51
|
Xu T, Xie K, Wang C, Ivanovski S, Zhou Y. Immunomodulatory nanotherapeutic approaches for periodontal tissue regeneration. NANOSCALE 2023; 15:5992-6008. [PMID: 36896757 DOI: 10.1039/d2nr06149j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Periodontitis is an infection-induced inflammatory disease characterized by progressive destruction of tooth supporting tissues, which, if left untreated, can result in tooth loss. The destruction of periodontal tissues is primarily caused by an imbalance between the host immune protection and immune destruction mechanisms. The ultimate goal of periodontal therapy is to eliminate inflammation and promote the repair and regeneration of both hard and soft tissues, so as to restore the physiological structure and function of periodontium. Advancement in nanotechnologies has enabled the development of nanomaterials with immunomodulatory properties for regenerative dentistry. This review discusses the immune mechanisms of the major effector cells in the innate and adaptive immune systems, the physicochemical and biological properties of nanomaterials, and the research advancements in immunomodulatory nanotherapeutic approaches for the management of periodontitis and the regeneration of periodontal tissues. The current challenges, and prospects for future applications of nanomaterials are then discussed so that researchers at the intersections of osteoimmunology, regenerative dentistry and materiobiology will continue to advance the development of nanomaterials for improved periodontal tissue regeneration.
Collapse
Affiliation(s)
- Tian Xu
- School of Dentistry, The University of Queensland, QLD 4006, Australia.
| | - Kunke Xie
- Clinical Laboratory, Bo'Ai Hospital of Zhongshan, 6 Chenggui Road, East District, Zhongshan 528403, Guangdong, China
| | - Cong Wang
- School of Dentistry, The University of Queensland, QLD 4006, Australia.
| | - Sašo Ivanovski
- School of Dentistry, The University of Queensland, QLD 4006, Australia.
| | - Yinghong Zhou
- School of Dentistry, The University of Queensland, QLD 4006, Australia.
| |
Collapse
|
52
|
Bomb K, LeValley PJ, Woodward I, Cassel SE, Sutherland BP, Bhattacharjee A, Yun Z, Steen J, Kurdzo E, McCoskey J, Burris D, Levine K, Carbrello C, Lenhoff AM, Fromen CA, Kloxin AM. Cell therapy biomanufacturing: integrating biomaterial and flow-based membrane technologies for production of engineered T-cells. ADVANCED MATERIALS TECHNOLOGIES 2023; 8:2201155. [PMID: 37600966 PMCID: PMC10437131 DOI: 10.1002/admt.202201155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Indexed: 08/22/2023]
Abstract
Adoptive T-cell therapies (ATCTs) are increasingly important for the treatment of cancer, where patient immune cells are engineered to target and eradicate diseased cells. The biomanufacturing of ATCTs involves a series of time-intensive, lab-scale steps, including isolation, activation, genetic modification, and expansion of a patient's T-cells prior to achieving a final product. Innovative modular technologies are needed to produce cell therapies at improved scale and enhanced efficacy. In this work, well-defined, bioinspired soft materials were integrated within flow-based membrane devices for improving the activation and transduction of T cells. Hydrogel coated membranes (HCM) functionalized with cell-activating antibodies were produced as a tunable biomaterial for the activation of primary human T-cells. T-cell activation utilizing HCMs led to highly proliferative T-cells that expressed a memory phenotype. Further, transduction efficiency was improved by several fold over static conditions by using a tangential flow filtration (TFF) flow-cell, commonly used in the production of protein therapeutics, to transduce T-cells under flow. The combination of HCMs and TFF technology led to increased cell activation, proliferation, and transduction compared to current industrial biomanufacturing processes. The combined power of biomaterials with scalable flow-through transduction techniques provides future opportunities for improving the biomanufacturing of ATCTs.
Collapse
Affiliation(s)
- Kartik Bomb
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
| | - Paige J. LeValley
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
| | - Ian Woodward
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
| | - Samantha E. Cassel
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
| | | | | | - Zaining Yun
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
| | - Jonathan Steen
- EMD Millipore Corporation, Bedford, MA, an affiliate of Merck, Newark, DE
| | - Emily Kurdzo
- EMD Millipore Corporation, Bedford, MA, an affiliate of Merck, Newark, DE
| | - Jacob McCoskey
- EMD Millipore Corporation, Bedford, MA, an affiliate of Merck, Newark, DE
| | - David Burris
- Mechanical Engineering, University of Delaware, Newark, DE
| | - Kara Levine
- EMD Millipore Corporation, Bedford, MA, an affiliate of Merck, Newark, DE
| | | | - Abraham M. Lenhoff
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
| | | | - April M. Kloxin
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
- Material Science and Engineering, University of Delaware, Newark, DE
| |
Collapse
|
53
|
Yuan Z, Li Y, Zhang S, Wang X, Dou H, Yu X, Zhang Z, Yang S, Xiao M. Extracellular matrix remodeling in tumor progression and immune escape: from mechanisms to treatments. Mol Cancer 2023; 22:48. [PMID: 36906534 PMCID: PMC10007858 DOI: 10.1186/s12943-023-01744-8] [Citation(s) in RCA: 273] [Impact Index Per Article: 136.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/11/2023] [Indexed: 03/13/2023] Open
Abstract
The malignant tumor is a multi-etiological, systemic and complex disease characterized by uncontrolled cell proliferation and distant metastasis. Anticancer treatments including adjuvant therapies and targeted therapies are effective in eliminating cancer cells but in a limited number of patients. Increasing evidence suggests that the extracellular matrix (ECM) plays an important role in tumor development through changes in macromolecule components, degradation enzymes and stiffness. These variations are under the control of cellular components in tumor tissue via the aberrant activation of signaling pathways, the interaction of the ECM components to multiple surface receptors, and mechanical impact. Additionally, the ECM shaped by cancer regulates immune cells which results in an immune suppressive microenvironment and hinders the efficacy of immunotherapies. Thus, the ECM acts as a barrier to protect cancer from treatments and supports tumor progression. Nevertheless, the profound regulatory network of the ECM remodeling hampers the design of individualized antitumor treatment. Here, we elaborate on the composition of the malignant ECM, and discuss the specific mechanisms of the ECM remodeling. Precisely, we highlight the impact of the ECM remodeling on tumor development, including proliferation, anoikis, metastasis, angiogenesis, lymphangiogenesis, and immune escape. Finally, we emphasize ECM "normalization" as a potential strategy for anti-malignant treatment.
Collapse
Affiliation(s)
- Zhennan Yuan
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Yingpu Li
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Sifan Zhang
- Department of Neurobiology, Harbin Medical University, Harbin, 150081, China
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - He Dou
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xi Yu
- Department of Gynecological Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Zhiren Zhang
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.,Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin, 150001, China
| | - Shanshan Yang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150000, China.
| | - Min Xiao
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
54
|
Lee CY, Chooi WH, Ng S, Chew SY. Modulating neuroinflammation through molecular, cellular and biomaterial-based approaches to treat spinal cord injury. Bioeng Transl Med 2023; 8:e10389. [PMID: 36925680 PMCID: PMC10013833 DOI: 10.1002/btm2.10389] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/02/2022] [Accepted: 07/16/2022] [Indexed: 11/09/2022] Open
Abstract
The neuroinflammatory response that is elicited after spinal cord injury contributes to both tissue damage and reparative processes. The complex and dynamic cellular and molecular changes within the spinal cord microenvironment result in a functional imbalance of immune cells and their modulatory factors. To facilitate wound healing and repair, it is necessary to manipulate the immunological pathways during neuroinflammation to achieve successful therapeutic interventions. In this review, recent advancements and fresh perspectives on the consequences of neuroinflammation after SCI and modulation of the inflammatory responses through the use of molecular-, cellular-, and biomaterial-based therapies to promote tissue regeneration and functional recovery will be discussed.
Collapse
Affiliation(s)
- Cheryl Yi‐Pin Lee
- Institute of Molecular and Cell BiologyA*STAR Research EntitiesSingaporeSingapore
| | - Wai Hon Chooi
- Institute of Molecular and Cell BiologyA*STAR Research EntitiesSingaporeSingapore
| | - Shi‐Yan Ng
- Institute of Molecular and Cell BiologyA*STAR Research EntitiesSingaporeSingapore
| | - Sing Yian Chew
- School of Chemical and Biomedical EngineeringNanyang Technological UniversitySingaporeSingapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
- School of Materials Science and EngineeringNanyang Technological UniversitySingaporeSingapore
| |
Collapse
|
55
|
VanBlunk M, Srikanth V, Pandit SS, Kuznetsov AV, Brudno Y. Absorption rate governs cell transduction in dry macroporous scaffolds. Biomater Sci 2023; 11:2372-2382. [PMID: 36744434 PMCID: PMC10050106 DOI: 10.1039/d2bm01753a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Developing the next generation of cellular therapies will depend on fast, versatile, and efficient cellular reprogramming. Novel biomaterials will play a central role in this process by providing scaffolding and bioactive signals that shape cell fate and function. Previously, our lab reported that dry macroporous alginate scaffolds mediate retroviral transduction of primary T cells with efficiencies that rival the gold-standard clinical spinoculation procedures, which involve centrifugation on Retronectin-coated plates. This scaffold transduction required the scaffolds to be both macroporous and dry. Transduction by dry, macroporous scaffolds, termed "Drydux transduction," provides a fast and inexpensive method for transducing cells for cellular therapy, including for the production of CAR T cells. In this study, we investigate the mechanism of action by which Drydux transduction works through exploring the impact of pore size, stiffness, viral concentration, and absorption speed on transduction efficiency. We report that Drydux scaffolds with macropores ranging from 50-230 μm and with Young's moduli ranging from 25-620 kPa all effectively transduce primary T cells, suggesting that these parameters are not central to the mechanism of action, but also demonstrating that Drydux scaffolds can be tuned without losing functionality. Increasing viral concentrations led to significantly higher transduction efficiencies, demonstrating that increased cell-virus interaction is necessary for optimal transduction. Finally, we discovered that the rate with which the cell-virus solution is absorbed into the scaffold is closely correlated to viral transduction efficiency, with faster absorption producing significantly higher transduction. A computational model of liquid flow through porous media validates this finding by showing that increased fluid flow substantially increases collisions between virus particles and cells in a porous scaffold. Taken together, we conclude that the rate of liquid flow through the scaffolds, rather than pore size or stiffness, serves as a central regulator for efficient Drydux transduction.
Collapse
Affiliation(s)
- Madelyn VanBlunk
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA. .,Comparative Medicine Institute, North Carolina State University, USA
| | - Vishal Srikanth
- Department of Mechanical and Aerospace Engineering, North Carolina State University, USA
| | - Sharda S Pandit
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA. .,Comparative Medicine Institute, North Carolina State University, USA
| | - Andrey V Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, USA.,Comparative Medicine Institute, North Carolina State University, USA
| | - Yevgeny Brudno
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA. .,Comparative Medicine Institute, North Carolina State University, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, USA
| |
Collapse
|
56
|
Materials and extracellular matrix rigidity highlighted in tissue damages and diseases: Implication for biomaterials design and therapeutic targets. Bioact Mater 2023; 20:381-403. [PMID: 35784640 PMCID: PMC9234013 DOI: 10.1016/j.bioactmat.2022.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/05/2022] [Accepted: 06/10/2022] [Indexed: 11/21/2022] Open
Abstract
Rigidity (or stiffness) of materials and extracellular matrix has proven to be one of the most significant extracellular physicochemical cues that can control diverse cell behaviors, such as contractility, motility, and spreading, and the resultant pathophysiological phenomena. Many 2D materials engineered with tunable rigidity have enabled researchers to elucidate the roles of matrix biophysical cues in diverse cellular events, including migration, lineage specification, and mechanical memory. Moreover, the recent findings accumulated under 3D environments with viscoelastic and remodeling properties pointed to the importance of dynamically changing rigidity in cell fate control, tissue repair, and disease progression. Thus, here we aim to highlight the works related with material/matrix-rigidity-mediated cell and tissue behaviors, with a brief outlook into the studies on the effects of material/matrix rigidity on cell behaviors in 2D systems, further discussion of the events and considerations in tissue-mimicking 3D conditions, and then examination of the in vivo findings that concern material/matrix rigidity. The current discussion will help understand the material/matrix-rigidity-mediated biological phenomena and further leverage the concepts to find therapeutic targets and to design implantable materials for the treatment of damaged and diseased tissues. Discuss the cutting-edge findings on the role of matrix rigidity in dictating diverse cell behaviors. Underscore the dynamic matrix rigidity that interplays with cells, and the related pathophysiological phenomena. Illuminate the significance of matrix rigidity in clinically-relevant settings.
Collapse
|
57
|
Sarkar M, Nguyen T, Gundre E, Ogunlusi O, El-Sobky M, Giri B, Sarkar TR. Cancer-associated fibroblasts: The chief architect in the tumor microenvironment. Front Cell Dev Biol 2023; 11:1089068. [PMID: 36793444 PMCID: PMC9923123 DOI: 10.3389/fcell.2023.1089068] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
Stromal heterogeneity of tumor microenvironment (TME) plays a crucial role in malignancy and therapeutic resistance. Cancer-associated fibroblasts (CAFs) are one of the major players in tumor stroma. The heterogeneous sources of origin and subsequent impacts of crosstalk with breast cancer cells flaunt serious challenges before current therapies to cure triple-negative breast cancer (TNBC) and other cancers. The positive and reciprocal feedback of CAFs to induce cancer cells dictates their mutual synergy in establishing malignancy. Their substantial role in creating a tumor-promoting niche has reduced the efficacy of several anti-cancer treatments, including radiation, chemotherapy, immunotherapy, and endocrine therapy. Over the years, there has been an emphasis on understanding CAF-induced therapeutic resistance in order to enhance cancer therapy results. CAFs, in the majority of cases, employ crosstalk, stromal management, and other strategies to generate resilience in surrounding tumor cells. This emphasizes the significance of developing novel strategies that target particular tumor-promoting CAF subpopulations, which will improve treatment sensitivity and impede tumor growth. In this review, we discuss the current understanding of the origin and heterogeneity of CAFs, their role in tumor progression, and altering the tumor response to therapeutic agents in breast cancer. In addition, we also discuss the potential and possible approaches for CAF-mediated therapies.
Collapse
Affiliation(s)
- Mrinmoy Sarkar
- Department of Biology, Texas A&M University, College Station, TX, United States
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Tristan Nguyen
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Esheksha Gundre
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Olajumoke Ogunlusi
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Mohanad El-Sobky
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Biplab Giri
- Department of Physiology, University of Gour Banga, English Bazar, India
| | - Tapasree Roy Sarkar
- Department of Biology, Texas A&M University, College Station, TX, United States
| |
Collapse
|
58
|
Understanding How Cells Probe the World: A Preliminary Step towards Modeling Cell Behavior? Int J Mol Sci 2023; 24:ijms24032266. [PMID: 36768586 PMCID: PMC9916635 DOI: 10.3390/ijms24032266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/16/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Cell biologists have long aimed at quantitatively modeling cell function. Recently, the outstanding progress of high-throughput measurement methods and data processing tools has made this a realistic goal. The aim of this paper is twofold: First, to suggest that, while much progress has been done in modeling cell states and transitions, current accounts of environmental cues driving these transitions remain insufficient. There is a need to provide an integrated view of the biochemical, topographical and mechanical information processed by cells to take decisions. It might be rewarding in the near future to try to connect cell environmental cues to physiologically relevant outcomes rather than modeling relationships between these cues and internal signaling networks. The second aim of this paper is to review exogenous signals that are sensed by living cells and significantly influence fate decisions. Indeed, in addition to the composition of the surrounding medium, cells are highly sensitive to the properties of neighboring surfaces, including the spatial organization of anchored molecules and substrate mechanical and topographical properties. These properties should thus be included in models of cell behavior. It is also suggested that attempts at cell modeling could strongly benefit from two research lines: (i) trying to decipher the way cells encode the information they retrieve from environment analysis, and (ii) developing more standardized means of assessing the quality of proposed models, as was done in other research domains such as protein structure prediction.
Collapse
|
59
|
Hiltensperger M, Krackhardt AM. Current and future concepts for the generation and application of genetically engineered CAR-T and TCR-T cells. Front Immunol 2023; 14:1121030. [PMID: 36949949 PMCID: PMC10025359 DOI: 10.3389/fimmu.2023.1121030] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
Adoptive cell therapy (ACT) has seen a steep rise of new therapeutic approaches in its immune-oncology pipeline over the last years. This is in great part due to the recent approvals of chimeric antigen receptor (CAR)-T cell therapies and their remarkable efficacy in certain soluble tumors. A big focus of ACT lies on T cells and how to genetically modify them to target and kill tumor cells. Genetically modified T cells that are currently utilized are either equipped with an engineered CAR or a T cell receptor (TCR) for this purpose. Both strategies have their advantages and limitations. While CAR-T cell therapies are already used in the clinic, these therapies face challenges when it comes to the treatment of solid tumors. New designs of next-generation CAR-T cells might be able to overcome these hurdles. Moreover, CARs are restricted to surface antigens. Genetically engineered TCR-T cells targeting intracellular antigens might provide necessary qualities for the treatment of solid tumors. In this review, we will summarize the major advancements of the CAR-T and TCR-T cell technology. Moreover, we will cover ongoing clinical trials, discuss current challenges, and provide an assessment of future directions within the field.
Collapse
Affiliation(s)
- Michael Hiltensperger
- German Cancer Consortium (DKTK), partner site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
- IIIrd Medical Department, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- *Correspondence: Michael Hiltensperger, ; Angela M. Krackhardt,
| | - Angela M. Krackhardt
- German Cancer Consortium (DKTK), partner site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
- IIIrd Medical Department, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- *Correspondence: Michael Hiltensperger, ; Angela M. Krackhardt,
| |
Collapse
|
60
|
Wang K, Wen D, Xu X, Zhao R, Jiang F, Yuan S, Zhang Y, Gao Y, Li Q. Extracellular matrix stiffness-The central cue for skin fibrosis. Front Mol Biosci 2023; 10:1132353. [PMID: 36968277 PMCID: PMC10031116 DOI: 10.3389/fmolb.2023.1132353] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/20/2023] [Indexed: 03/29/2023] Open
Abstract
Skin fibrosis is a physiopathological process featuring the excessive deposition of extracellular matrix (ECM), which is the main architecture that provides structural support and constitutes the microenvironment for various cellular behaviors. Recently, increasing interest has been drawn to the relationship between the mechanical properties of the ECM and the initiation and modulation of skin fibrosis, with the engagement of a complex network of signaling pathways, the activation of mechanosensitive proteins, and changes in immunoregulation and metabolism. Simultaneous with the progression of skin fibrosis, the stiffness of ECM increases, which in turn perturbs mechanical and humoral homeostasis to drive cell fate toward an outcome that maintains and enhances the fibrosis process, thus forming a pro-fibrotic "positive feedback loop". In this review, we highlighted the central role of the ECM and its dynamic changes at both the molecular and cellular levels in skin fibrosis. We paid special attention to signaling pathways regulated by mechanical cues in ECM remodeling. We also systematically summarized antifibrotic interventions targeting the ECM, hopefully enlightening new strategies for fibrotic diseases.
Collapse
Affiliation(s)
- Kang Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dongsheng Wen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuewen Xu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rui Zhao
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Feipeng Jiang
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Shengqin Yuan
- School of Public Administration, Sichuan University, Chengdu, Sichuan, China
| | - Yifan Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| | - Ya Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| |
Collapse
|
61
|
Shi L, Lim JY, Kam LC. Substrate stiffness enhances human regulatory T cell induction and metabolism. Biomaterials 2023; 292:121928. [PMID: 36455488 PMCID: PMC9772289 DOI: 10.1016/j.biomaterials.2022.121928] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 11/09/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
Regulatory T cells (Tregs) provide an essential tolerance mechanism to suppress the immune response. Induced Tregs hold the potential to treat autoimmune diseases in adoptive therapy and can be produced with stimulating signals to CD3 and CD28 in presence of the cytokine TGF-β and IL-2. This report examines the modulation of human Treg induction by leveraging the ability of T cells to sense the mechanical stiffness of an activating substrate. Treg induction on polyacrylamide gels (PA-gels) was sensitive to the substrate's elastic modulus, increasing with greater material stiffness. Single-cell RNA-Seq analysis revealed that Treg induction on stiffer substrates involved greater use of oxidative phosphorylation (OXPHOS). Inhibition of ATP synthase significantly reduced the rate of Treg induction and abrogated the difference among gels while activation of AMPK (AMP-activated protein kinase) increased Treg induction on the softer sample but not on the harder sample. Treg induction is thus mechanosensitive and OXPHOS-dependent, providing new strategies for improving the production of these cells for cellular immunotherapy.
Collapse
Affiliation(s)
- Lingting Shi
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Ave, New York, NY, 10027, USA
| | - Jee Yoon Lim
- Department of Biological Sciences, Columbia University, 1212 Amsterdam Ave, New York, NY, 10027, USA
| | - Lance C Kam
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Ave, New York, NY, 10027, USA.
| |
Collapse
|
62
|
Alatoom A, ElGindi M, Sapudom J, Teo JCM. The T Cell Journey: A Tour de Force. Adv Biol (Weinh) 2023; 7:e2200173. [PMID: 36190140 DOI: 10.1002/adbi.202200173] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/30/2022] [Indexed: 11/07/2022]
Abstract
T cells act as the puppeteers in the adaptive immune response, and their dysfunction leads to the initiation and progression of pathological conditions. During their lifetime, T cells experience myriad forces that modulate their effector functions. These forces are imposed by interacting cells, surrounding tissues, and shear forces from fluid movement. In this review, a journey with T cells is made, from their development to their unique characteristics, including the early studies that uncovered their mechanosensitivity. Then the studies pertaining to the responses of T cell activation to changes in antigen-presenting cells' physical properties, to their immediate surrounding extracellular matrix microenvironment, and flow conditions are highlighted. In addition, it is explored how pathological conditions like the tumor microenvironment can hinder T cells and allow cancer cells to escape elimination.
Collapse
Affiliation(s)
- Aseel Alatoom
- Laboratory for Immuno Bioengineering Research and Applications Division of Engineering, New York University Abu Dhabi, Saadiyat Campus, P.O. Box 127788, Abu Dhabi, UAE.,Department of Mechanical Engineering Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
| | - Mei ElGindi
- Laboratory for Immuno Bioengineering Research and Applications Division of Engineering, New York University Abu Dhabi, Saadiyat Campus, P.O. Box 127788, Abu Dhabi, UAE
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications Division of Engineering, New York University Abu Dhabi, Saadiyat Campus, P.O. Box 127788, Abu Dhabi, UAE
| | - Jeremy C M Teo
- Laboratory for Immuno Bioengineering Research and Applications Division of Engineering, New York University Abu Dhabi, Saadiyat Campus, P.O. Box 127788, Abu Dhabi, UAE.,Department of Mechanical Engineering Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA.,Department of Biomedical Engineering Tandon School of Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
| |
Collapse
|
63
|
Blache U, Ford EM, Ha B, Rijns L, Chaudhuri O, Dankers PY, Kloxin AM, Snedeker JG, Gentleman E. Engineered hydrogels for mechanobiology. NATURE REVIEWS. METHODS PRIMERS 2022; 2:98. [PMID: 37461429 PMCID: PMC7614763 DOI: 10.1038/s43586-022-00179-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/17/2022] [Indexed: 07/20/2023]
Abstract
Cells' local mechanical environment can be as important in guiding cellular responses as many well-characterized biochemical cues. Hydrogels that mimic the native extracellular matrix can provide these mechanical cues to encapsulated cells, allowing for the study of their impact on cellular behaviours. Moreover, by harnessing cellular responses to mechanical cues, hydrogels can be used to create tissues in vitro for regenerative medicine applications and for disease modelling. This Primer outlines the importance and challenges of creating hydrogels that mimic the mechanical and biological properties of the native extracellular matrix. The design of hydrogels for mechanobiology studies is discussed, including appropriate choice of cross-linking chemistry and strategies to tailor hydrogel mechanical cues. Techniques for characterizing hydrogels are explained, highlighting methods used to analyze cell behaviour. Example applications for studying fundamental mechanobiological processes and regenerative therapies are provided, along with a discussion of the limitations of hydrogels as mimetics of the native extracellular matrix. The article ends with an outlook for the field, focusing on emerging technologies that will enable new insights into mechanobiology and its role in tissue homeostasis and disease.
Collapse
Affiliation(s)
- Ulrich Blache
- Fraunhofer Institute for Cell Therapy and Immunology and Fraunhofer Cluster of Excellence for Immune-Mediated Disease, Leipzig, Germany
| | - Eden M. Ford
- Department of Chemical and Biomolecular Engineering, University of Delaware, DE, USA
| | - Byunghang Ha
- Department of Mechanical Engineering, Stanford University, CA, USA
| | - Laura Rijns
- Institute for Complex Molecular Systems, Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, CA, USA
| | - Patricia Y.W. Dankers
- Institute for Complex Molecular Systems, Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | - April M. Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, DE, USA
- Department of Material Science and Engineering, University of Delaware, DE, USA
| | - Jess G. Snedeker
- University Hospital Balgrist and ETH Zurich, Zurich, Switzerland
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King’s College London, London SE1 9RT, UK
| |
Collapse
|
64
|
Lee M, Du H, Winer DA, Clemente-Casares X, Tsai S. Mechanosensing in macrophages and dendritic cells in steady-state and disease. Front Cell Dev Biol 2022; 10:1044729. [PMID: 36467420 PMCID: PMC9712790 DOI: 10.3389/fcell.2022.1044729] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022] Open
Abstract
Macrophages and dendritic cells are myeloid cells that play critical roles in immune responses. Macrophages help to maintain homeostasis through tissue regeneration and the clearance of dead cells, but also mediate inflammatory processes against invading pathogens. As the most potent antigen-presenting cells, dendritic cells are important in connecting innate to adaptive immune responses via activation of T cells, and inducing tolerance under physiological conditions. While it is known that macrophages and dendritic cells respond to biochemical cues in the microenvironment, the role of extracellular mechanical stimuli is becoming increasingly apparent. Immune cell mechanotransduction is an emerging field, where accumulating evidence suggests a role for extracellular physical cues coming from tissue stiffness in promoting immune cell recruitment, activation, metabolism and inflammatory function. Additionally, many diseases such as pulmonary fibrosis, cardiovascular disease, cancer, and cirrhosis are associated with changes to the tissue biophysical environment. This review will discuss current knowledge about the effects of biophysical cues including matrix stiffness, topography, and mechanical forces on macrophage and dendritic cell behavior under steady-state and pathophysiological conditions. In addition, we will also provide insight on molecular mediators and signaling pathways important in macrophage and dendritic cell mechanotransduction.
Collapse
Affiliation(s)
- Megan Lee
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Huixun Du
- Buck Institute for Research on Aging, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| | - Daniel A. Winer
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Pathology, University Health Network, Toronto, ON, Canada
- Buck Institute for Research on Aging, Novato, CA, United States
| | - Xavier Clemente-Casares
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Sue Tsai
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
65
|
Xu M, Zhang T, Xia R, Wei Y, Wei X. Targeting the tumor stroma for cancer therapy. Mol Cancer 2022; 21:208. [PMID: 36324128 PMCID: PMC9628074 DOI: 10.1186/s12943-022-01670-1] [Citation(s) in RCA: 128] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/13/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
Tumors are comprised of both cancer cells and surrounding stromal components. As an essential part of the tumor microenvironment, the tumor stroma is highly dynamic, heterogeneous and commonly tumor-type specific, and it mainly includes noncellular compositions such as the extracellular matrix and the unique cancer-associated vascular system as well as a wide variety of cellular components including activated cancer-associated fibroblasts, mesenchymal stromal cells, pericytes. All these elements operate with each other in a coordinated fashion and collectively promote cancer initiation, progression, metastasis and therapeutic resistance. Over the past few decades, numerous studies have been conducted to study the interaction and crosstalk between stromal components and neoplastic cells. Meanwhile, we have also witnessed an exponential increase in the investigation and recognition of the critical roles of tumor stroma in solid tumors. A series of clinical trials targeting the tumor stroma have been launched continually. In this review, we introduce and discuss current advances in the understanding of various stromal elements and their roles in cancers. We also elaborate on potential novel approaches for tumor-stroma-based therapeutic targeting, with the aim to promote the leap from bench to bedside.
Collapse
Affiliation(s)
- Maosen Xu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Tao Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Ruolan Xia
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China.
| |
Collapse
|
66
|
Jurj A, Ionescu C, Berindan-Neagoe I, Braicu C. The extracellular matrix alteration, implication in modulation of drug resistance mechanism: friends or foes? J Exp Clin Cancer Res 2022; 41:276. [PMID: 36114508 PMCID: PMC9479349 DOI: 10.1186/s13046-022-02484-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 09/01/2022] [Indexed: 11/10/2022] Open
Abstract
The extracellular matrix (ECM) is an important component of the tumor microenvironment (TME), having several important roles related to the hallmarks of cancer. In cancer, multiple components of the ECM have been shown to be altered. Although most of these alterations are represented by the increased or decreased quantity of the ECM components, changes regarding the functional alteration of a particular ECM component or of the ECM as a whole have been described. These alterations can be induced by the cancer cells directly or by the TME cells, with cancer-associated fibroblasts being of particular interest in this regard. Because the ECM has this wide array of functions in the tumor, preclinical and clinical studies have assessed the possibility of targeting the ECM, with some of them showing encouraging results. In the present review, we will highlight the most relevant ECM components presenting a comprehensive description of their physical, cellular and molecular properties which can alter the therapy response of the tumor cells. Lastly, some evidences regarding important biological processes were discussed, offering a more detailed understanding of how to modulate altered signalling pathways and to counteract drug resistance mechanisms in tumor cells.
Collapse
Affiliation(s)
- Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400337, Cluj-Napoca, Romania
| | - Calin Ionescu
- 7Th Surgical Department, Iuliu Hațieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012, Cluj-Napoca, Romania
- Surgical Department, Municipal Hospital, 400139, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400337, Cluj-Napoca, Romania.
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400337, Cluj-Napoca, Romania.
- Research Center for Oncopathology and Translational Medicine (CCOMT), George Emil Palade University of Medicine, Pharmacy, Sciences and Technology, 540139, Targu Mures, Romania.
| |
Collapse
|
67
|
Tian Q, Gao H, Ma Y, Zhu L, Zhou Y, Shen Y, Wang B. The regulatory roles of T helper cells in distinct extracellular matrix characterization in breast cancer. Front Immunol 2022; 13:871742. [PMID: 36159822 PMCID: PMC9493030 DOI: 10.3389/fimmu.2022.871742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 08/16/2022] [Indexed: 11/21/2022] Open
Abstract
Background Tumors are characterized by extracellular matrix (ECM) remodeling and stiffening. The ECM has been recognized as an important determinant of breast cancer progression and prognosis. Recent studies have revealed a strong link between ECM remodeling and immune cell infiltration in a variety of tumor types. However, the landscape and specific regulatory mechanisms between ECM and immune microenvironment in breast cancer have not been fully understood. Methods Using genomic data and clinical information of breast cancer patients obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, we conducted an extensive multi-omics analysis to explore the heterogeneity and prognostic significance of the ECM microenvironment. Masson and Sirius red staining were applied to quantify the contents of collagen in the ECM microenvironment. Tissue immunofluorescence (IF) staining was applied to identify T helper (Th) cells. Results We classified breast cancer patients into two ECM-clusters and three gene-clusters by consensus clustering. Significant heterogeneity in prognosis and immune cell infiltration have been found in these distinct clusters. Specifically, in the ECM-cluster with better prognosis, the expression levels of Th2 and regulatory T (Treg) cells were reduced, while the Th1, Th17 and T follicular helper (Tfh) cells-associated activities were significantly enhanced. The correlations between ECM characteristics and Th cells infiltration were then validated by clinical tissue samples from our hospital. The ECM-associated prognostic model was then constructed by 10 core prognostic genes and stratified breast cancer patients into two risk groups. Kaplan-Meier analysis showed that the overall survival (OS) of breast cancer patients in the high-risk group was significantly worse than that of the low-risk group. The risk scores for breast cancer patients obtained from our prognostic model were further confirmed to be associated with immune cell infiltration, tumor mutation burden (TMB) and stem cell indexes. Finally, the half-maximal inhibitory concentration (IC50) values of antitumor agents for patients in different risk groups were calculated to provide references for therapy targeting distinct ECM characteristics. Conclusion Our findings identify a novel strategy for breast cancer subtyping based on the ECM characterization and reveal the regulatory roles of Th cells in ECM remodeling. Targeting ECM remodeling and Th cells hold potential to be a therapeutic alternative for breast cancer in the future.
Collapse
Affiliation(s)
- Qi Tian
- Department of Radiology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Huan Gao
- Department of Medical Oncology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yingying Ma
- Department of Medical Oncology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Lizhe Zhu
- Department of Breast Surgery, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yan Zhou
- Department of Medical Oncology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yanwei Shen
- Department of Surgery Oncology, Shaanxi Provincial People’s Hospital, Xi’an, China
- *Correspondence: Yanwei Shen, ; Bo Wang,
| | - Bo Wang
- Center for Translational Medicine, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Yanwei Shen, ; Bo Wang,
| |
Collapse
|
68
|
Wheatley BA, Rey-Suarez I, Hourwitz MJ, Kerr S, Shroff H, Fourkas JT, Upadhyaya A. Nanotopography modulates cytoskeletal organization and dynamics during T cell activation. Mol Biol Cell 2022; 33:ar88. [PMID: 35830602 PMCID: PMC9582624 DOI: 10.1091/mbc.e21-12-0601] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Exposure to MHC-antigen complexes on the surface of antigen-presenting cells (APCs) activates T cells, inducing the formation of the immune synapse (IS). Antigen detection at the APC surface is thus a critical step in the adaptive immune response. The physical properties of antigen-presenting surfaces encountered by T cells in vivo are believed to modulate T cell activation and proliferation. Although stiffness and ligand mobility influence IS formation, the effect of the complex topography of the APC surface on this process is not well understood. Here we investigate how nanotopography modulates cytoskeletal dynamics and signaling during the early stages of T cell activation using high-resolution fluorescence microscopy on nanofabricated surfaces with parallel nanoridges of different spacings. We find that although nanoridges reduce the maximum spread area as compared with cells on flat surfaces, the ridges enhance the accumulation of actin and the signaling kinase ZAP-70 at the IS. Actin polymerization is more dynamic in the presence of ridges, which influence the directionality of both actin flows and microtubule (MT) growth. Our results demonstrate that the topography of the activating surface exerts both global effects on T cell morphology and local changes in actin and MT dynamics, collectively influencing T cell signaling.
Collapse
Affiliation(s)
- Brittany A Wheatley
- Department of Integrative Structural and Computational Biology and.,Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, Jupiter, FL 33458
| | - Ivan Rey-Suarez
- Institute for Physical Science and Technology, University of Maryland, College Park, MD 20742
| | - Matt J Hourwitz
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742
| | - Sarah Kerr
- Department of Physics, University of Colorado, Boulder, CO 80302
| | - Hari Shroff
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892
| | - John T Fourkas
- Institute for Physical Science and Technology, University of Maryland, College Park, MD 20742.,Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742.,Maryland Quantum Materials Center, University of Maryland, College Park, MD 20742
| | - Arpita Upadhyaya
- Institute for Physical Science and Technology, University of Maryland, College Park, MD 20742.,Department of Physics, University of Maryland, College Park, MD 20742
| |
Collapse
|
69
|
Oh J, Xia X, Wong WKR, Wong SHD, Yuan W, Wang H, Lai CHN, Tian Y, Ho YP, Zhang H, Zhang Y, Li G, Lin Y, Bian L. The Effect of the Nanoparticle Shape on T Cell Activation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107373. [PMID: 35297179 DOI: 10.1002/smll.202107373] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/08/2022] [Indexed: 06/14/2023]
Abstract
The mechanism of extracellular ligand nano-geometry in ex vivo T cell activation for immunotherapy remains elusive. Herein, the authors demonstrate large aspect ratio (AR) of gold nanorods (AuNRs) conjugated on cell culture substrate enhancing both murine and human T cell activation through the nanoscale anisotropic presentation of stimulatory ligands (anti-CD3(αCD3) and anti-CD28(αCD28) antibodies). AuNRs with large AR bearing αCD3 and αCD28 antibodies significantly promote T cell expansion and key cytokine secretion including interleukin-2 (IL-2), interferon-gamma (IFN-γ), and tumor necrosis factor-alpha (TNF-α). High membrane tension observed in large AR AuNRs regulates actin filament and focal adhesion assembly and develops maturation-related morphological features in T cells such as membrane ruffle formation, cell spreading, and large T cell receptor (TCR) cluster formation. Anisotropic stimulatory ligand presentation promotes differentiation of naïve CD8+ T cells toward the effector phenotype inducing CD137 expression upon co-culture with human cervical carcinoma. The findings suggest the importance of manipulating extracellular ligand nano-geometry in optimizing T cell behaviors to enhance therapeutic outcomes.
Collapse
Affiliation(s)
- Jiwon Oh
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Xingyu Xia
- Department of Mechanical Engineering, University of Hong Kong, Hong Kong, 999077, China
| | - Wai Ki Ricky Wong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, 999077, China
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | - Siu Hong Dexter Wong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, 999077, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Weihao Yuan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, 999077, China
- Department of Orthopedic and Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, Hong Kong, 999077, China
| | - Haixing Wang
- Department of Orthopedic and Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, Hong Kong, 999077, China
| | - Chun Him Nathanael Lai
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Ye Tian
- Department of Mechanical Engineering, University of Hong Kong, Hong Kong, 999077, China
| | - Yi-Ping Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Honglu Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Yuan Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Gang Li
- Department of Orthopedic and Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, Hong Kong, 999077, China
| | - Yuan Lin
- Department of Mechanical Engineering, University of Hong Kong, Hong Kong, 999077, China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New territories, Hong Kong, 999077, China
- HKU-Shenzhen Institute of Research and Innovation (HKU-SIRI), Shenzhen, Guang Dong, 518000, China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| |
Collapse
|
70
|
Jain N, Lord JM, Vogel V. Mechanoimmunology: Are inflammatory epigenetic states of macrophages tuned by biophysical factors? APL Bioeng 2022; 6:031502. [PMID: 36051106 PMCID: PMC9427154 DOI: 10.1063/5.0087699] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022] Open
Abstract
Many inflammatory diseases that are responsible for a majority of deaths are still uncurable, in part as the underpinning pathomechanisms and how to combat them is still poorly understood. Tissue-resident macrophages play pivotal roles in the maintenance of tissue homeostasis, but if they gradually convert to proinflammatory phenotypes, or if blood-born proinflammatory macrophages persist long-term after activation, they contribute to chronic inflammation and fibrosis. While biochemical factors and how they regulate the inflammatory transcriptional response of macrophages have been at the forefront of research to identify targets for therapeutic interventions, evidence is increasing that physical factors also tune the macrophage phenotype. Recently, several mechanisms have emerged as to how physical factors impact the mechanobiology of macrophages, from the nuclear translocation of transcription factors to epigenetic modifications, perhaps even DNA methylation. Insight into the mechanobiology of macrophages and associated epigenetic modifications will deliver novel therapeutic options going forward, particularly in the context of increased inflammation with advancing age and age-related diseases. We review here how biophysical factors can co-regulate pro-inflammatory gene expression and epigenetic modifications and identify knowledge gaps that require urgent attention if this therapeutic potential is to be realized.
Collapse
Affiliation(s)
- Nikhil Jain
- Authors to whom correspondence should be addressed: and
| | | | - Viola Vogel
- Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
71
|
Wang H, Xu T, Fu Y, Wang Z, Leeson MS, Jiang J, Liu T. Liquid Crystal Biosensors: Principles, Structure and Applications. BIOSENSORS 2022; 12:639. [PMID: 36005035 PMCID: PMC9406233 DOI: 10.3390/bios12080639] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/06/2022] [Accepted: 08/12/2022] [Indexed: 12/31/2022]
Abstract
Liquid crystals (LCs) have been widely used as sensitive elements to construct LC biosensors based on the principle that specific bonding events between biomolecules can affect the orientation of LC molecules. On the basis of the sensing interface of LC molecules, LC biosensors can be classified into three types: LC-solid interface sensing platforms, LC-aqueous interface sensing platforms, and LC-droplet interface sensing platforms. In addition, as a signal amplification method, the combination of LCs and whispering gallery mode (WGM) optical microcavities can provide higher detection sensitivity due to the extremely high quality factor and the small mode volume of the WGM optical microcavity, which enhances the interaction between the light field and biotargets. In this review, we present an overview of the basic principles, the structure, and the applications of LC biosensors. We discuss the important properties of LC and the principle of LC biosensors. The different geometries of LCs in the biosensing systems as well as their applications in the biological detection are then described. The fabrication and the application of the LC-based WGM microcavity optofluidic sensor in the biological detection are also introduced. Finally, challenges and potential research opportunities in the development of LC-based biosensors are discussed.
Collapse
Affiliation(s)
- Haonan Wang
- School of Precision Instruments and Opto-Electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Tianhua Xu
- School of Precision Instruments and Opto-Electronics Engineering, Tianjin University, Tianjin 300072, China
- School of Engineering, University of Warwick, Coventry CV4 7AL, UK
| | - Yaoxin Fu
- School of Precision Instruments and Opto-Electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Ziyihui Wang
- School of Precision Instruments and Opto-Electronics Engineering, Tianjin University, Tianjin 300072, China
- School of Electrical and Electronics Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Mark S. Leeson
- School of Engineering, University of Warwick, Coventry CV4 7AL, UK
| | - Junfeng Jiang
- School of Precision Instruments and Opto-Electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Tiegen Liu
- School of Precision Instruments and Opto-Electronics Engineering, Tianjin University, Tianjin 300072, China
| |
Collapse
|
72
|
Guo T, He C, Venado A, Zhou Y. Extracellular Matrix Stiffness in Lung Health and Disease. Compr Physiol 2022; 12:3523-3558. [PMID: 35766837 PMCID: PMC10088466 DOI: 10.1002/cphy.c210032] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The extracellular matrix (ECM) provides structural support and imparts a wide variety of environmental cues to cells. In the past decade, a growing body of work revealed that the mechanical properties of the ECM, commonly known as matrix stiffness, regulate the fundamental cellular processes of the lung. There is growing appreciation that mechanical interplays between cells and associated ECM are essential to maintain lung homeostasis. Dysregulation of ECM-derived mechanical signaling via altered mechanosensing and mechanotransduction pathways is associated with many common lung diseases. Matrix stiffening is a hallmark of lung fibrosis. The stiffened ECM is not merely a sequelae of lung fibrosis but can actively drive the progression of fibrotic lung disease. In this article, we provide a comprehensive view on the role of matrix stiffness in lung health and disease. We begin by summarizing the effects of matrix stiffness on the function and behavior of various lung cell types and on regulation of biomolecule activity and key physiological processes, including host immune response and cellular metabolism. We discuss the potential mechanisms by which cells probe matrix stiffness and convert mechanical signals to regulate gene expression. We highlight the factors that govern matrix stiffness and outline the role of matrix stiffness in lung development and the pathogenesis of pulmonary fibrosis, pulmonary hypertension, asthma, chronic obstructive pulmonary disease (COPD), and lung cancer. We envision targeting of deleterious matrix mechanical cues for treatment of fibrotic lung disease. Advances in technologies for matrix stiffness measurements and design of stiffness-tunable matrix substrates are also explored. © 2022 American Physiological Society. Compr Physiol 12:3523-3558, 2022.
Collapse
Affiliation(s)
- Ting Guo
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Alabama, USA.,Department of Respiratory Medicine, the Second Xiangya Hospital, Central-South University, Changsha, Hunan, China
| | - Chao He
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Alabama, USA
| | - Aida Venado
- Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Yong Zhou
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Alabama, USA
| |
Collapse
|
73
|
Wang MS, Hu Y, Sanchez EE, Xie X, Roy NH, de Jesus M, Winer BY, Zale EA, Jin W, Sachar C, Lee JH, Hong Y, Kim M, Kam LC, Salaita K, Huse M. Mechanically active integrins target lytic secretion at the immune synapse to facilitate cellular cytotoxicity. Nat Commun 2022; 13:3222. [PMID: 35680882 PMCID: PMC9184626 DOI: 10.1038/s41467-022-30809-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 05/19/2022] [Indexed: 01/25/2023] Open
Abstract
Cytotoxic lymphocytes fight pathogens and cancer by forming immune synapses with infected or transformed target cells and then secreting cytotoxic perforin and granzyme into the synaptic space, with potent and specific killing achieved by this focused delivery. The mechanisms that establish the precise location of secretory events, however, remain poorly understood. Here we use single cell biophysical measurements, micropatterning, and functional assays to demonstrate that localized mechanotransduction helps define the position of secretory events within the synapse. Ligand-bound integrins, predominantly the αLβ2 isoform LFA-1, function as spatial cues to attract lytic granules containing perforin and granzyme and induce their fusion with the plasma membrane for content release. LFA-1 is subjected to pulling forces within secretory domains, and disruption of these forces via depletion of the adaptor molecule talin abrogates cytotoxicity. We thus conclude that lymphocytes employ an integrin-dependent mechanical checkpoint to enhance their cytotoxic power and fidelity.
Collapse
Affiliation(s)
- Mitchell S Wang
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Pharmacology Program, Weill-Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Yuesong Hu
- Department of Chemistry, Emory University, Atlanta, GA, USA
| | - Elisa E Sanchez
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Biochemistry and Molecular Biology Program, Weill-Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Xihe Xie
- Neuroscience Program, Weill-Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Nathan H Roy
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Miguel de Jesus
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Benjamin Y Winer
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elizabeth A Zale
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Weiyang Jin
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Chirag Sachar
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Joanne H Lee
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Yeonsun Hong
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Lance C Kam
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, GA, USA
| | - Morgan Huse
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
74
|
Göhring J, Schrangl L, Schütz GJ, Huppa JB. Mechanosurveillance: Tiptoeing T Cells. Front Immunol 2022; 13:886328. [PMID: 35693808 PMCID: PMC9178122 DOI: 10.3389/fimmu.2022.886328] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/19/2022] [Indexed: 11/28/2022] Open
Abstract
Efficient scanning of tissue that T cells encounter during their migratory life is pivotal to protective adaptive immunity. In fact, T cells can detect even a single antigenic peptide/MHC complex (pMHC) among thousands of structurally similar yet non-stimulatory endogenous pMHCs on the surface of antigen-presenting cells (APCs) or target cells. Of note, the glycocalyx of target cells, being composed of proteoglycans and bulky proteins, is bound to affect and even modulate antigen recognition by posing as a physical barrier. T cell-resident microvilli are actin-rich membrane protrusions that puncture through such barriers and thereby actively place the considerably smaller T-cell antigen receptors (TCRs) in close enough proximity to APC-presented pMHCs so that productive interactions may occur efficiently yet under force. We here review our current understanding of how the plasticity of T-cell microvilli and physicochemical properties of the glycocalyx may affect early events in T-cell activation. We assess insights gained from studies on T-cell plasma membrane ultrastructure and provide an update on current efforts to integrate biophysical aspects such as the amplitude and directionality of TCR-imposed mechanical forces and the distribution and lateral mobility of plasma membrane-resident signaling molecules into a more comprehensive view on sensitized T-cell antigen recognition.
Collapse
Affiliation(s)
- Janett Göhring
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Institute of Applied Physics, TU Wien, Vienna, Austria
- *Correspondence: Janett Göhring,
| | | | | | - Johannes B. Huppa
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
75
|
Yan J, Gundsambuu B, Krasowska M, Platts K, Facal Marina P, Gerber C, Barry SC, Blencowe A. Injectable Diels-Alder cycloaddition hydrogels with tuneable gelation, stiffness and degradation for the sustained release of T-lymphocytes. J Mater Chem B 2022; 10:3329-3343. [PMID: 35380575 DOI: 10.1039/d2tb00274d] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Engineered T-cell therapies have proven highly efficacious for the treatment of haematological cancers, but translation of this success to solid tumours has been limited, in part, due to difficulties in maintaining high doses at specific target sites. Hydrogel delivery systems that provide a sustained release of T-cells at the target site are emerging as a promising strategy. Therefore, in this study we aimed to develop an injectable hydrogel that gels in situ via efficient Diels-Alder cycloaddition (DAC) chemistry and provides a sustained release of T-cells through gradual hydrolysis of the hydrogel matrix. Hydrogels were prepared via the DAC between fulvene and maleimide functionalised poly(ethylene glycol) (PEG) derivatives. By adjusting the concentration and molecular weight of the functionalised PEGs in the hydrogel formulation the in vitro gelation time (Tgel), initial Young's modulus (E) and degradation time (Td) could be tailored from 15-150 min, 5-179 kPa and 7-114 h, respectively. Prior to gelation, the formulations could be readily injected through narrow gauge (26 G) needles with the working time correlating closely with the Tgel. A 5 wt% hydrogel formation with conjugated cyclic RGD motif was found to be optimal for the encapsulation and release of CD3+ T-cells with a near linear release profile and >70% cell viability over the first 4 d and release continuing out to 7 d. With their tuneable Tgel, Td and stiffness, the DAC hydrogels provide the opportunity to control the release period and profile of encapsulated cells.
Collapse
Affiliation(s)
- Jie Yan
- Applied Chemistry and Translational Biomaterials (ACTB) Group, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia.
| | - Batjargal Gundsambuu
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Marta Krasowska
- Surface Interaction and Soft Matter (SISM) Group, Future Industries Institute (FII), UniSA STEM, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Kirsten Platts
- Applied Chemistry and Translational Biomaterials (ACTB) Group, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia.
| | - Paula Facal Marina
- Applied Chemistry and Translational Biomaterials (ACTB) Group, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia.
| | - Cobus Gerber
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Simon C Barry
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, South Australia 5005, Australia.,Department of Gastroenterology, Women's and Children's Hospital, SA Health, Adelaide, South Australia 5006, Australia
| | - Anton Blencowe
- Applied Chemistry and Translational Biomaterials (ACTB) Group, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia.
| |
Collapse
|
76
|
Ghassemi S, Durgin JS, Nunez-Cruz S, Patel J, Leferovich J, Pinzone M, Shen F, Cummins KD, Plesa G, Cantu VA, Reddy S, Bushman FD, Gill SI, O'Doherty U, O'Connor RS, Milone MC. Rapid manufacturing of non-activated potent CAR T cells. Nat Biomed Eng 2022; 6:118-128. [PMID: 35190680 PMCID: PMC8860360 DOI: 10.1038/s41551-021-00842-6] [Citation(s) in RCA: 153] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 12/17/2021] [Indexed: 12/14/2022]
Abstract
Chimaeric antigen receptor (CAR) T cells can generate durable clinical responses in B-cell haematologic malignancies. The manufacturing of these T cells typically involves their activation, followed by viral transduction and expansion ex vivo for at least 6 days. However, the activation and expansion of CAR T cells leads to their progressive differentiation and the associated loss of anti-leukaemic activity. Here we show that functional CAR T cells can be generated within 24 hours from T cells derived from peripheral blood without the need for T-cell activation or ex vivo expansion, and that the efficiency of viral transduction in this process is substantially influenced by the formulation of the medium and the surface area-to-volume ratio of the culture vessel. In mouse xenograft models of human leukaemias, the rapidly generated non-activated CAR T cells exhibited higher anti-leukaemic in vivo activity per cell than the corresponding activated CAR T cells produced using the standard protocol. The rapid manufacturing of CAR T cells may reduce production costs and broaden their applicability.
Collapse
Affiliation(s)
- Saba Ghassemi
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Joseph S Durgin
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Selene Nunez-Cruz
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jai Patel
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Leferovich
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marilia Pinzone
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Feng Shen
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katherine D Cummins
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gabriela Plesa
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vito Adrian Cantu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shantan Reddy
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Saar I Gill
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Una O'Doherty
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Roddy S O'Connor
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael C Milone
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
77
|
Han B, Song Y, Park J, Doh J. Nanomaterials to improve cancer immunotherapy based on ex vivo engineered T cells and NK cells. J Control Release 2022; 343:379-391. [DOI: 10.1016/j.jconrel.2022.01.049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/15/2022] [Accepted: 01/31/2022] [Indexed: 02/08/2023]
|
78
|
Tello-Lafoz M, de Jesus MM, Huse M. Harder, better, faster, stronger: biochemistry and biophysics in the immunosurveillance concert. Trends Immunol 2022; 43:96-105. [PMID: 34973924 PMCID: PMC8810625 DOI: 10.1016/j.it.2021.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/03/2021] [Accepted: 12/03/2021] [Indexed: 02/08/2023]
Abstract
Antitumor immunosurveillance is triggered by immune cell recognition of characteristic biochemical signals on the surfaces of cancer cells. Recent data suggest that the mechanical properties of cancer cells influence the strength of these signals, with physically harder target cells (more rigid) eliciting better, faster, and stronger cytotoxic responses against metastasis. Using analogies to a certain electronic music duo, we argue that the biophysical properties of cancer cells and their environment can adjust the volume and tone of the antitumor immune response. We also consider the potential influence of biomechanics-based immunosurveillance in disease progression and posit that targeting the biophysical properties of cancer cells in concert with their biochemical features could increase the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Maria Tello-Lafoz
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Miguel M de Jesus
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Morgan Huse
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
79
|
Al-Aghbar MA, Jainarayanan AK, Dustin ML, Roffler SR. The interplay between membrane topology and mechanical forces in regulating T cell receptor activity. Commun Biol 2022; 5:40. [PMID: 35017678 PMCID: PMC8752658 DOI: 10.1038/s42003-021-02995-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/21/2021] [Indexed: 12/20/2022] Open
Abstract
T cells are critically important for host defense against infections. T cell activation is specific because signal initiation requires T cell receptor (TCR) recognition of foreign antigen peptides presented by major histocompatibility complexes (pMHC) on antigen presenting cells (APCs). Recent advances reveal that the TCR acts as a mechanoreceptor, but it remains unclear how pMHC/TCR engagement generates mechanical forces that are converted to intracellular signals. Here we propose a TCR Bending Mechanosignal (TBM) model, in which local bending of the T cell membrane on the nanometer scale allows sustained contact of relatively small pMHC/TCR complexes interspersed among large surface receptors and adhesion molecules on the opposing surfaces of T cells and APCs. Localized T cell membrane bending is suggested to increase accessibility of TCR signaling domains to phosphorylation, facilitate selective recognition of agonists that form catch bonds, and reduce noise signals associated with slip bonds.
Collapse
Affiliation(s)
- Mohammad Ameen Al-Aghbar
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Department of Translational Medicine, Sidra Medicine, Doha, Qatar
| | - Ashwin K Jainarayanan
- Interdisciplinary Bioscience Doctoral Training Program and Exeter College, University of Oxford, Oxford, UK
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
80
|
Engineering tumor stromal mechanics for improved T cell therapy. Biochim Biophys Acta Gen Subj 2022; 1866:130095. [DOI: 10.1016/j.bbagen.2022.130095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/23/2021] [Accepted: 01/14/2022] [Indexed: 12/17/2022]
|
81
|
Rømer AMA, Thorseth ML, Madsen DH. Immune Modulatory Properties of Collagen in Cancer. Front Immunol 2021; 12:791453. [PMID: 34956223 PMCID: PMC8692250 DOI: 10.3389/fimmu.2021.791453] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/22/2021] [Indexed: 12/22/2022] Open
Abstract
During tumor growth the extracellular matrix (ECM) undergoes dramatic remodeling. The normal ECM is degraded and substituted with a tumor-specific ECM, which is often of higher collagen density and increased stiffness. The structure and collagen density of the tumor-specific ECM has been associated with poor prognosis in several types of cancer. However, the reason for this association is still largely unknown. Collagen can promote cancer cell growth and migration, but recent studies have shown that collagens can also affect the function and phenotype of various types of tumor-infiltrating immune cells such as tumor-associated macrophages (TAMs) and T cells. This suggests that tumor-associated collagen could have important immune modulatory functions within the tumor microenvironment, affecting cancer progression as well as the efficacy of cancer immunotherapy. The effects of tumor-associated collagen on immune cells could help explain why a high collagen density in tumors is often correlated with a poor prognosis. Knowledge about immune modulatory functions of collagen could potentially identify targets for improving current cancer therapies or for development of new treatments. In this review, the current knowledge about the ability of collagen to influence T cell activity will be summarized. This includes direct interactions with T cells as well as induction of immune suppressive activity in other immune cells such as macrophages. Additionally, the potential effects of collagen on the efficacy of cancer immunotherapy will be discussed.
Collapse
Affiliation(s)
- Anne Mette Askehøj Rømer
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark.,Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Marie-Louise Thorseth
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Daniel Hargbøl Madsen
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
82
|
Taeb S, Ashrafizadeh M, Zarrabi A, Rezapoor S, Musa AE, Farhood B, Najafi M. Role of Tumor Microenvironment in Cancer Stem Cells Resistance to Radiotherapy. Curr Cancer Drug Targets 2021; 22:18-30. [PMID: 34951575 DOI: 10.2174/1568009622666211224154952] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/29/2021] [Accepted: 08/24/2021] [Indexed: 11/22/2022]
Abstract
Cancer is a chronic disorder that involves several elements of both the tumor and the host stromal cells. At present, the complex relationship between the various factors presents in the tumor microenvironment (TME) and tumor cells, as well as immune cells located within the TME, is still poorly known. Within the TME, the crosstalk of these factors and immune cells essentially determines how a tumor reacts to the treatment and how the tumor can ultimately be destroyed, remain dormant, or develop and metastasize. Also, in TME, reciprocal crosstalk between cancer-associated fibroblasts (CAFs), extracellular matrix (ECM), hypoxia-inducible factor (HIF) intensifies the proliferation capacity of cancer stem cells (CSCs). CSCs are subpopulation of cells that reside within the tumor bulk and have the capacity to self-renew, differentiate, and repair DNA damage. These characteristics make CSCs develop resistance to a variety of treatments, such as radiotherapy (RT). RT is a frequent and often curative treatment for local cancer which mediates tumor elimination by cytotoxic actions. Also, cytokines and growth factors that are released into TME, have been involved in the activation of tumor radioresistance and the induction of different immune cells, altering local immune responses. In this review, we discuss the pivotal role of TME in resistance of CSCs to RT.
Collapse
Affiliation(s)
- Shahram Taeb
- Department of Radiology, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 , Turkey
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Turkey
| | - Saeed Rezapoor
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Iran
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences (International Campus), Iran
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences., Iraq
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Iran
| |
Collapse
|
83
|
Aghlara-Fotovat S, Nash A, Kim B, Krencik R, Veiseh O. Targeting the extracellular matrix for immunomodulation: applications in drug delivery and cell therapies. Drug Deliv Transl Res 2021; 11:2394-2413. [PMID: 34176099 DOI: 10.1007/s13346-021-01018-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2021] [Indexed: 12/12/2022]
Abstract
Host immune cells interact bi-directionally with their extracellular matrix (ECM) to receive and deposit molecular signals, which orchestrate cellular activation, proliferation, differentiation, and function to maintain healthy tissue homeostasis. In response to pathogens or damage, immune cells infiltrate diseased sites and synthesize critical ECM molecules such as glycoproteins, proteoglycans, and glycosaminoglycans to promote healing. When the immune system misidentifies pathogens or fails to survey damaged cells effectively, maladies such as chronic inflammation, autoimmune diseases, and cancer can develop. In these conditions, it is essential to restore balance to the body through modulation of the immune system and the ECM. This review details the components of dysregulated ECM implicated in pathogenic environments and therapeutic approaches to restore tissue homeostasis. We evaluate emerging strategies to overcome inflamed, immune inhibitory, and otherwise diseased microenvironments, including mechanical stimulation, targeted proteases, adoptive cell therapy, mechanomedicine, and biomaterial-based cell therapeutics. We highlight various strategies that have produced efficacious responses in both pre-clinical and human trials and identify additional opportunities to develop next-generation interventions. Significantly, we identify a need for therapies to address dense or fibrotic tissue for the treatment of organ tissue damage and various cancer subtypes. Finally, we conclude that therapeutic techniques that disrupt, evade, or specifically target the pathogenic microenvironment have a high potential for improving therapeutic outcomes and should be considered a priority for immediate exploration. A schematic showing the various methods of extracellular matrix disruption/targeting in both fibrotic and cancerous environments. a Biomaterial-based cell therapy can be used to deliver anti-inflammatory cytokines, chemotherapeutics, or other factors for localized, slow release of therapeutics. b Mechanotherapeutics can be used to inhibit the deposition of molecules such as collagen that affect stiffness. c Ablation of the ECM and target tissue can be accomplished via mechanical degradation such as focused ultrasound. d Proteases can be used to improve the distribution of therapies such as oncolytic virus. e Localization of therapeutics such as checkpoint inhibitors can be improved with the targeting of specific ECM components, reducing off-target effects and toxicity.
Collapse
Affiliation(s)
| | - Amanda Nash
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Boram Kim
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Robert Krencik
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA.
| |
Collapse
|
84
|
He Y, Liu T, Dai S, Xu Z, Wang L, Luo F. Tumor-Associated Extracellular Matrix: How to Be a Potential Aide to Anti-tumor Immunotherapy? Front Cell Dev Biol 2021; 9:739161. [PMID: 34733848 PMCID: PMC8558531 DOI: 10.3389/fcell.2021.739161] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/28/2021] [Indexed: 02/05/2023] Open
Abstract
The development of cancer immunotherapy, particularly immune checkpoint blockade therapy, has made major breakthroughs in the therapy of cancers. However, less than one-third of the cancer patients obtain significant and long-lasting therapeutic effects by cancer immunotherapy. Over the past few decades, cancer-related inflammations have been gradually more familiar to us. It’s known that chronic inflammation in tumor microenvironment (TME) plays a predominant role in tumor immunosuppression. Tumor-associated extracellular matrix (ECM), as a core member of TME, has been a research hotspot recently. A growing number of studies indicate that tumor-associated ECM is one of the major obstacles to realizing more successful cases of cancer immunotherapy. In this review, we discussed the potential application of tumor-associated ECM in the cancer immunity and its aide potentialities to anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Yingying He
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,Oncology Department, People's Hospital of Deyang City, Deyang, China
| | - Tao Liu
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
| | - Shuang Dai
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zihan Xu
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Li Wang
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Feng Luo
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
85
|
Puech PH, Bongrand P. Mechanotransduction as a major driver of cell behaviour: mechanisms, and relevance to cell organization and future research. Open Biol 2021; 11:210256. [PMID: 34753321 PMCID: PMC8586914 DOI: 10.1098/rsob.210256] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/18/2021] [Indexed: 01/04/2023] Open
Abstract
How do cells process environmental cues to make decisions? This simple question is still generating much experimental and theoretical work, at the border of physics, chemistry and biology, with strong implications in medicine. The purpose of mechanobiology is to understand how biochemical and physical cues are turned into signals through mechanotransduction. Here, we review recent evidence showing that (i) mechanotransduction plays a major role in triggering signalling cascades following cell-neighbourhood interaction; (ii) the cell capacity to continually generate forces, and biomolecule properties to undergo conformational changes in response to piconewton forces, provide a molecular basis for understanding mechanotransduction; and (iii) mechanotransduction shapes the guidance cues retrieved by living cells and the information flow they generate. This includes the temporal and spatial properties of intracellular signalling cascades. In conclusion, it is suggested that the described concepts may provide guidelines to define experimentally accessible parameters to describe cell structure and dynamics, as a prerequisite to take advantage of recent progress in high-throughput data gathering, computer simulation and artificial intelligence, in order to build a workable, hopefully predictive, account of cell signalling networks.
Collapse
Affiliation(s)
- Pierre-Henri Puech
- Lab Adhesion and Inflammation (LAI), Inserm UMR 1067, CNRS UMR 7333, Aix-Marseille Université UM61, Marseille, France
| | - Pierre Bongrand
- Lab Adhesion and Inflammation (LAI), Inserm UMR 1067, CNRS UMR 7333, Aix-Marseille Université UM61, Marseille, France
| |
Collapse
|
86
|
Zhou B, Gao Z, Liu W, Wu X, Wang W. Important role of mechanical microenvironment on macrophage dysfunction during keloid pathogenesis. Exp Dermatol 2021; 31:375-380. [PMID: 34665886 DOI: 10.1111/exd.14473] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 09/27/2021] [Accepted: 10/17/2021] [Indexed: 12/17/2022]
Abstract
Keloid is considered as a tumor-like skin disease with multiple aetiologies including immunological factors and mechanical microenvironment. Macrophages are plastic and diverse immune cells that play a critical role in maintaining tissue homeostasis by removing dead cells, debris, pathogens and repairing tissues after inflammation. The imbalance of M1/M2 macrophages and disturbances in macrophage functions can steer the progression of chronic inflammation and lead to the development of pathological fibrosis in keloid disease. Recently, it has been shown that macrophages are sensitive to mechanical signals, especially stretching tension and tissue stiffness, which can determine macrophage polarization and functions. Higher stretching tension is known to be an important pathogenic factor of keloid, and the formation of keloid will lead to an increase in tissue stiffness. As little is known about the underlying reasons of macrophages dysfunction in keloid, an understanding of how the mechanical microenvironment interacting with macrophages and affecting their behaviours may help provide mechanism insights into keloid pathogenesis. We thus hypothesize that the synergistic effect of stretching tension and matrix stiffness may contribute to the major pathophysiological niche attributes of macrophages' in vivo mechanical microenvironment in keloids. These mechanism insights of how macrophages sense and respond to their mechanical microenvironment would propel the development of novel strategies for keloid treatment.
Collapse
Affiliation(s)
- Boya Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
| | - Zhen Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
| | - Wei Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China
| | - Xiaoli Wu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
| | - Wenbo Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
| |
Collapse
|
87
|
Matrix biophysical cues direct mesenchymal stromal cell functions in immunity. Acta Biomater 2021; 133:126-138. [PMID: 34365041 DOI: 10.1016/j.actbio.2021.07.075] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 07/14/2021] [Accepted: 07/30/2021] [Indexed: 12/25/2022]
Abstract
Hydrogels have been used to design synthetic matrices that capture salient features of matrix microenvironments to study and control cellular functions. Recent advances in understanding of both extracellular matrix biology and biomaterial design have shown that biophysical cues are powerful mediators of cell biology, especially that of mesenchymal stromal cells (MSCs). MSCs have been tested in many clinical trials because of their ability to modulate immune cells in different pathological conditions. While roles of biophysical cues in MSC biology have been studied in the context of multilineage differentiation, their significance in regulating immunomodulatory functions of MSCs is just beginning to be elucidated. This review first describes design principles behind how biophysical cues in native microenvironments influence the ability of MSCs to regulate immune cell production and functions. We will then discuss how biophysical cues can be leveraged to optimize cell isolation, priming, and delivery, which can help improve the success of MSC therapy for immunomodulation. Finally, a perspective is presented on how implementing biophysical cues in MSC potency assay can be important in predicting clinical outcomes. STATEMENT OF SIGNIFICANCE: Stromal cells of mesenchymal origin are known to direct immune cell functions in vivo by secreting paracrine mediators. This property has been leveraged in developing mesenchymal stromal cell (MSC)-based therapeutics by adoptive transfer to treat immunological rejection and tissue injuries, which have been tested in over one thousand clinical trials to date, but with mixed success. Advances in biomaterial design have enabled precise control of biophysical cues based on how stromal cells interact with the extracellular matrix in microenvironments in situ. Investigators have begun to use this approach to understand how different matrix biophysical parameters, such as fiber orientation, porosity, dimensionality, and viscoelasticity impact stromal cell-mediated immunomodulation. The insights gained from this effort can potentially be used to precisely define the microenvironmental cues for isolation, priming, and delivery of MSCs, which can be tailored based on different disease indications for optimal therapeutic outcomes.
Collapse
|
88
|
Jones JO, Moody WM, Shields JD. Microenvironmental modulation of the developing tumour: an immune-stromal dialogue. Mol Oncol 2021; 15:2600-2633. [PMID: 32741067 PMCID: PMC8486574 DOI: 10.1002/1878-0261.12773] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/03/2020] [Accepted: 07/27/2020] [Indexed: 12/17/2022] Open
Abstract
Successful establishment of a tumour relies on a cascade of interactions between cancer cells and stromal cells within an evolving microenvironment. Both immune and nonimmune cellular components are key factors in this process, and the individual players may change their role from tumour elimination to tumour promotion as the microenvironment develops. While the tumour-stroma crosstalk present in an established tumour is well-studied, aspects in the early tumour or premalignant microenvironment have received less attention. This is in part due to the challenges in studying this process in the clinic or in mouse models. Here, we review the key anti- and pro-tumour factors in the early microenvironment and discuss how understanding this process may be exploited in the clinic.
Collapse
Affiliation(s)
- James O. Jones
- MRC Cancer UnitHutchison/MRC Research CentreUniversity of CambridgeCambridgeUK
- Department of OncologyCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - William M. Moody
- MRC Cancer UnitHutchison/MRC Research CentreUniversity of CambridgeCambridgeUK
| | | |
Collapse
|
89
|
Sachar C, Kam LC. Probing T Cell 3D Mechanosensing With Magnetically-Actuated Structures. Front Immunol 2021; 12:704693. [PMID: 34566962 PMCID: PMC8458571 DOI: 10.3389/fimmu.2021.704693] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/23/2021] [Indexed: 12/02/2022] Open
Abstract
The ability of cells to recognize and respond to the mechanical properties of their environment is of increasing importance in T cell physiology. However, initial studies in this direction focused on planar hydrogel and elastomer surfaces, presenting several challenges in interpretation including difficulties in separating mechanical stiffness from changes in chemistry needed to modulate this property. We introduce here the use of magnetic fields to change the structural rigidity of microscale elastomer pillars loaded with superparamagnetic nanoparticles, independent of substrate chemistry. This magnetic modulation of rigidity, embodied as the pillar spring constant, changed the interaction of mouse naïve CD4+ T cells from a contractile morphology to one involving deep embedding into the array. Furthermore, increasing spring constant was associated with higher IL-2 secretion, showing a functional impact on mechanosensing. The system introduced here thus separates local substrate stiffness and long-range structural rigidity, revealing new facets of T cell interaction with their environment.
Collapse
Affiliation(s)
- Chirag Sachar
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Lance C Kam
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| |
Collapse
|
90
|
CAR T-Cells Depend on the Coupling of NADH Oxidation with ATP Production. Cells 2021; 10:cells10092334. [PMID: 34571983 PMCID: PMC8472053 DOI: 10.3390/cells10092334] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/20/2021] [Accepted: 08/25/2021] [Indexed: 12/29/2022] Open
Abstract
The metabolic milieu of solid tumors provides a barrier to chimeric antigen receptor (CAR) T-cell therapies. Excessive lactate or hypoxia suppresses T-cell growth, through mechanisms including NADH buildup and the depletion of oxidized metabolites. NADH is converted into NAD+ by the enzyme Lactobacillus brevis NADH Oxidase (LbNOX), which mimics the oxidative function of the electron transport chain without generating ATP. Here we determine if LbNOX promotes human CAR T-cell metabolic activity and antitumor efficacy. CAR T-cells expressing LbNOX have enhanced oxygen as well as lactate consumption and increased pyruvate production. LbNOX renders CAR T-cells resilient to lactate dehydrogenase inhibition. But in vivo in a model of mesothelioma, CAR T-cell's expressing LbNOX showed no increased antitumor efficacy over control CAR T-cells. We hypothesize that T cells in hostile environments face dual metabolic stressors of excessive NADH and insufficient ATP production. Accordingly, futile T-cell NADH oxidation by LbNOX is insufficient to promote tumor clearance.
Collapse
|
91
|
Record J, Saeed MB, Venit T, Percipalle P, Westerberg LS. Journey to the Center of the Cell: Cytoplasmic and Nuclear Actin in Immune Cell Functions. Front Cell Dev Biol 2021; 9:682294. [PMID: 34422807 PMCID: PMC8375500 DOI: 10.3389/fcell.2021.682294] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
Actin cytoskeletal dynamics drive cellular shape changes, linking numerous cell functions to physiological and pathological cues. Mutations in actin regulators that are differentially expressed or enriched in immune cells cause severe human diseases known as primary immunodeficiencies underscoring the importance of efficienct actin remodeling in immune cell homeostasis. Here we discuss recent findings on how immune cells sense the mechanical properties of their environement. Moreover, while the organization and biochemical regulation of cytoplasmic actin have been extensively studied, nuclear actin reorganization is a rapidly emerging field that has only begun to be explored in immune cells. Based on the critical and multifaceted contributions of cytoplasmic actin in immune cell functionality, nuclear actin regulation is anticipated to have a large impact on our understanding of immune cell development and functionality.
Collapse
Affiliation(s)
- Julien Record
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Mezida B. Saeed
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Tomas Venit
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates
| | - Piergiorgio Percipalle
- Science Division, Biology Program, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Lisa S. Westerberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
92
|
Zhang B, Su Y, Zhou J, Zheng Y, Zhu D. Toward a Better Regeneration through Implant-Mediated Immunomodulation: Harnessing the Immune Responses. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100446. [PMID: 34117732 PMCID: PMC8373114 DOI: 10.1002/advs.202100446] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/08/2021] [Indexed: 05/06/2023]
Abstract
Tissue repair/regeneration, after implantation or injury, involves comprehensive physiological processes wherein immune responses play a crucial role to enable tissue restoration, amidst the immune cells early-stage response to tissue damages. These cells break down extracellular matrix, clear debris, and secret cytokines to orchestrate regeneration. However, the immune response can also lead to abnormal tissue healing or scar formation if not well directed. This review first introduces the general immune response post injury, with focus on the major immune cells including neutrophils, macrophages, and T cells. Next, a variety of implant-mediated immunomodulation strategies to regulate immune response through physical, chemical, and biological cues are discussed. At last, various scaffold-facilitated regenerations of different tissue types, such as, bone, cartilage, blood vessel, and nerve system, by harnessing the immunomodulation are presented. Therefore, the most recent data in biomaterials and immunomodulation is presented here in a bid to shape expert perspectives, inspire researchers to go in new directions, and drive development of future strategies focusing on targeted, sequential, and dynamic immunomodulation elicited by implants.
Collapse
Affiliation(s)
- Ben Zhang
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew York11794USA
| | - Yingchao Su
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew York11794USA
| | - Juncen Zhou
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew York11794USA
| | - Yufeng Zheng
- Department of Materials Science and EngineeringCollege of EngineeringPeking UniversityBeijing100871China
| | - Donghui Zhu
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew York11794USA
| |
Collapse
|
93
|
Abdeen AA, Cosgrove BD, Gersbach CA, Saha K. Integrating Biomaterials and Genome Editing Approaches to Advance Biomedical Science. Annu Rev Biomed Eng 2021; 23:493-516. [PMID: 33909475 DOI: 10.1146/annurev-bioeng-122019-121602] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The recent discovery and subsequent development of the CRISPR-Cas9 (clustered regularly interspaced short palindromic repeat-CRISPR-associated protein 9) platform as a precise genome editing tool have transformed biomedicine. As these CRISPR-based tools have matured, multiple stages of the gene editing process and the bioengineering of human cells and tissues have advanced. Here, we highlight recent intersections in the development of biomaterials and genome editing technologies. These intersections include the delivery of macromolecules, where biomaterial platforms have been harnessed to enable nonviral delivery of genome engineering tools to cells and tissues in vivo. Further, engineering native-like biomaterial platforms for cell culture facilitates complex modeling of human development and disease when combined with genome engineering tools. Deeper integration of biomaterial platforms in these fields could play a significant role in enabling new breakthroughs in the application of gene editing for the treatment of human disease.
Collapse
Affiliation(s)
- Amr A Abdeen
- Department of Biomedical Engineering, Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin 53715, USA
| | - Brian D Cosgrove
- Department of Biomedical Engineering and Center for Advanced Genomic Technologies, Duke University, Durham, North Carolina 27708, USA;
| | - Charles A Gersbach
- Department of Biomedical Engineering and Center for Advanced Genomic Technologies, Duke University, Durham, North Carolina 27708, USA;
- Department of Surgery, Duke University Medical Center, Durham, North Carolina 27708, USA
| | - Krishanu Saha
- Department of Biomedical Engineering, Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin 53715, USA
- McPherson Eye Research Institute, Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA;
| |
Collapse
|
94
|
Belhabib I, Zaghdoudi S, Lac C, Bousquet C, Jean C. Extracellular Matrices and Cancer-Associated Fibroblasts: Targets for Cancer Diagnosis and Therapy? Cancers (Basel) 2021; 13:3466. [PMID: 34298680 PMCID: PMC8303391 DOI: 10.3390/cancers13143466] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/25/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Solid cancer progression is dictated by neoplastic cell features and pro-tumoral crosstalks with their microenvironment. Stroma modifications, such as fibroblast activation into cancer-associated fibroblasts (CAFs) and extracellular matrix (ECM) remodeling, are now recognized as critical events for cancer progression and as potential therapeutic or diagnostic targets. The recent appreciation of the key, complex and multiple roles of the ECM in cancer and of the CAF diversity, has revolutionized the field and raised innovative but challenging questions. Here, we rapidly present CAF heterogeneity in link with their specific ECM remodeling features observed in cancer, before developing each of the impacts of such ECM modifications on tumor progression (survival, angiogenesis, pre-metastatic niche, chemoresistance, etc.), and on patient prognosis. Finally, based on preclinical studies and recent results obtained from clinical trials, we highlight key mechanisms or proteins that are, or may be, used as potential therapeutic or diagnostic targets, and we report and discuss benefits, disappointments, or even failures, of recently reported stroma-targeting strategies.
Collapse
Affiliation(s)
| | | | | | | | - Christine Jean
- Centre de Recherche en Cancérologie de Toulouse (CRCT), INSERM U1037, Université Toulouse III Paul Sabatier, ERL5294 CNRS, 31037 Toulouse, France; (I.B.); (S.Z.); (C.L.); (C.B.)
| |
Collapse
|
95
|
Kim SHJ, Hammer DA. Integrin cross-talk modulates stiffness-independent motility of CD4+ T lymphocytes. Mol Biol Cell 2021; 32:1749-1757. [PMID: 34232700 PMCID: PMC8684734 DOI: 10.1091/mbc.e21-03-0131] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
To carry out their physiological responsibilities, CD4+ T lymphocytes interact with various tissues of different mechanical properties. Recent studies suggest that T cells migrate upstream on surfaces expressing intracellular adhesion molecule-1 (ICAM-1) through interaction with leukocyte function-associated antigen-1 (αLβ2) (LFA-1) integrins. LFA-1 likely behaves as a mechanosensor, and thus we hypothesized that substrate mechanics might affect the ability of LFA-1 to support upstream migration of T cells under flow. Here we measured motility of CD4+ T lymphocytes on polyacrylamide gels with predetermined stiffnesses containing ICAM-1, vascular cell adhesion molecule-1 (VCAM-1), or a 1:1 mixture of VCAM-1/ICAM-1. Under static conditions, we found that CD4+ T cells exhibit an increase in motility on ICAM-1, but not on VCAM-1 or VCAM-1/ICAM-1 mixed, surfaces as a function of matrix stiffness. The mechanosensitivity of T-cell motility on ICAM-1 is overcome when VLA-4 (very late antigen-4 [α4β1]) is ligated with soluble VCAM-1. Last, we observed that CD4+ T cells migrate upstream under flow on ICAM-1-functionalized hydrogels, independent of substrate stiffness. In summary, we show that CD4+ T cells under no flow respond to matrix stiffness through LFA-1, and that the cross-talk of VLA-4 and LFA-1 can compensate for deformable substrates. Interestingly, CD4+ T lymphocytes migrated upstream on ICAM-1 regardless of the substrate stiffness, suggesting that flow can compensate for substrate stiffness.
Collapse
Affiliation(s)
- Sarah Hyun Ji Kim
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104
| | - Daniel A Hammer
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
96
|
Abstract
T cell activation is a critical event in the adaptive immune response, indispensable for cell-mediated and humoral immunity as well as for immune regulation. Recent years have witnessed an emerging trend emphasizing the essential role that physical force and mechanical properties play at the T cell interface. In this review, we integrate current knowledge of T cell antigen recognition and the different models of T cell activation from the perspective of mechanobiology, focusing on the interaction between the T cell receptor (TCR) and the peptide-major histocompatibility complex (pMHC) antigen. We address the shortcomings of TCR affinity alone in explaining T cell functional outcomes and the rising status of force-regulated TCR bond lifetimes, most notably the TCR catch bond. Ultimately, T cell activation and the ensuing physiological responses result from mechanical interaction between TCRs and the pMHC. 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)
- Baoyu Liu
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA; , ,
| | - Elizabeth M Kolawole
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA; , ,
| | - Brian D Evavold
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA; , ,
| |
Collapse
|
97
|
Britten CM, Shalabi A, Hoos A. Industrializing engineered autologous T cells as medicines for solid tumours. Nat Rev Drug Discov 2021; 20:476-488. [PMID: 33833444 DOI: 10.1038/s41573-021-00175-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2021] [Indexed: 02/06/2023]
Abstract
Cell therapy is one of the fastest growing areas in the pharmaceutical industry, with considerable therapeutic potential. However, substantial challenges regarding the utility of these therapies will need to be addressed before they can become mainstream medicines with applicability similar to that of small molecules or monoclonal antibodies. Engineered T cells have achieved success in the treatment of blood cancers, with four chimeric antigen receptor (CAR)-T cell therapies now approved for the treatment of B cell malignancies based on their unprecedented efficacy in clinical trials. However, similar results have not yet been achieved in the treatment of the much larger patient population with solid tumours. For cell therapies to become mainstream medicines, they may need to offer transformational clinical effects for patients and be applicable in disease settings that remain unaddressed by simpler approaches. This Perspective provides an industry perspective on the progress achieved by engineered T cell therapies to date and the opportunities and current barriers for accessing broader patient populations, and discusses the solutions and new development strategies required to fully industrialize the therapeutic potential of engineered T cells as medicines.
Collapse
Affiliation(s)
- Cedrik M Britten
- Oncology R&D, GlaxoSmithKline, Stevenage, UK
- Immatics Biotechnologies, Munich, Germany
| | - Aiman Shalabi
- Oncology R&D, GlaxoSmithKline, Philadelphia, PA, USA
| | - Axel Hoos
- Oncology R&D, GlaxoSmithKline, Philadelphia, PA, USA.
| |
Collapse
|
98
|
Bhingardive V, Kossover A, Iraqi M, Khand B, Le Saux G, Porgador A, Schvartzman M. Antibody-Functionalized Nanowires: A Tuner for the Activation of T Cells. NANO LETTERS 2021; 21:4241-4248. [PMID: 33989498 DOI: 10.1021/acs.nanolett.1c00245] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
T cells sense both chemical cues delivered by antigen molecules and physical cues delivered by the environmental elasticity and topography; yet, it is still largely unclear how these cues cumulatively regulate the immune activity of T cells. Here, we engineered a nanoscale platform for ex vivo stimulation of T cells based on antigen-functionalized nanowires. The nanowire topography and elasticity, as well as the immobilized antigens, deliver the physical and chemical cues, respectively, enabling the systematic study of the integrated effect of these cues on a T cell's immune response. We found that T cells sense both the topography and bending modulus of the nanowires and modulate their signaling, degranulation, and cytotoxicity with the variation in these physical features. Our study provides an important insight into the physical mechanism of T cell activation and paves the way to novel nanomaterials for the controlled ex vivo activation of T cells in immunotherapy.
Collapse
|
99
|
Zhu Y, Reinach PS, Ge C, Li Y, Wu B, Xie Q, Tong L, Chen W. Corneal Collagen Cross-Linking Pretreatment Mitigates Injury-Induced Inflammation, Hemangiogenesis and Lymphangiogenesis In Vivo. Transl Vis Sci Technol 2021; 10:11. [PMID: 34550310 PMCID: PMC8475280 DOI: 10.1167/tvst.10.5.11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Purpose The purpose of this study was to evaluate if corneal collagen cross-linking (CXL) pretreatment dampens suture-induced hemangiogenesis and lymphangiogenesis driven by inflammation. Methods Four weeks after CXL pretreatment, suture emplacement was performed in rats. The time dependent effects were compared of this procedure in three groups: (1) suture-induced neovascularization (SNV group); (2) CXL treatment prior to suture-induced neovascularization (CXL + SNV group); (3) Normal control (NC group). Serial morphometric measurements evaluated suture-induced hemangiogenesis and lymphangiogenesis. CD45 and CD68 immunofluorescent staining pattern changes determined immune cell activation, stromal leucocyte, and macrophage infiltration. The real-time quantitative polymerase chain reaction (RT-qPCR) determined angiogenic and lymphangiogenic gene expression level changes. Western blots evaluated protein expression levels of vascular endothelial cell CD31 and lymphatic vessel endothelial hyaluronan receptor (LYVE-1). Results On days 7 and 14 after suture emplacement, the rises in angiogenesis, lymphangiogenesis, CD45+ and CD68+ cell infiltration were less in the CXL pretreated (CXL + SNV) group than in the untreated (SNV) group. Angiogenic and lymphangiogenic mRNA levels and CD31 and LYVE-1 protein and proinflammatory cytokines were also suppressed, confirming that CXL pretreatment improved the wound healing response. Conclusions CXL pretreatment inhibits injury-induced angiogenesis and lymphangiogenesis. These reductions suggest that prior CXL therapy decrease ocular inflammation reactivated by secondary trauma. Translational Relevance CXL pretreatment induces increases in stromal stiffness which in turn reduces trauma or microbial driven increases in inflammation, angiogenesis, and lymphangiogenesis. These beneficial effects suggest that this novel procedure may improve therapeutic management of trauma-induced corneal disease in a clinical setting.
Collapse
Affiliation(s)
- Yirui Zhu
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Zhejiang, China.,Eye Center of the 2nd Affiliated Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, China
| | - Peter S Reinach
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Zhejiang, China
| | - Chaoxiang Ge
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Zhejiang, China
| | - Yun Li
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Zhejiang, China
| | - Biao Wu
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Zhejiang, China
| | - Qi Xie
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Zhejiang, China
| | - Louis Tong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Singapore National Eye Centre, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Wei Chen
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Zhejiang, China
| |
Collapse
|
100
|
Hammink R, Weiden J, Voerman D, Popelier C, Eggermont LJ, Schluck M, Figdor CG, Verdoes M. Semiflexible Immunobrushes Induce Enhanced T Cell Activation and Expansion. ACS APPLIED MATERIALS & INTERFACES 2021; 13:16007-16018. [PMID: 33797875 PMCID: PMC8045021 DOI: 10.1021/acsami.0c21994] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
A variety of bioactive materials developed to expand T cells for adoptive transfer into cancer patients are currently evaluated in the clinic. In most cases, T cell activating biomolecules are attached to rigid surfaces or matrices and form a static interface between materials and the signaling receptors on the T cells. We hypothesized that a T cell activating polymer brush interface might better mimic the cell surface of a natural antigen-presenting cell, facilitating receptor movement and concomitant advantageous mechanical forces to provide enhanced T cell activating capacities. Here, as a proof of concept, we synthesized semiflexible polyisocyanopeptide (PIC) polymer-based immunobrushes equipped with T cell activating agonistic anti-CD3 (αCD3) and αCD28 antibodies placed on magnetic microbeads. We demonstrated enhanced efficiency of ex vivo expansion of activated primary human T cells even at very low numbers of stimulating antibodies compared to rigid beads. Importantly, the immunobrush architecture appeared crucial for this improved T cell activating capacity. Immunobrushes outperform current benchmarks by producing higher numbers of T cells exhibiting a combination of beneficial phenotypic characteristics, such as reduced exhaustion marker expression, high cytokine production, and robust expression of cytotoxic hallmarks. This study indicates that semiflexible immunobrushes have great potential in making T cell-based immunotherapies more effective.
Collapse
Affiliation(s)
- Roel Hammink
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Jorieke Weiden
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Dion Voerman
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Carlijn Popelier
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
| | - Loek J. Eggermont
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Marjolein Schluck
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Carl G. Figdor
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Martijn Verdoes
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| |
Collapse
|