Surgery, University of Rochester Medical Center, Rochester, NY, USA
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
65
|
Eshaghi B, Alsharif N, An X, Akiyama H, Brown KA, Gummuluru S, Reinhard BM. Stiffness of HIV-1 Mimicking Polymer Nanoparticles Modulates Ganglioside-Mediated Cellular Uptake and Trafficking. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000649. [PMID: 32999830 PMCID: PMC7509657 DOI: 10.1002/advs.202000649] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/19/2020] [Indexed: 05/12/2023]
Abstract
The monosialodihexosylganglioside, GM3, and its binding to CD169 (Siglec-1) have been indicated as key factors in the glycoprotein-independent sequestration of the human immunodeficiency virus-1 (HIV-1) in virus-containing compartments (VCCs) in myeloid cells. Here, lipid-wrapped polymer nanoparticles (NPs) are applied as a virus-mimicking model to characterize the effect of core stiffness on NP uptake and intracellular fate triggered by GM3-CD169 binding in macrophages. GM3-functionalized lipid-wrapped NPs are assembled with poly(lactic-co-glycolic) acid (PLGA) as well as with low and high molecular weight polylactic acid (PLAlMW and PLAhMW) cores. The NPs have an average diameter of 146 ± 17 nm and comparable surface properties defined by the self-assembled lipid layer. Due to differences in the glass transition temperature, the Young's modulus (E) differs substantially under physiological conditions between PLGA (E PLGA = 60 ± 32 MPa), PLAlMW (E PLA lMW = 86 ± 25 MPa), and PLAhMW (E PLA hMW = 1.41 ± 0.67 GPa) NPs. Only the stiff GM3-presenting PLAhMW NPs but not the softer PLGA or PLAlMW NPs avoid a lysosomal pathway and localize in tetraspanin (CD9)-positive compartments that resemble VCCs. These observations suggest that GM3-CD169-induced sequestration of NPs in nonlysosomal compartments is not entirely determined by ligand-receptor interactions but also depends on core stiffness.
Collapse
Affiliation(s)
- Behnaz Eshaghi
- Department of Chemistry and The Photonics CenterBoston UniversityBostonMA02215USA
| | - Nourin Alsharif
- Department of Mechanical Engineering and The Photonics CenterBoston UniversityBostonMA02215USA
| | - Xingda An
- Department of Chemistry and The Photonics CenterBoston UniversityBostonMA02215USA
| | - Hisashi Akiyama
- Department of MicrobiologyBoston University School of MedicineBostonMA02118USA
| | - Keith A. Brown
- Department of Mechanical Engineering and The Photonics CenterBoston UniversityBostonMA02215USA
| | - Suryaram Gummuluru
- Department of MicrobiologyBoston University School of MedicineBostonMA02118USA
| | - Björn M. Reinhard
- Department of Chemistry and The Photonics CenterBoston UniversityBostonMA02215USA
| |
Collapse
|
66
|
Zhang B, Zhu M, Li Z, Lung PS, Chrzanowski W, Kwok CT, Lu J, Li Q. Cellular fate of deformable needle-shaped PLGA-PEG fibers. Acta Biomater 2020; 112:182-189. [PMID: 32470525 DOI: 10.1016/j.actbio.2020.05.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022]
Abstract
Deformability of micro/nanometer sized particles plays an important role in particle-cell interactions and thus becomes a key parameter in carrier design in biomedicine application such as drug delivery and vaccinology. Yet the influence of material's deformability on the cellular fate of the particles as well as physiology response of live cells are to be understood. Here we show the cellular fate of needle shaped (high aspect ratio ~25) PLGA-PEG copolymer fibers depending on their deformability. We found that all the fibers entered murine macrophage cells (RAW 264.7) via phagocytosis. While the fibers of high apparent Young's modulus (average value = 872 kPa) maintained their original shape upon phagocytosis, their counterparts of low apparent Young's modulus (average value = 56 kPa) curled in cells. The observed deformation of fibers of low apparent Young's modulus in cells coincided with abnormal intracellular actin translocation and absence of lysosome/phagosome fusion in macrophages, suggesting the important role of material mechanical properties and mechano-related cellular pathway in affecting cell physiology. STATEMENT OF SIGNIFICANCE: Particles are increasingly important in the field of biomedicine, especially when they are serving as drug carriers. Physical cues, such as mechanical properties, were shown to provide insight into their stability and influence on physiology inside the cell. In the current study, we managed to fabricate 5 types of needle shaped PLGA-PEG fibers with controlled Young's modulus. We found that hard fibers maintained their original shape upon phagocytosis, while soft fibers were curled by actin compressive force inside the cell, causing abnormal actin translocation and impediment of lysosome/phagosome fusion, suggesting the important role of material mechanical properties and mechano-related cellular pathway in affecting cell physiology.
Collapse
|
67
|
Jain N, Moeller J, Vogel V. Mechanobiology of Macrophages: How Physical Factors Coregulate Macrophage Plasticity and Phagocytosis. Annu Rev Biomed Eng 2020; 21:267-297. [PMID: 31167103 DOI: 10.1146/annurev-bioeng-062117-121224] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In addition to their early-recognized functions in host defense and the clearance of apoptotic cell debris, macrophages play vital roles in tissue development, homeostasis, and repair. If misregulated, they steer the progression of many inflammatory diseases. Much progress has been made in understanding the mechanisms underlying macrophage signaling, transcriptomics, and proteomics, under physiological and pathological conditions. Yet, the detailed mechanisms that tune circulating monocytes/macrophages and tissue-resident macrophage polarization, differentiation, specification, and their functional plasticity remain elusive. We review how physical factors affect macrophage phenotype and function, including how they hunt for particles and pathogens, as well as the implications for phagocytosis, autophagy, and polarization from proinflammatory to prohealing phenotype. We further discuss how this knowledge can be harnessed in regenerative medicine and for the design of new drugs and immune-modulatory drug delivery systems, biomaterials, and tissue scaffolds.
Collapse
Affiliation(s)
- Nikhil Jain
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, and Department of Health Sciences and Technology, ETH Zurich, CH-8093 Zurich, Switzerland;
| | - Jens Moeller
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, and Department of Health Sciences and Technology, ETH Zurich, CH-8093 Zurich, Switzerland;
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, and Department of Health Sciences and Technology, ETH Zurich, CH-8093 Zurich, Switzerland;
| |
Collapse
|
68
|
Saelens X. The Role of Matrix Protein 2 Ectodomain in the Development of Universal Influenza Vaccines. J Infect Dis 2020; 219:S68-S74. [PMID: 30715367 PMCID: PMC6452325 DOI: 10.1093/infdis/jiz003] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The influenza A virus matrix protein 2 ectodomain (M2e) is a universal influenza A vaccine candidate. Numerous studies in laboratory mice, but very few in natural influenza A virus hosts, have demonstrated that M2e-based vaccines can provide protection against any influenza A virus challenge. M2e-based immunity is largely accomplished by IgG and early stage clinical studies have demonstrated that the vaccine is safe. Yet M2e is considered a difficult target to develop as a vaccine: it does not offer sterilizing immunity and its mode of action relies on Fcγ receptor-mediated effector mechanisms, most likely in concert with alveolar macrophages. In a human challenge study with an H3N2 virus, treatment with a monoclonal M2e-specific human IgG was associated with a faster recovery compared to placebo treatment. If the universal influenza vaccine field incorporates this antigen into next generation vaccines, M2e could prove its merit when the next influenza pandemic strikes.
Collapse
Affiliation(s)
- Xavier Saelens
- VIB-UGent Center for Medical Biotechnology, Ghent.,Department of Biomedical Molecular Biology, Ghent University, Belgium.,Department of Biochemistry and Microbiology, Ghent University, Belgium
| |
Collapse
|
69
|
Chen Y, Li X, Wang M, Peng L, Yu Z, Peng X, Song J, Qu J. Virus-Inspired Deformable Mesoporous Nanocomposites for High Efficiency Drug Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1906028. [PMID: 31994359 DOI: 10.1002/smll.201906028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/14/2020] [Indexed: 06/10/2023]
Abstract
Mesoporous nanoparticles as a versatile platform for cancer theranostics have been widely used, but their cellular delivery efficiency is still far from satisfactory. Although deformability is emerging as an important parameter influencing cellular uptake enhancement, the facile synthesis of deformable mesoporous nanocomposite with adjustable mechanical property is challenging but meaningful for a deeper understanding of cellular uptake mechanisms and significantly improving cancer therapy. In this work, yolk-shell structured eccentric mesoporous organosilica (YEMO) nanocomposites with adjustable mechanical property are successfully prepared by an organosilane-assisted anisotropic self-assembly approach. The feasibility to precisely control the mechanical property of the YEMO by manipulating the structural parameters, the crosslinking degree of mesoporous framework, and the rotation rate of the reaction is demonstrated. The study of the fabrication mechanism and mechanical properties of YEMO are discussed in detail. The Young's modulus (EY ) of YEMO can be adjusted from 2.4 to 65 MPa. Thereby, the continuous control of the cellular uptake from ≈15% to ≈80% under the same incubation time is achieved. To further prove the higher efficiency drug delivery of YEMO with soft characteristics, the higher toxicity of the "soft" YEMO loaded with the anticancer drug doxorubicin compared to the "stiff" one is demonstrated.
Collapse
Affiliation(s)
- Yu Chen
- Key Laboratory of Optoelectronic Devices and Systems of the Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Xiaobin Li
- Key Laboratory of Optoelectronic Devices and Systems of the Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Meng Wang
- Key Laboratory of Optoelectronic Devices and Systems of the Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Lucheng Peng
- Key Laboratory of Optoelectronic Devices and Systems of the Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Zhongzheng Yu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637459, Singapore
| | - Xiao Peng
- Key Laboratory of Optoelectronic Devices and Systems of the Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Jun Song
- Key Laboratory of Optoelectronic Devices and Systems of the Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of the Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China
| |
Collapse
|
70
|
Huang X, Cavalcante DP, Townley HE. Macrophage-like THP-1 cells show effective uptake of silica nanoparticles carrying inactivated diphtheria toxoid for vaccination. JOURNAL OF NANOPARTICLE RESEARCH : AN INTERDISCIPLINARY FORUM FOR NANOSCALE SCIENCE AND TECHNOLOGY 2020; 22:23. [PMID: 32435151 PMCID: PMC7223038 DOI: 10.1007/s11051-019-4720-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/22/2019] [Indexed: 06/11/2023]
Abstract
Nanoparticles may be used in vaccinology as an antigen delivery and/or an immunostimulant to enhance immunity. Porous silica has been identified as an effective adjuvant for more than a decade, and we have therefore investigated the take up rate by an immortalized macrophage-like cell line of a number of mesoporous silica nanoparticles (MSNPs) with differing diameter and pore size. The MSNPs were synthesized using a sol-gel reaction and post-synthesis removal of the template. The MSNPs showed a clear distribution in take up rate peaking at 217 nm, whereas a comparison with solid spherical nanoparticles showed a similar distribution peaking at 377 nm. The MSNPs were investigated before and after loading with antigen. Diphtheria toxoid was used as a proof-of-concept antigen and showed a peak macrophage internalization of 53.42% for loaded LP3 particles which had a diameter of 217.75 ± 5.44 nm and large 16.5 nm pores. Optimal MSNP sizes appeared to be in the 200-400 nm range, and larger pores showed better antigen loading. The mesoporous silica particles were shown to be generally biocompatible, and cell viability was not altered by the loading of particles with or without antigen. Graphical abstract.
Collapse
Affiliation(s)
- Xinyue Huang
- Nuffield Department of women’s and reproductive health, Oxford University, John Radcliffe Hospital, Oxford, UK
| | | | - Helen E Townley
- Nuffield Department of women’s and reproductive health, Oxford University, John Radcliffe Hospital, Oxford, UK
- Department of Engineering Science, Oxford University, Park’s Road, Oxford, UK
| |
Collapse
|
71
|
Benne N, Leboux RJT, Glandrup M, van Duijn J, Lozano Vigario F, Neustrup MA, Romeijn S, Galli F, Kuiper J, Jiskoot W, Slütter B. Atomic force microscopy measurements of anionic liposomes reveal the effect of liposomal rigidity on antigen-specific regulatory T cell responses. J Control Release 2019; 318:246-255. [PMID: 31812539 DOI: 10.1016/j.jconrel.2019.12.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/03/2019] [Accepted: 12/04/2019] [Indexed: 12/21/2022]
Abstract
Regulatory T cells (Tregs) are vital for maintaining a balanced immune response and their dysfunction is often associated with auto-immune disorders. We have previously shown that antigen-loaded anionic liposomes composed of phosphatidylcholine (PC) and phosphatidylglycerol (PG) and cholesterol can induce strong antigen-specific Treg responses. We hypothesized that altering the rigidity of these liposomes while maintaining their size and surface charge would affect their capability of inducing Treg responses. The rigidity of liposomes is affected in part by the length and saturation of carbon chains of the phospholipids in the bilayer, and in part by the presence of cholesterol. We used atomic force microscopy (AFM) to measure the rigidity of anionic OVA323-containing liposomes composed of different types of PC and PG, with or without cholesterol, in a molar ratio of 4:1(:2) distearoyl (DS)PC:DSPG (Young's modulus (YM) 3611 ± 1271 kPa), DSPC:DSPG:CHOL (1498 ± 531 kPa), DSPC:dipalmitoyl (DP)PG:CHOL (1208 ± 538), DPPC:DPPG:CHOL (1195 ± 348 kPa), DSPC:dioleoyl (DO)PG:CHOL (825 ± 307 kPa), DOPC:DOPG:CHOL (911 ± 447 kPa), and DOPC:DOPG (494 ± 365 kPa). Next, we assessed if rigidity affects the association of liposomes to bone marrow-derived dendritic cells (BMDCs) in vitro. Aside from DOPC:DOPG liposomes, we observed a positive correlation between liposomal rigidity and cellular association. Finally, we show that rigidity positively correlates with Treg responses in vitro in murine DCs and in vivo in mice. Our findings underline the suitability of AFM to measure liposome rigidity and the importance of this parameter when designing liposomes as a vaccine delivery system.
Collapse
Affiliation(s)
- Naomi Benne
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, the Netherlands
| | - Romain J T Leboux
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, the Netherlands
| | - Marco Glandrup
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, the Netherlands
| | - Janine van Duijn
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, the Netherlands
| | - Fernando Lozano Vigario
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, the Netherlands
| | - Malene Aaby Neustrup
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, the Netherlands
| | - Stefan Romeijn
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, the Netherlands
| | | | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, the Netherlands
| | - Wim Jiskoot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, the Netherlands
| | - Bram Slütter
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden, the Netherlands.
| |
Collapse
|
72
|
Mosaiab T, Farr DC, Kiefel MJ, Houston TA. Carbohydrate-based nanocarriers and their application to target macrophages and deliver antimicrobial agents. Adv Drug Deliv Rev 2019; 151-152:94-129. [PMID: 31513827 DOI: 10.1016/j.addr.2019.09.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/02/2019] [Accepted: 09/05/2019] [Indexed: 12/18/2022]
Abstract
Many deadly infections are produced by microorganisms capable of sustained survival in macrophages. This reduces exposure to chemadrotherapy, prevents immune detection, and is akin to criminals hiding in police stations. Therefore, the use of glyco-nanoparticles (GNPs) as carriers of therapeutic agents is a burgeoning field. Such an approach can enhance the penetration of drugs into macrophages with specific carbohydrate targeting molecules on the nanocarrier to interact with macrophage lectins. Carbohydrates are natural biological molecules and the key constituents in a large variety of biological events such as cellular communication, infection, inflammation, enzyme trafficking, cellular migration, cancer metastasis and immune functions. The prominent characteristics of carbohydrates including biodegradability, biocompatibility, hydrophilicity and the highly specific interaction of targeting cell-surface receptors support their potential application to drug delivery systems (DDS). This review presents the 21st century development of carbohydrate-based nanocarriers for drug targeting of therapeutic agents for diseases localized in macrophages. The significance of natural carbohydrate-derived nanoparticles (GNPs) as anti-microbial drug carriers is highlighted in several areas of treatment including tuberculosis, salmonellosis, leishmaniasis, candidiasis, and HIV/AIDS.
Collapse
Affiliation(s)
- Tamim Mosaiab
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Dylan C Farr
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD 4222, Australia
| | - Milton J Kiefel
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD 4222, Australia.
| | - Todd A Houston
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD 4222, Australia.
| |
Collapse
|
73
|
Coupling of β 2 integrins to actin by a mechanosensitive molecular clutch drives complement receptor-mediated phagocytosis. Nat Cell Biol 2019; 21:1357-1369. [PMID: 31659275 PMCID: PMC6858589 DOI: 10.1038/s41556-019-0414-2] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 09/25/2019] [Indexed: 12/21/2022]
Abstract
αMβ2 integrin (complement receptor 3) is a major receptor for phagocytosis in macrophages. In other contexts, integrins’ activities and functions are mechanically linked to actin dynamics through focal adhesions (FAs). We asked whether mechanical coupling of αMβ2 integrin to the actin cytoskeleton mediates phagocytosis. We found that particle internalization was driven by formation of Arp2/3 and formin-dependent actin protrusions that wrapped around the particle. Focal complex-like adhesions formed in the phagocytic cup that contained β2 integrins, FA proteins and tyrosine kinases. Perturbation of talin and Syk demonstrated that a talin-dependent link between integrin and actin and Syk-mediated recruitment of vinculin enable force transmission to target particles and promote phagocytosis. Altering target mechanical properties demonstrated more efficient phagocytosis of stiffer targets. Thus, macrophages use tyrosine kinase signaling to build a mechanosensitive, talin- and vinculin-mediated, FA-like molecular clutch, which couples integrins to cytoskeletal forces to drive particle engulfment.
Collapse
|
74
|
Assaying How Phagocytic Success Depends on the Elasticity of a Large Target Structure. Biophys J 2019; 117:1496-1507. [PMID: 31586520 DOI: 10.1016/j.bpj.2019.08.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 08/14/2019] [Accepted: 08/26/2019] [Indexed: 01/26/2023] Open
Abstract
Biofilm infections can consist of bacterial aggregates that are an order of magnitude larger than neutrophils, phagocytic immune cells that densely surround aggregates but do not enter them. Because a neutrophil is too small to engulf the entire aggregate, it must be able to detach and engulf a few bacteria at a time if it is to use phagocytosis to clear the infection. Current research techniques do not provide a method for determining how the success of phagocytosis, here defined as the complete engulfment of a piece of foreign material, depends on the mechanical properties of a larger object from which the piece must be removed before being engulfed. This article presents a step toward such a method. By varying polymer concentration or cross-linking density, the elastic moduli of centimeter-sized gels are varied over the range that was previously measured for Pseudomonas aeruginosa biofilms grown from clinical bacterial isolates. Human neutrophils are isolated from blood freshly drawn from healthy adult volunteers, exposed to gel containing embedded beads for 1 h, and removed from the gel. The percentage of collected neutrophils that contain beads that had previously been within the gels is used to measure successful phagocytic engulfment. Both increased polymer concentration in agarose gels and increased cross-linking density in alginate gels are associated with a decreased success of phagocytic engulfment. Upon plotting the percentage of neutrophils showing successful engulfment as a function of the elastic modulus of the gel to which they were applied, it is found that data from both alginate and agarose gels collapse onto the same curve. This suggests that gel mechanics may be impacting the success of phagocytosis and demonstrates that this experiment is a step toward realizing methods for measuring how the mechanics of a large target, or a large structure in which smaller targets are embedded, impact the success of phagocytic engulfment.
Collapse
|
75
|
Gangotra A, Biviano M, Dagastine RR, Berry JD, Willmott GR. Use of microaspiration to study the mechanical properties of polymer gel microparticles. SOFT MATTER 2019; 15:7286-7294. [PMID: 31498362 DOI: 10.1039/c9sm00862d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The mechanical properties of polyacrylamide (PA) and polydimethylsiloxane (PDMS) microparticle populations have been measured using microaspiration, a recently developed experimental technique. Microaspiration is an augmented version of micropipette aspiration, in which optical microscopy data are obtained as individual soft particles pass through the tip of a micropipette. During microaspiration, the ion current passing through the pipette tip is also measured, and the synchronised optical and current data streams are used to study and quantify mechanical properties. Ion current signatures for the poroelastic PA particles were qualitatively different from those of the viscoelastic PDMS particles. For PA particles the current gradually reduced during each aspiration event, whereas for PDMS particles the current trace resembled a negative top hat function. For PA particles it was found that the maximum change in current during aspiration (ΔIh) increased with particle size. By considering the initial elastic response, a mean effective shear modulus (G') of 6.6 ± 0.2 kPa was found for aspiration of 115 PA particles of ∼10-20 μm diameter. Using a viscoelastic model to describe flow into the pipette, a mean initial effective elastic modulus (E0') of 3.5 ± 1.7 MPa was found for aspiration of 17 PDMS particles of ∼ 9-11 μm diameter. These moduli are consistent with previously reported literature values, providing initial validation of the microaspiration method.
Collapse
Affiliation(s)
- Ankita Gangotra
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, New Zealand
| | | | | | | | | |
Collapse
|
76
|
Friess F, Roch T, Seifert B, Lendlein A, Wischke C. Phagocytosis of spherical and ellipsoidal micronetwork colloids from crosslinked poly(ε-caprolactone). Int J Pharm 2019; 567:118461. [PMID: 31247276 DOI: 10.1016/j.ijpharm.2019.118461] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/14/2019] [Accepted: 06/23/2019] [Indexed: 12/31/2022]
Abstract
The effect of non-spherical particle shapes on cellular uptake has been reported as a general design parameter to control cellular recognition of particulate drug carriers. Beside shape, also size and cell-particle ratio should mutually effect phagocytosis. Here, the capability to control cellular uptake of poly(ɛ-caprolactone) (PCL) based polymer micronetwork colloids (MNC), a carrier system that can be transferred to various shapes, is explored in vitro at test conditions allowing multiple cell-particle contacts. PCL-based MNC were synthesized as spheres with a diameter of ∼6, ∼10, and 13 µm, loaded with a fluorescent dye by a specific technique of swelling, re-dispersion and drying, and transferred into different ellipsoidal shapes by a phantom stretching method. The boundaries of MNC deformability to prolate ellipsoid target shapes were systematically analyzed and found to be at an aspect ratio AR of ∼4 as obtained by a phantom elongation εph of ∼150%. Uptake studies with a murine macrophages cell line showed shape dependency of phagocytosis for selected conditions when varying particle sizes (∼6 and 10 μm),and shapes (εph: 0, 75 or 150%), cell-particle ratios (1:1, 1:2, 1:10, 1:50), and time points (1-24 h). For larger-sized MNC, there was no significant shape effect on phagocytosis as these particles may associate with more than one cell, thus increasing the possibility of phagocytosis by any of these cells. Accordingly, controlling shape effects on phagocytosis for carriers made from degradable polymers relevant for medical applications requires considering further parameters besides shape, such as kinetic aspects of the exposure and uptake by cells.
Collapse
Affiliation(s)
- Fabian Friess
- Institute of Biomaterial Science and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14153 Teltow, Germany; Institute of Chemistry, University of Potsdam, 14476 Potsdam, Germany
| | - Toralf Roch
- Institute of Biomaterial Science and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14153 Teltow, Germany
| | - Barbara Seifert
- Institute of Biomaterial Science and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14153 Teltow, Germany
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14153 Teltow, Germany; Institute of Chemistry, University of Potsdam, 14476 Potsdam, Germany
| | - Christian Wischke
- Institute of Biomaterial Science and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14153 Teltow, Germany.
| |
Collapse
|
77
|
Meli VS, Veerasubramanian PK, Atcha H, Reitz Z, Downing TL, Liu WF. Biophysical regulation of macrophages in health and disease. J Leukoc Biol 2019; 106:283-299. [PMID: 30861205 PMCID: PMC7001617 DOI: 10.1002/jlb.mr0318-126r] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Macrophages perform critical functions for homeostasis and immune defense in tissues throughout the body. These innate immune cells are capable of recognizing and clearing dead cells and pathogens, and orchestrating inflammatory and healing processes that occur in response to injury. In addition, macrophages are involved in the progression of many inflammatory diseases including cardiovascular disease, fibrosis, and cancer. Although it has long been known that macrophages respond dynamically to biochemical signals in their microenvironment, the role of biophysical cues has only recently emerged. Furthermore, many diseases that involve macrophages are also characterized by changes to the tissue biophysical environment. This review will discuss current knowledge about the effects of biophysical cues including matrix stiffness, material topography, and applied mechanical forces, on macrophage behavior. We will also describe the role of molecules that are known to be important for mechanotransduction, including adhesion molecules, ion channels, as well as nuclear mediators such as transcription factors, scaffolding proteins, and epigenetic regulators. Together, this review will illustrate a developing role of biophysical cues in macrophage biology, and also speculate upon molecular targets that may potentially be exploited therapeutically to treat disease.
Collapse
Affiliation(s)
- Vijaykumar S. Meli
- Department of Biomedical Engineering, University of California Irvine, CA 92697
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, CA 92697
| | - Praveen K. Veerasubramanian
- Department of Biomedical Engineering, University of California Irvine, CA 92697
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, CA 92697
| | - Hamza Atcha
- Department of Biomedical Engineering, University of California Irvine, CA 92697
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, CA 92697
| | - Zachary Reitz
- Department of Biomedical Engineering, University of California Irvine, CA 92697
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, CA 92697
| | - Timothy L. Downing
- Department of Biomedical Engineering, University of California Irvine, CA 92697
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, CA 92697
- Department of Microbiology and Molecular Genetics, University of California Irvine, CA 92697
| | - Wendy F. Liu
- Department of Biomedical Engineering, University of California Irvine, CA 92697
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California Irvine, CA 92697
- Department of Chemical and Biomolecular Engineering, University of California Irvine, CA 92697
| |
Collapse
|
78
|
Lang M, Ferron PJ, Bursztyka J, Montjarret A, Duteil E, Bazire A, Bedoux G. Evaluation of immunomodulatory activities of essential oils by high content analysis. J Biotechnol 2019; 303:65-71. [PMID: 31369773 DOI: 10.1016/j.jbiotec.2019.07.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/12/2019] [Accepted: 07/28/2019] [Indexed: 12/23/2022]
Abstract
Nowadays, research concerning immunomodulatory products are of great interest, particularly in the treatment of inflammatory diseases or the prevention of infectious diseases. These activities are usually evaluated on cell cultures, by tracking different factors requiring dedicated manipulation. Evaluation of the immunomodulatory activities of essential oils and pure compounds using several technics adapted to high content analysis is described in this study. This approach allows a multiparametric evaluation on a single cell batch, in order to obtain an overall response. The developed method is based on the simultaneous evaluation of phagocytosis, production of iNOS and secretion of IL-6, induced by contact of RAW 264.7 cells with LPS. The results highlight the immunomodulatory activities of cinnamon and clove essential oils. They also provide information, particularly concerning the inhibitory activity of mint essential oil, which inhibits the LPS-induced phagocytosis of RAW 264.7 cells by 42%, at 100 μg/ml. This work presents for the first time the adaptation of high content analyses for the monitoring of immunomodulatory activities of essential oils. This protocol could be adjustable to other cell types and supplemented by the evaluation of additional parameters.
Collapse
Affiliation(s)
- Marie Lang
- Université Bretagne-Sud, EA 3884, LBCM, IUEM, F-56100 Lorient, France.
| | - Pierre-Jean Ferron
- HCS-Pharma, Rennes Biopôle, 6 rue Pierre Joseph Colin, 35000 Rennes, France
| | - Julian Bursztyka
- HCS-Pharma, Rennes Biopôle, 6 rue Pierre Joseph Colin, 35000 Rennes, France
| | | | | | - Alexis Bazire
- Université Bretagne-Sud, EA 3884, LBCM, IUEM, F-56100 Lorient, France
| | - Gilles Bedoux
- Université Bretagne-Sud, EA 3884, LBCM, IUEM, F-56100 Lorient, France
| |
Collapse
|
79
|
Andrechak JC, Dooling LJ, Discher DE. The macrophage checkpoint CD47 : SIRPα for recognition of 'self' cells: from clinical trials of blocking antibodies to mechanobiological fundamentals. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180217. [PMID: 31431181 DOI: 10.1098/rstb.2018.0217] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Immunotherapies against some solid tumour types have recently shown unprecedented, durable cures in the clinic, and the most successful thus far involves blocking inhibitory receptor 'checkpoints' on T cells. A similar approach with macrophages is emerging by blocking the ubiquitously expressed 'marker of self' CD47 from binding the inhibitory receptor SIRPα on macrophages. Here, we first summarize available information on the safety and efficacy of CD47 blockade, which raises some safety concerns with the clearance of 'self' cells but also suggests some success against haematological (liquid) and solid cancers. Checkpoint blockade generally benefits from parallel activation of the immune cell, which can occur for macrophages in multiple ways, such as by combination with a second, tumour-opsonizing antibody and perhaps also via rigidity sensing. Cytoskeletal forces in phagocytosis and inhibitory 'self'-signalling are thus reviewed together with macrophage mechanosensing, which extends to regulating levels of SIRPα and the nuclear protein lamin A, which affects phenotype and cell trafficking. Considerations of such physical factors in cancer and the immune system can inform the design of new immunotherapies and help to refine existing therapies to improve safety and efficacy. This article is part of a discussion meeting issue 'Forces in cancer: interdisciplinary approaches in tumour mechanobiology'.
Collapse
Affiliation(s)
- Jason C Andrechak
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA, USA.,Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Lawrence J Dooling
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA, USA
| | - Dennis E Discher
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
80
|
Montel L, Pinon L, Fattaccioli J. A Multiparametric and High-Throughput Assay to Quantify the Influence of Target Size on Phagocytosis. Biophys J 2019; 117:408-419. [PMID: 31301802 DOI: 10.1016/j.bpj.2019.06.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/10/2019] [Accepted: 06/17/2019] [Indexed: 11/26/2022] Open
Abstract
Phagocytosis by macrophages represents a fundamental process essential for both immunity and tissue homeostasis. It consists in the uptake of pathogenic or cellular targets larger than 0.5 μm. For the biggest particles, the phagocytic process involves a massive reorganization of membrane and actin cytoskeleton as well as an important intracellular deformation all in a matter of minutes. The study of the role of the size of objects in their phagocytosis has led to contradictory results in the last decades. We designed a method using confocal microscopy, automated image analysis, and databases for fast quantitative analysis of phagocytosis assays. It yields comprehensive data on the cells and targets geometric and fluorescence intensity parameters, automatically discriminates internalized from external targets, and stores the relationship between a cell and the targets it has engulfed. We used two types of targets (solid polystyrene beads and liquid lipid droplets) to investigate the influence of size on the phagocytic uptake of macrophages. The method made it possible not only to perform phagocytic assays with functionalized droplets and beads of different sizes but to use polydisperse particles to further our understanding of the role of size in phagocytosis. The use of monodisperse and polydisperse objects shows that whereas smaller monodisperse objects are internalized in greater numbers, objects of different sizes presented simultaneously are internalized without preferred size. The total surface engulfed by the cell is thus the main factor limiting the uptake of particles, regardless of their nature or size. A meta-analysis of the literature reveals that this dependence in surface is consistently conserved throughout cell types, targets' nature, or activated receptors.
Collapse
Affiliation(s)
- Lorraine Montel
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, Paris, France; Institut Pierre-Gilles de Gennes pour la Microfluidique, Paris, France
| | - Léa Pinon
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, Paris, France; Institut Curie, PSL University, INSERM U932, Paris, France; Institut Pierre-Gilles de Gennes pour la Microfluidique, Paris, France
| | - Jacques Fattaccioli
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, Paris, France; Institut Pierre-Gilles de Gennes pour la Microfluidique, Paris, France.
| |
Collapse
|
81
|
Abstract
Bioavailability is an ancient but effective terminology by which the entire therapeutic efficacy of a drug directly or indirectly relays. Despite considering general plasma bioavailability, specific organ/tissue bioavailability will pave the path to broad spectrum dose calculation. Clear knowledge and calculative vision on bioavailability can improve the research and organ-targeting phenomenon. This article comprises a detailed introduction on bioavailability along with regulatory aspects, kinetic data and novel bioformulative approaches to achieve improved organ specific bioavailability, which may not be readily related to blood plasma bioavailability.
Collapse
|
82
|
Jung H, Jang HE, Kang YY, Song J, Mok H. PLGA Microspheres Coated with Cancer Cell-Derived Vesicles for Improved Internalization into Antigen-Presenting Cells and Immune Stimulation. Bioconjug Chem 2019; 30:1690-1701. [DOI: 10.1021/acs.bioconjchem.9b00240] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Heesun Jung
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyo-Eun Jang
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Yoon Young Kang
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Jihyeon Song
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyejung Mok
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
83
|
Sridharan R, Cavanagh B, Cameron AR, Kelly DJ, O'Brien FJ. Material stiffness influences the polarization state, function and migration mode of macrophages. Acta Biomater 2019; 89:47-59. [PMID: 30826478 DOI: 10.1016/j.actbio.2019.02.048] [Citation(s) in RCA: 265] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 02/20/2019] [Accepted: 02/27/2019] [Indexed: 01/25/2023]
Abstract
Biomaterial implantation is followed by an inflammatory cascade dominated by macrophages, which determine implant acceptance or rejection through pro- and anti-inflammatory polarization states (Anderson et al., 2008; Brown and Badylak, 2013). It is known that chemical signals such as bacterial endotoxins and cytokines (IL4) can direct macrophage polarization (Mantovani et al., 2004); however, recent evidence implicates biophysical cues in this process (McWhorter et al., 2015; Patel et al., 2012). Here we report that THP-1 derived macrophages cultured on collagen-coated polyacrylamide gels of varying stiffness adapt their polarization state, functional roles and migration mode according to the stiffness of the underlying substrate. Through gene expression and protein secretion analysis, we show that stiff polyacrylamide gels (323 kPa) prime macrophages towards a pro-inflammatory phenotype with impaired phagocytosis in macrophages, while soft (11 kPa) and medium (88 kPa) stiffness gels prime cells towards an anti-inflammatory, highly phagocytic phenotype. Furthermore, we show that stiffness dictates the migration mode of macrophages; on soft and medium stiffness gels, cells display Rho-A kinase (ROCK)-dependent, podosome-independent fast amoeboid migration and on stiff gels they adopt a ROCK-independent, podosome-dependent slow mesenchymal migration mode. We also provide a mechanistic insight into this process by showing that the anti-inflammatory property of macrophages on soft and medium gels is ROCK-dependent and independent of the ligand presented to them. Together, our results demonstrate that macrophages adapt their polarization, function and migration mode in response to the stiffness of the underlying substrate and suggest that biomaterial stiffness is capable of directing macrophage behaviour independent of the biochemical cues being presented to them. The results from this study establish an important role for substrate stiffness in directing macrophage behaviour, and will lead to the design of immuno-informed biomaterials that are capable of modulating the macrophage response after implantation. STATEMENT OF SIGNIFICANCE: Biomaterial implantation is followed by an inflammatory cascade dominated by macrophages, which determine implant acceptance or rejection through pro- and anti-inflammatory polarization states. It is known that chemical signals can direct macrophage polarization; however, recent evidence implicates biophysical cues in this process. Here we report that macrophages cultured on gels of varying stiffness adapt their polarization state, functional roles and migration mode according to the stiffness of the underlying substrate. The results from this study establish an important role for substrate stiffness in directing macrophage behaviour, and will lead to the design of immuno-informed biomaterials that are capable of modulating the macrophage response after implantation.
Collapse
Affiliation(s)
- Rukmani Sridharan
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials Bio-Engineering Research (AMBER) Centre, Trinity College Dublin, Dublin 2, Ireland
| | - Brenton Cavanagh
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Cellular and Molecular Imaging Core, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Andrew R Cameron
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials Bio-Engineering Research (AMBER) Centre, Trinity College Dublin, Dublin 2, Ireland
| | - Daniel J Kelly
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials Bio-Engineering Research (AMBER) Centre, Trinity College Dublin, Dublin 2, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials Bio-Engineering Research (AMBER) Centre, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
84
|
Barger SR, Reilly NS, Shutova MS, Li Q, Maiuri P, Heddleston JM, Mooseker MS, Flavell RA, Svitkina T, Oakes PW, Krendel M, Gauthier NC. Membrane-cytoskeletal crosstalk mediated by myosin-I regulates adhesion turnover during phagocytosis. Nat Commun 2019; 10:1249. [PMID: 30890704 PMCID: PMC6425032 DOI: 10.1038/s41467-019-09104-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 02/21/2019] [Indexed: 11/09/2022] Open
Abstract
Phagocytosis of invading pathogens or cellular debris requires a dramatic change in cell shape driven by actin polymerization. For antibody-covered targets, phagocytosis is thought to proceed through the sequential engagement of Fc-receptors on the phagocyte with antibodies on the target surface, leading to the extension and closure of the phagocytic cup around the target. We find that two actin-dependent molecular motors, class 1 myosins myosin 1e and myosin 1f, are specifically localized to Fc-receptor adhesions and required for efficient phagocytosis of antibody-opsonized targets. Using primary macrophages lacking both myosin 1e and myosin 1f, we find that without the actin-membrane linkage mediated by these myosins, the organization of individual adhesions is compromised, leading to excessive actin polymerization, slower adhesion turnover, and deficient phagocytic internalization. This work identifies a role for class 1 myosins in coordinated adhesion turnover during phagocytosis and supports a mechanism involving membrane-cytoskeletal crosstalk for phagocytic cup closure.
Collapse
Affiliation(s)
- Sarah R Barger
- Cell and Developmental Biology Department, State University of New York Upstate Medical University, Syracuse, 13210, NY, USA
| | - Nicholas S Reilly
- Department of Physics, University of Rochester, Rochester, 14627, NY, USA
| | - Maria S Shutova
- Department of Biology, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Qingsen Li
- IFOM, FIRC Institute of Molecular Oncology, Milan, 20139, Italy
| | - Paolo Maiuri
- IFOM, FIRC Institute of Molecular Oncology, Milan, 20139, Italy
| | - John M Heddleston
- Advanced Imaging Center, Howard Hughes Medical Institute Janelia Research Campus, Ashburn, 20147, VA, USA
| | - Mark S Mooseker
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, 06520, CT, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, 06519, CT, USA
- Howard Hughes Medical Institute, Yale University, New Haven, 06519, CT, USA
| | - Tatyana Svitkina
- Department of Biology, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Patrick W Oakes
- Department of Physics, University of Rochester, Rochester, 14627, NY, USA
- Department of Biology, University of Rochester, Rochester, 14627, NY, USA
| | - Mira Krendel
- Cell and Developmental Biology Department, State University of New York Upstate Medical University, Syracuse, 13210, NY, USA.
| | - Nils C Gauthier
- IFOM, FIRC Institute of Molecular Oncology, Milan, 20139, Italy.
| |
Collapse
|
85
|
Tay MZ, Wiehe K, Pollara J. Antibody-Dependent Cellular Phagocytosis in Antiviral Immune Responses. Front Immunol 2019; 10:332. [PMID: 30873178 PMCID: PMC6404786 DOI: 10.3389/fimmu.2019.00332] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/08/2019] [Indexed: 12/20/2022] Open
Abstract
Antiviral activities of antibodies may either be dependent only on interactions between the antibody and cognate antigen, as in binding and neutralization of an infectious virion, or instead may require interactions between antibody-antigen immune complexes and immunoproteins or Fc receptor expressing immune effector cells. These Fc receptor-dependent antibody functions provide a direct link between the innate and adaptive immune systems by combining the potent antiviral activity of innate effector cells with the diversity and specificity of the adaptive humoral response. The Fc receptor-dependent function of antibody-dependent cellular phagocytosis (ADCP) provides mechanisms for clearance of virus and virus-infected cells, as well as for stimulation of downstream adaptive immune responses by facilitating antigen presentation, or by stimulating the secretion of inflammatory mediators. In this review, we discuss the properties of Fc receptors, antibodies, and effector cells that influence ADCP. We also provide and interpret evidence from studies that support a potential role for ADCP in either inhibiting or enhancing viral infection. Finally, we describe current approaches used to measure antiviral ADCP and discuss considerations for the translation of studies performed in animal models. We propose that additional investigation into the role of ADCP in protective viral responses, the specific virus epitopes targeted by ADCP antibodies, and the types of phagocytes and Fc receptors involved in ADCP at sites of virus infection will provide insight into strategies to successfully leverage this important immune response for improved antiviral immunity through rational vaccine design.
Collapse
Affiliation(s)
- Matthew Zirui Tay
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
| | - Kevin Wiehe
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Justin Pollara
- Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
86
|
Patel KR, Roberts JT, Barb AW. Multiple Variables at the Leukocyte Cell Surface Impact Fc γ Receptor-Dependent Mechanisms. Front Immunol 2019; 10:223. [PMID: 30837990 PMCID: PMC6382684 DOI: 10.3389/fimmu.2019.00223] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/25/2019] [Indexed: 12/18/2022] Open
Abstract
Fc γ receptors (FcγR) expressed on the surface of human leukocytes bind clusters of immunoglobulin G (IgG) to induce a variety of responses. Many therapeutic antibodies and vaccine-elicited antibodies prevent or treat infectious diseases, cancers and autoimmune disorders by binding FcγRs, thus there is a need to fully define the variables that impact antibody-induced mechanisms to properly evaluate candidate therapies and design new intervention strategies. A multitude of factors influence the IgG-FcγR interaction; one well-described factor is the differential affinity of the six distinct FcγRs for the four human IgG subclasses. However, there are several other recently described factors that may prove more relevant for disease treatment. This review covers recent reports of several aspects found at the leukocyte membrane or outside the cell that contribute to the cell-based response to antibody-coated targets. One major focus is recent reports covering post-translational modification of the FcγRs, including asparagine-linked glycosylation. This review also covers the organization of FcγRs at the cell surface, and properties of the immune complex. Recent technical advances provide high-resolution measurements of these often-overlooked variables in leukocyte function and immune system activation.
Collapse
Affiliation(s)
- Kashyap R Patel
- Roy J. Carver Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University, Ames, IA, United States
| | - Jacob T Roberts
- Roy J. Carver Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University, Ames, IA, United States
| | - Adam W Barb
- Roy J. Carver Department of Biochemistry, Biophysics, and Molecular Biology, Iowa State University, Ames, IA, United States
| |
Collapse
|
87
|
Visser JG, Van Staden ADP, Smith C. Harnessing Macrophages for Controlled-Release Drug Delivery: Lessons From Microbes. Front Pharmacol 2019; 10:22. [PMID: 30740053 PMCID: PMC6355695 DOI: 10.3389/fphar.2019.00022] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/09/2019] [Indexed: 01/15/2023] Open
Abstract
With the effectiveness of therapeutic agents ever decreasing and the increased incidence of multi-drug resistant pathogens, there is a clear need for administration of more potent, potentially more toxic, drugs. Alternatively, biopharmaceuticals may hold potential but require specialized protection from premature in vivo degradation. Thus, a paralleled need for specialized drug delivery systems has arisen. Although cell-mediated drug delivery is not a completely novel concept, the few applications described to date are not yet ready for in vivo application, for various reasons such as drug-induced carrier cell death, limited control over the site and timing of drug release and/or drug degradation by the host immune system. Here, we present our hypothesis for a new drug delivery system, which aims to negate these limitations. We propose transport of nanoparticle-encapsulated drugs inside autologous macrophages polarized to M1 phenotype for high mobility and treated to induce transient phagosome maturation arrest. In addition, we propose a significant shift of existing paradigms in the study of host-microbe interactions, in order to study microbial host immune evasion and dissemination patterns for their therapeutic utilization in the context of drug delivery. We describe a system in which microbial strategies may be adopted to facilitate absolute control over drug delivery, and without sacrificing the host carrier cells. We provide a comprehensive summary of the lessons we can learn from microbes in the context of drug delivery and discuss their feasibility for in vivo therapeutic application. We then describe our proposed "synthetic microbe drug delivery system" in detail. In our opinion, this multidisciplinary approach may hold the solution to effective, controlled drug delivery.
Collapse
Affiliation(s)
- Johan Georg Visser
- Department of Physiological Sciences, Stellenbosch University, Matieland, South Africa
| | | | - Carine Smith
- Department of Physiological Sciences, Stellenbosch University, Matieland, South Africa
| |
Collapse
|
88
|
Shen Z, Ye H, Yi X, Li Y. Membrane Wrapping Efficiency of Elastic Nanoparticles during Endocytosis: Size and Shape Matter. ACS NANO 2019; 13:215-228. [PMID: 30557506 DOI: 10.1021/acsnano.8b05340] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Using coarse-grained molecular dynamics simulations, we systematically investigate the receptor-mediated endocytosis of elastic nanoparticles (NPs) with different sizes, ranging from 25 to 100 nm, and shapes, including sphere-like, oblate-like, and prolate-like. Simulation results provide clear evidence that the membrane wrapping efficiency of NPs during endocytosis is a result of competition between receptor diffusion kinetics and thermodynamic driving force. The receptor diffusion kinetics refer to the kinetics of receptor recruitment that are affected by the contact edge length between the NP and membrane. The thermodynamic driving force represents the amount of required free energy to drive NPs into a cell. Under the volume constraint of elastic NPs, the soft spherical NPs are found to have similar contact edge lengths to rigid ones and to less efficiently be fully wrapped due to their elastic deformation. Moreover, the difference in wrapping efficiency between soft and rigid spherical NPs increases with their sizes, due to the increment of their elastic energy change. Furthermore, because of its prominent large contact edge length, the oblate ellipsoid is found to be the least sensitive geometry to the variation in NP's elasticity among the spherical, prolate, and oblate shapes during the membrane wrapping. In addition, simulation results indicate that conflicting experimental observations on the efficiency of cellular uptake of elastic NPs could be caused by their different mechanical properties. Our simulations provide a detailed mechanistic understanding about the influence of NPs' size, shape, and elasticity on their membrane wrapping efficiency, which serves as a rational guidance for the design of NP-based drug carriers.
Collapse
Affiliation(s)
- Zhiqiang Shen
- Department of Mechanical Engineering , University of Connecticut , Storrs , Connecticut 06269 , United States
| | - Huilin Ye
- Department of Mechanical Engineering , University of Connecticut , Storrs , Connecticut 06269 , United States
| | - Xin Yi
- Department of Mechanics and Engineering Science, College of Engineering, and Beijing Innovation Center for Engineering Science and Advanced Technology , Peking University , Beijing 100871 , China
| | - Ying Li
- Department of Mechanical Engineering and Institute of Materials Science , University of Connecticut , Storrs , Connecticut 06269 , United States
| |
Collapse
|
89
|
Dispersible hydrogel force sensors reveal patterns of solid mechanical stress in multicellular spheroid cultures. Nat Commun 2019; 10:144. [PMID: 30635553 PMCID: PMC6329783 DOI: 10.1038/s41467-018-07967-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 12/07/2018] [Indexed: 12/24/2022] Open
Abstract
Understanding how forces orchestrate tissue formation requires technologies to map internal tissue stress at cellular length scales. Here, we develop ultrasoft mechanosensors that visibly deform under less than 10 Pascals of cell-generated stress. By incorporating these mechanosensors into multicellular spheroids, we capture the patterns of internal stress that arise during spheroid formation. We experimentally demonstrate the spontaneous generation of a tensional 'skin', only a few cell layers thick, at the spheroid surface, which correlates with activation of mechanobiological signalling pathways, and balances a compressive stress profile within the tissue. These stresses develop through cell-driven mechanical compaction at the tissue periphery, and suggest that the tissue formation process plays a critically important role in specifying mechanobiological function. The broad applicability of this technique should ultimately provide a quantitative basis to design tissues that leverage the mechanical activity of constituent cells to evolve towards a desired form and function.
Collapse
|
90
|
|
91
|
Ashton JR, Gottlin EB, Patz EF, West JL, Badea CT. A comparative analysis of EGFR-targeting antibodies for gold nanoparticle CT imaging of lung cancer. PLoS One 2018; 13:e0206950. [PMID: 30408128 PMCID: PMC6224087 DOI: 10.1371/journal.pone.0206950] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 10/21/2018] [Indexed: 11/29/2022] Open
Abstract
Computed tomography (CT) is the standard imaging test used for the screening and assessment of suspected lung cancer, but distinguishing malignant from benign nodules by CT is an ongoing challenge. Consequently, a large number of avoidable invasive procedures are performed on patients with benign nodules in order to exclude malignancy. Improving cancer discrimination by non-invasive imaging could reduce the need for invasive diagnostics. In this work we focus on developing a gold nanoparticle contrast agent that targets the epidermal growth factor receptor (EGFR), which is expressed on the cell surface of most lung adenocarcinomas. Three different contrast agents were compared for their tumor targeting effectiveness: non-targeted nanoparticles, nanoparticles conjugated with full-sized anti-EGFR antibodies (cetuximab), and nanoparticles conjugated with a single-domain llama-derived anti-EGFR antibody, which is smaller than the cetuximab, but has a lower binding affinity. Nanoparticle targeting effectiveness was evaluated in vitro by EGFR-binding assays and in cell culture with A431 cells, which highly express EGFR. In vivo CT imaging performance was evaluated in both C57BL/6 mice and in nude mice with A431 subcutaneous tumors. The cetuximab nanoparticles had a significantly shorter blood residence time than either the non-targeted or the single-domain antibody nanoparticles. All of the nanoparticle contrast agents demonstrated tumor accumulation; however, the cetuximab-targeted group had significantly higher tumor gold accumulation than the other two groups, which were statistically indistinguishable from one another. In this study we found that the relative binding affinity of the targeting ligands had more of an effect on tumor accumulation than the circulation half life of the nanoparticles. This study provides useful insight into targeted nanoparticle design and demonstrates that nanoparticle contrast agents can be used to detect tumor receptor overexpression. Combining receptor status data with traditional imaging characteristics has the potential for better differentiation of malignant lung tumors from benign lesions.
Collapse
Affiliation(s)
- Jeffrey R. Ashton
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
- Department of Radiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Elizabeth B. Gottlin
- Department of Radiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Edward F. Patz
- Department of Radiology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Jennifer L. West
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
| | - Cristian T. Badea
- Department of Radiology, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
92
|
Affiliation(s)
- Brandon M Johnson
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Robert D Junkins
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Jenny P-Y Ting
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Center for Translation Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Institute for Inflammatory Diseases, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
93
|
Shah MK, Leary EA, Darling EM. Integration of hyper-compliant microparticles into a 3D melanoma tumor model. J Biomech 2018; 82:46-53. [PMID: 30392774 DOI: 10.1016/j.jbiomech.2018.10.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/11/2018] [Accepted: 10/17/2018] [Indexed: 12/19/2022]
Abstract
Multicellular spheroids provide a physiologically relevant platform to study the microenvironment of tumors and therapeutic applications, such as microparticle-based drug delivery. The goal of this study was to investigate the incorporation/penetration of compliant polyacrylamide microparticles (MPs), into either cancer or normal human cell spheroids. Incorporation of collagen-1-coated MPs (stiffness: 0.1 and 9 kPa; diameter: 15-30 µm) into spheroids (diameter ∼100 µm) was tracked for up to 22 h. Results indicated that cells within melanoma spheroids were more influenced by MP mechanical properties than cells within normal cell spheroids. Melanoma spheroids had a greater propensity to incorporate and displace the more compliant MPs over time. Mature spheroids composed of either cell type were able to recognize and integrate MPs. While many tumor models exist to study drug delivery and efficacy, the study of uptake and incorporation of cell-sized MPs into established spheroids/tissues or tumors has been limited. The ability of hyper-compliant MPs to successfully penetrate 3D tumor models with natural extracellular matrix deposition provides a novel platform for potential delivery of drugs and other therapeutics into the core of tumors and micrometastases.
Collapse
Affiliation(s)
- Manisha K Shah
- Center for Biomedical Engineering, Brown University, RI, USA
| | | | - Eric M Darling
- Center for Biomedical Engineering, Brown University, RI, USA; Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, RI, USA; Department of Orthopaedics, Brown University, RI, USA; School of Engineering, Brown University, RI, USA.
| |
Collapse
|
94
|
Kruger TM, Givens BE, Lansakara TI, Bell KJ, Mohapatra H, Salem AK, Tivanski AV, Stevens LL. Mechanosensitive Endocytosis of High-Stiffness, Submicron Microgels in Macrophage and Hepatocarcinoma Cell Lines. ACS APPLIED BIO MATERIALS 2018; 1:1254-1265. [PMID: 34996229 DOI: 10.1021/acsabm.8b00111] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The mechanical properties of submicron particles offer a unique design space for advanced drug-delivery particle engineering. However, the recognition of this potential is limited by a poor consensus about both the specificity and sensitivity of mechanosensitive endocytosis over a broad particle stiffness range. In this report, our model series of polystyrene-co-poly(N-isopropylacrylamide) (pS-co-NIPAM) microgels have been prepared with a nominally constant monomer composition (50 mol % styrene and 50 mol % NIPAM) with varied bis-acrylamide cross-linking densities to introduce a tuned spectrum of particle mechanics without significant variation in particle size and surface charge. While previous mechanosensitive studies use particles with moduli ranging from 15 kPa to 20 MPa, the pS-co-NIPAM particles have Young's moduli (E) ranging from 300 to 700 MPa, which is drastically stiffer than these previous studies as well as pure pNIPAM. Despite this elevated stiffness, particle uptake in RAW264.7 murine macrophages displays a clear stiffness dependence, with a significant increase in particle uptake for our softest microgels after a 4 h incubation. Preferential uptake of the softest microgel, pS-co-NIPAM-1 (E = 310 kPa), was similarly observed with nonphagocytic HepG2 hepatoma cells; however, the uptake kinetics were distinct relative to that observed for RAW264.7 cells. Pharmacological inhibitors, used to probe for specific routes of particle internalization, identify actin- and microtubule-dependent pathways in RAW264.7 cells as sensitive particle mechanics. For our pS-co-NIPAM particles at nominally 300-400 nm in size, this microtubule-dependent pathway was interpreted as a phagocytic route. For our high-stiffness microgel series, this study provides evidence of cell-specific, mechanosensitive endocytosis in a distinctly new stiffness regime that will further broaden the functional landscape of mechanics as a design space for particle engineering.
Collapse
Affiliation(s)
- Terra M. Kruger
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Brittany E. Givens
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, Iowa 52242, United States
- Department of Chemical and Biochemical Engineering, College of Engineering, The University of Iowa, Iowa City, Iowa 52242, United States
| | | | - Kendra J. Bell
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Himansu Mohapatra
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Aliasger K. Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Alexei V. Tivanski
- Department of Chemistry, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Lewis L. Stevens
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, Iowa 52242, United States
| |
Collapse
|
95
|
Jenkins E, Santos AM, O'Brien-Ball C, Felce JH, Wilcock MJ, Hatherley D, Dustin ML, Davis SJ, Eggeling C, Sezgin E. Reconstitution of immune cell interactions in free-standing membranes. J Cell Sci 2018; 132:jcs219709. [PMID: 30209137 PMCID: PMC6398472 DOI: 10.1242/jcs.219709] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 09/04/2018] [Indexed: 12/28/2022] Open
Abstract
The spatiotemporal regulation of signalling proteins at the contacts formed between immune cells and their targets determines how and when immune responses begin and end. Therapeutic control of immune responses therefore relies on thorough elucidation of the molecular processes occurring at these interfaces. However, the detailed investigation of each component's contribution to the formation and regulation of the contact is hampered by the complexities of cell composition and architecture. Moreover, the transient nature of these interactions creates additional challenges, especially in the use of advanced imaging technology. One approach that circumvents these problems is to establish in vitro systems that faithfully mimic immune cell interactions, but allow complexity to be 'dialled-in' as needed. Here, we present an in vitro system that makes use of synthetic vesicles that mimic important aspects of immune cell surfaces. Using this system, we began to explore the spatial distribution of signalling molecules (receptors, kinases and phosphatases) and how this changes during the initiation of signalling. The GUV/cell system presented here is expected to be widely applicable.
Collapse
Affiliation(s)
- Edward Jenkins
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Ana Mafalda Santos
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Caitlin O'Brien-Ball
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - James H Felce
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| | - Martin J Wilcock
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Deborah Hatherley
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| | - Simon J Davis
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Christian Eggeling
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
- Institute of Applied Optics Friedrich-Schiller-University Jena, Max-Wien Platz 4, 07743 Jena, Germany
- Leibniz Institute of Photonic Technology e.V., Albert-Einstein-Straße 9, 07745 Jena, Germany
| | - Erdinc Sezgin
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| |
Collapse
|
96
|
Agrawal G, Agrawal R. Functional Microgels: Recent Advances in Their Biomedical Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1801724. [PMID: 30035853 DOI: 10.1002/smll.201801724] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 06/11/2018] [Indexed: 06/08/2023]
Abstract
Here, a spotlight is shown on aqueous microgel particles which exhibit a great potential for various biomedical applications such as drug delivery, cell imaging, and tissue engineering. Herein, different synthetic methods to develop microgels with desirable functionality and properties along with degradable strategies to ensure their renal clearance are briefly presented. A special focus is given on the ability of microgels to respond to various stimuli such as temperature, pH, redox potential, magnetic field, light, etc., which helps not only to adjust their physical and chemical properties, and degradability on demand, but also the release of encapsulated bioactive molecules and thus making them suitable for drug delivery. Furthermore, recent developments in using the functional microgels for cell imaging and tissue regeneration are reviewed. The results reviewed here encourage the development of a new class of microgels which are able to intelligently perform in a complex biological environment. Finally, various challenges and possibilities are discussed in order to achieve their successful clinical use in future.
Collapse
Affiliation(s)
- Garima Agrawal
- Department of Polymer and Process Engineering, Indian Institute of Technology Roorkee, Saharanpur Campus, Paper Mill Road, Saharanpur, 247001, Uttar Pradesh, India
| | - Rahul Agrawal
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-1500, USA
| |
Collapse
|
97
|
Shen Z, Ye H, Li Y. Understanding receptor-mediated endocytosis of elastic nanoparticles through coarse grained molecular dynamic simulation. Phys Chem Chem Phys 2018; 20:16372-16385. [PMID: 29445792 DOI: 10.1039/c7cp08644j] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
For nanoparticle (NP)-based drug delivery platforms, the elasticity of the NPs has a significant influence on their blood circulation time and cellular uptake efficiency. However, due to the complexity of the endocytosis process and the inconsistency in the definition of elasticity for NPs in experiments, the understanding about the receptor-mediated endocytosis process of elastic NPs is still limited. In this work, we developed a coarse-grained molecular dynamics (CGMD) model for elastic NPs. The energy change of the elastic NPs can be precisely controlled by the bond, area, volume and bending potentials of this CGMD model. To represent liposomes with different elasticities, we systematically varied the bending rigidity of elastic NPs in CGMD simulations. Additionally, we changed the radius of the elastic NPs to explore the potential size effect. Through virtual nano-indentation tests, we found that the effective stiffness of elastic NPs was determined by their bending rigidity and size. Afterwards, we investigated the receptor-mediated endocytosis process of elastic NPs with different sizes and bending rigidities. We found that the membrane wrapping of soft NPs was faster than that of the stiff ones at the early stage, due to the NP deformation induced large contact area between the NPs and the membrane. However, because of the large energy penalties induced by the NP deformation, the membrane wrapping speed of soft NPs slows down during the late stage. Eventually, the soft NPs are wrapped less efficiently than the stiff ones during the membrane wrapping process. Through systematic CGMD simulations, we found a scaling law between the cellular uptake efficiency and the phenomenal bending rigidity of elastic NPs, which agrees reasonably well with experimental observations. Furthermore, we observed that the membrane wrapping efficiencies of soft and stiff NPs with large sizes were close to each other, due to the stronger ligand-receptor binding force and smaller difference in the stiffness of elastic NPs. Our computational model provides an effective tool to investigate the receptor-mediated endocytosis of elastic NPs with well controlled mechanical properties. This study can also be applied to guide the design of NP-based drug carriers with high efficacy, by utilizing their elastic properties.
Collapse
Affiliation(s)
- Zhiqiang Shen
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT 06269, USA.
| | | | | |
Collapse
|
98
|
Morrissey MA, Williamson AP, Steinbach AM, Roberts EW, Kern N, Headley MB, Vale RD. Chimeric antigen receptors that trigger phagocytosis. eLife 2018; 7:36688. [PMID: 29862966 PMCID: PMC6008046 DOI: 10.7554/elife.36688] [Citation(s) in RCA: 249] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/26/2018] [Indexed: 12/14/2022] Open
Abstract
Chimeric antigen receptors (CARs) are synthetic receptors that reprogram T cells to kill cancer. The success of CAR-T cell therapies highlights the promise of programmed immunity and suggests that applying CAR strategies to other immune cell lineages may be beneficial. Here, we engineered a family of Chimeric Antigen Receptors for Phagocytosis (CAR-Ps) that direct macrophages to engulf specific targets, including cancer cells. CAR-Ps consist of an extracellular antibody fragment, which can be modified to direct CAR-P activity towards specific antigens. By screening a panel of engulfment receptor intracellular domains, we found that the cytosolic domains from Megf10 and FcRɣ robustly triggered engulfment independently of their native extracellular domain. We show that CAR-Ps drive specific engulfment of antigen-coated synthetic particles and whole human cancer cells. Addition of a tandem PI3K recruitment domain increased cancer cell engulfment. Finally, we show that CAR-P expressing murine macrophages reduce cancer cell number in co-culture by over 40%. Our immune system constantly patrols our body, looking to eliminate cancerous cells and harmful microbes. It can spot these threats because it recognizes certain signals at the surface of dangerous cells. However, cancer cells often find ways to ‘hide’ from our immune system. Chimeric antigen receptors, or CARs, are receptors designed in a laboratory to attach to specific proteins that are found on a cancer cell. These receptors tell immune cells, such as T cells, to attack cancers. T cells that carry CARs are already used to treat people with blood cancers. Yet, these immune cells are not good at penetrating a solid tumor to kill the cells inside, which limits their use. Macrophages are a group of immune cells that can make their way inside tumors and travel to cancers that the rest of the immune system cannot reach. They defend our body by ‘swallowing’ harmful cells. Would it then be possible to use CARs to program macrophages to ‘eat’ cancer cells? Morrissey, Williamson et al. created a new type of CARs, named CAR-P, and introduced it in macrophages. These cells were then able to recognize and attack beads covered in proteins found on cancer cells. The modified macrophages could also limit the growth of live cancer cells in a dish by ‘biting’ and even ‘eating’ them. While these results are promising in the laboratory, the next step is to test whether these reprogrammed macrophages can recognize and fight cancers in living animals.
Collapse
Affiliation(s)
- Meghan A Morrissey
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, United States.,Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Adam P Williamson
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, United States.,Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Adriana M Steinbach
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, United States.,Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Edward W Roberts
- Department of Pathology, University of California, San Francisco, San Francisco, United States
| | - Nadja Kern
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, United States.,Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Mark B Headley
- Department of Pathology, University of California, San Francisco, San Francisco, United States
| | - Ronald D Vale
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, United States.,Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
99
|
Parhiz H, Khoshnejad M, Myerson JW, Hood E, Patel PN, Brenner JS, Muzykantov VR. Unintended effects of drug carriers: Big issues of small particles. Adv Drug Deliv Rev 2018; 130:90-112. [PMID: 30149885 PMCID: PMC6588191 DOI: 10.1016/j.addr.2018.06.023] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/11/2018] [Accepted: 06/26/2018] [Indexed: 02/06/2023]
Abstract
Humoral and cellular host defense mechanisms including diverse phagocytes, leukocytes, and immune cells have evolved over millions of years to protect the body from microbes and other external and internal threats. These policing forces recognize engineered sub-micron drug delivery systems (DDS) as such a threat, and react accordingly. This leads to impediment of the therapeutic action, extensively studied and discussed in the literature. Here, we focus on side effects of DDS interactions with host defenses. We argue that for nanomedicine to reach its clinical potential, the field must redouble its efforts in understanding the interaction between drug delivery systems and the host defenses, so that we can engineer safer interventions with the greatest potential for clinical success.
Collapse
Affiliation(s)
- Hamideh Parhiz
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Makan Khoshnejad
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacob W Myerson
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Hood
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Priyal N Patel
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacob S Brenner
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Vladimir R Muzykantov
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Targeted Therapeutics and Translational Nanomedicine (CT3N), University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
100
|
Evans MA, Huang PJ, Iwamoto Y, Ibsen KN, Chan EM, Hitomi Y, Ford PC, Mitragotri S. Macrophage-mediated delivery of light activated nitric oxide prodrugs with spatial, temporal and concentration control. Chem Sci 2018; 9:3729-3741. [PMID: 29780505 PMCID: PMC5939611 DOI: 10.1039/c8sc00015h] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 03/12/2018] [Indexed: 12/31/2022] Open
Abstract
Nitric oxide (NO) holds great promise as a treatment for cancer hypoxia, if its concentration and localization can be precisely controlled. Here, we report a "Trojan Horse" strategy to provide the necessary spatial, temporal, and dosage control of such drug-delivery therapies at targeted tissues. Described is a unique package consisting of (1) a manganese-nitrosyl complex, which is a photoactivated NO-releasing moiety (photoNORM), plus Nd3+-doped upconverting nanoparticles (Nd-UCNPs) incorporated into (2) biodegradable polymer microparticles that are taken up by (3) bone-marrow derived murine macrophages. Both the photoNORM [Mn(NO)dpaqNO2 ]BPh4(dpaqNO2 = 2-[N,N-bis(pyridin-2-yl-methyl)]-amino-N'-5-nitro-quinolin-8-yl-acetamido) and the Nd-UCNPs are activated by tissue-penetrating near-infrared (NIR) light at ∼800 nm. Thus, simultaneous therapeutic NO delivery and photoluminescence (PL) imaging can be achieved with a NIR diode laser source. The loaded microparticles are non-toxic to their macrophage hosts in the absence of light. The microparticle-carrying macrophages deeply penetrate into NIH-3T3/4T1 tumor spheroid models, and when the infiltrated spheroids are irradiated with NIR light, NO is released in quantifiable amounts while emission from the Nd-UCNPs provides images of microparticle location. Furthermore, varying the intensity of the NIR excitation allows photochemical control over NO release. Low doses reduce levels of hypoxia inducible factor 1 alpha (HIF-1α) in the tumor cells, while high doses are cytotoxic. The use of macrophages to carry microparticles with a NIR photo-activated theranostic payload into a tumor overcomes challenges often faced with therapeutic administration of NO and offers the potential of multiple treatment strategies with a single system.
Collapse
Affiliation(s)
- Michael A Evans
- Department of Chemistry and Biochemistry , University of California, Santa Barbara , Santa Barbara , CA , 93106 USA .
- Department of Chemical Engineering , Center for Bioengineering , University of California, Santa Barbara , Santa Barbara , CA , 93106 USA
- John A. Paulson School of Engineering and Applied Sciences , Harvard University , 29 Oxford St. , Cambridge , MA 02138 , USA .
| | - Po-Ju Huang
- Department of Chemistry and Biochemistry , University of California, Santa Barbara , Santa Barbara , CA , 93106 USA .
| | - Yuji Iwamoto
- Department of Chemistry and Biochemistry , Doshisha University , 1-3 Tatara Miyakodani, Kyotanabe , Kyoto 610-0394 , Japan
| | - Kelly N Ibsen
- Department of Chemical Engineering , Center for Bioengineering , University of California, Santa Barbara , Santa Barbara , CA , 93106 USA
| | - Emory M Chan
- Molecular Foundry , Lawrence Berkeley National Laboratory , Berkeley , CA , USA
| | - Yutaka Hitomi
- Department of Chemistry and Biochemistry , Doshisha University , 1-3 Tatara Miyakodani, Kyotanabe , Kyoto 610-0394 , Japan
| | - Peter C Ford
- Department of Chemistry and Biochemistry , University of California, Santa Barbara , Santa Barbara , CA , 93106 USA .
| | - Samir Mitragotri
- Department of Chemical Engineering , Center for Bioengineering , University of California, Santa Barbara , Santa Barbara , CA , 93106 USA
- John A. Paulson School of Engineering and Applied Sciences , Harvard University , 29 Oxford St. , Cambridge , MA 02138 , USA .
| |
Collapse
|