1
|
Xie X, Liu Y, Yang L, Zhang Z, Li H, Zhang W, Liu H, Wang H, Shao Z. Impaired LTB4-induced neutrophil chemotactic directionality in myelodysplastic neoplasms patients. Hematology 2025; 30:2483551. [PMID: 40170327 DOI: 10.1080/16078454.2025.2483551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 03/19/2025] [Indexed: 04/03/2025] Open
Abstract
OBJECTIVES Myelodysplastic neoplasm (MDS) patients are at a high risk of infections, contributing significantly to morbidity and mortality. While neutrophil dysfunction is considered a primary factor, specific functional defects remain elusive. METHODS We conducted a comprehensive study involving 90 participants, including controls and de novo MDS patients. We utilized the TAXIScan-FL system to evaluate neutrophil chemotaxis towards leukotriene B4 (LTB4). The global reactive oxygen species (ROS) production by neutrophils were measured by chemiluminescence assay, neutrophil alkaline phosphatase (NAP) was evaluated by enzymatic staining. RESULTS MDS patients, irrespective of absolute neutrophil count (ANC) levels, exhibited elevated empirical antimicrobial therapy (EAT) rate compared to controls. Neutrophil migration towards LTB4 was notably impaired, demonstrating reduced velocity and directionality. Interestingly, MDS patients with high ANC still displayed poor directionality and slower migration. MDS patients also had compromised ROS and NAP activity. A noteworthy correlation was observed between EAT rate and chemotactic directionality parameters. CONCLUSION MDS patients face a heightened risk of infection, potentially attributed to impaired neutrophil chemotactic speed and directionality, alongside compromised ROS and NAP activity. Notably, chemotactic directionality emerged as a pivotal factor correlated with antimicrobial therapy. These insights hold significant clinical implications for managing infections in MDS patients, underscoring the importance of targeting specific neutrophil defects for more effective therapeutic strategies.
Collapse
Affiliation(s)
- Xinyan Xie
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, People's Republic of China
- Tianjin Institute of Hematology, Tianjin, People's Republic of China
| | - Yumei Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, People's Republic of China
- Tianjin Institute of Hematology, Tianjin, People's Republic of China
| | - Liyan Yang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, People's Republic of China
- Tianjin Institute of Hematology, Tianjin, People's Republic of China
| | - Zhe Zhang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, People's Republic of China
- Tianjin Institute of Hematology, Tianjin, People's Republic of China
| | - Hongzhao Li
- Tianjin Children's Hospital, Tianjin, People's Republic of China
| | - Wei Zhang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, People's Republic of China
- Tianjin Institute of Hematology, Tianjin, People's Republic of China
| | - Hong Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, People's Republic of China
- Tianjin Institute of Hematology, Tianjin, People's Republic of China
| | - Huaquan Wang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, People's Republic of China
- Tianjin Institute of Hematology, Tianjin, People's Republic of China
| | - Zonghong Shao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, People's Republic of China
- Tianjin Institute of Hematology, Tianjin, People's Republic of China
| |
Collapse
|
2
|
Schrope JH, Horn A, Lazorchak K, Tinnen CW, Stevens JJ, Farooqui M, Robertson T, Li J, Bennin D, Juang T, Ahmed A, Li C, Huttenlocher A, Beebe DJ. Confinement by Liquid-Liquid Interface Replicates In Vivo Neutrophil Deformations and Elicits Bleb-Based Migration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2414024. [PMID: 40151891 DOI: 10.1002/advs.202414024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 03/05/2025] [Indexed: 03/29/2025]
Abstract
Leukocytes forge paths through interstitial spaces by exerting forces to overcome confining mechanical pressures provided by surrounding cells. While such mechanical cues regulate leukocyte motility, engineering an in vitro system that models the deformable cellular environment encountered in vivo has been challenging. Here, microchannels are constructed with a liquid-liquid interface that exerts confining pressures similar to cells in tissues, and thus, is deformable by cell-generated forces. Consequently, the balance between migratory cell-generated and interfacial pressures determines the degree of confinement. Pioneer cells that first contact the interfacial barrier require greater deformation forces to forge a path for migration, and as a result migrate slower than trailing cells. Critically, resistive pressures are tunable by controlling the curvature of the liquid interface, which regulates motility. By granting cells autonomy in determining their confinement, and tuning environmental resistance, interfacial deformations match those of surrounding cells in vivo during interstitial neutrophil migration in a larval zebrafish model. It is discovered that neutrophils employ a bleb-based mechanism of force generation to deform a soft barrier exerting cell-scale confining pressures. In all, this work introduces a tunable in vitro material interface that replicates confining pressures applied by soft tissue environments.
Collapse
Affiliation(s)
- Jonathan H Schrope
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Medical Scientist Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Adam Horn
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Kaitlyn Lazorchak
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Medical Scientist Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Clyde W Tinnen
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Jack J Stevens
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Mehtab Farooqui
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Tanner Robertson
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Jiayi Li
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - David Bennin
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Terry Juang
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Adeel Ahmed
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Chao Li
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| |
Collapse
|
3
|
Xu Q, Authi KS, Kirpotina LN, Schepetkin IA, Quinn MT, Cilibrizzi A. Development of small-molecule fluorescent probes targeting neutrophils via N-formyl peptide receptors. RSC Med Chem 2025; 16:1397-1409. [PMID: 39886349 PMCID: PMC11775818 DOI: 10.1039/d4md00849a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/19/2024] [Indexed: 02/01/2025] Open
Abstract
N-Formyl peptide receptors (FPRs) are membrane receptors that are abundantly expressed in innate immune cells, including neutrophils and platelets, demonstrating potential new targets for immune system regulation and the treatment of inflammatory conditions. We report here the development and bio-physical validation of new FPR imaging agents as effective tools to track FPR distribution, localisation and functions, ultimately helping to establish FPR exact roles and functions in pathological and physiological conditions. The new series of probes feature a small molecule-based FPR address system conjugated to suitable fluorophores, resulting in highly specific FPR agents, including a partial agonist endowed with high affinity (i.e. low/sub-nanomolar potency) on FPR-transfected cells and human neutrophils. Preliminary imaging studies via multiphoton microscopy demonstrate that the probes enable the visualisation of FPRs in live cells, thus representing valid bio-imaging tools for the analysis of FPR-mediated signalling, such as the activation of neutrophils in inflammatory events.
Collapse
Affiliation(s)
- Qi Xu
- Institute of Pharmaceutical Science, King's College London Stamford Street London SE1 9NH UK +44 (0) 20 7848 9532
| | - Kalwant S Authi
- BHF Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, King's College London London SE1 9NH UK
| | - Liliya N Kirpotina
- Department of Microbiology and Cell Biology, Montana State University Bozeman Montana 59717 USA
| | - Igor A Schepetkin
- Department of Microbiology and Cell Biology, Montana State University Bozeman Montana 59717 USA
| | - Mark T Quinn
- Department of Microbiology and Cell Biology, Montana State University Bozeman Montana 59717 USA
| | - Agostino Cilibrizzi
- Institute of Pharmaceutical Science, King's College London Stamford Street London SE1 9NH UK +44 (0) 20 7848 9532
| |
Collapse
|
4
|
Bose S, Do V, Testini C, Jadhav SS, Sailliet N, Kho AT, Komatsu M, Boneschansker L, Kong SW, Wedel J, Briscoe DM. Immunomodulation by allograft endothelial cells. FRONTIERS IN TRANSPLANTATION 2025; 4:1518772. [PMID: 39967861 PMCID: PMC11832486 DOI: 10.3389/frtra.2025.1518772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025]
Abstract
It is increasingly appreciated that the expression of immunoregulatory molecules within tumors have potential to shape a microenvironment that promotes local immunoevasion and immunoregulation. However, little is known about tissue-intrinsic immunomodulatory mechanisms following transplantation. We propose that differences in the phenotype of microvascular endothelial cells impact the alloantigenicity of the graft and its potential to promote immunoregulation following transplantation. We focus this review on the concept that graft-dependent immunoregulation may evolve post-transplantation, and that it is dependent on the phenotype of select subsets of intragraft endothelial cells. We also discuss evidence that long-term graft survival is critically dependent on adaptive interactions among immune cells and endothelial cells within the transplanted tissue microenvironment.
Collapse
Affiliation(s)
- Sayantan Bose
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Vicki Do
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States
| | - Chiara Testini
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Suchita S. Jadhav
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Nicolas Sailliet
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Alvin T. Kho
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA, United States
| | - Masaki Komatsu
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Leo Boneschansker
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Sek Won Kong
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA, United States
| | - Johannes Wedel
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA, United States
| | - David M. Briscoe
- Transplant Research Program, Boston Children’s Hospital, Boston, MA, United States
- Division of Nephrology, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
- The Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
5
|
Kwaku GN, Jensen KN, Simaku P, Floyd DJ, Saelens JW, Reardon CM, Ward RA, Basham KJ, Hepworth OW, Vyas TD, Zamith-Miranda D, Nosanchuk JD, Vyas JM, Harding HB. Extracellular vesicles from diverse fungal pathogens induce species-specific and endocytosis-dependent immunomodulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.03.631181. [PMID: 39803513 PMCID: PMC11722428 DOI: 10.1101/2025.01.03.631181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
Microbial pathogens generate extracellular vesicles (EVs) for intercellular communication and quorum sensing. Microbial EVs also induce inflammatory pathways within host innate immune cells. We previously demonstrated that EVs secreted by Candida albicans trigger type I interferon signaling in host cells specifically via the cGAS-STING innate immune signaling pathway. Here, we show that despite sharing similar properties of morphology and internal DNA content, the interactions between EVs and the innate immune system differ according to the parental fungal species. EVs secreted by C. albicans, Saccharomyces cerevisiae, Cryptococcus neoformans, and Aspergillus fumigatus are endocytosed at different rates by murine macrophages triggering varied cytokine responses, innate immune signaling, and subsequent immune cell recruitment. Notably, cell wall constituents that decorate C. neoformans and A. fumigatus EVs inhibit efficient internalization by macrophages and dampen innate immune activation. Our data uncover the transcriptional and functional consequences of the internalization of diverse fungal EVs by immune cells and reveal novel insights into the early innate immune response to distinct clinically significant fungal pathogens.
Collapse
Affiliation(s)
- Geneva N Kwaku
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kirstine Nolling Jensen
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Patricia Simaku
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Daniel J Floyd
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Joseph W Saelens
- Pfizer Worldwide Research Development and Medical, Machine Learning and Computational Sciences, Cambridge, MA, USA
| | - Christopher M Reardon
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Rebecca A Ward
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kyle J Basham
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Olivia W Hepworth
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Tammy D Vyas
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Daniel Zamith-Miranda
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Joshua D Nosanchuk
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jatin M Vyas
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Infectious Disease, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Hannah Brown Harding
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
6
|
Grandhi TSP, Mebrahtu M, Musso R, Fullman A, Nifong B, Wisdom K, Roh TT, Sender M, Poore D, Macdougall CE, Oren R, Griffin S, Cheng AT, Ekert JE. A microphysiological assay for studying T-cell chemotaxis, trafficking and tumor killing. Biofabrication 2024; 17:015004. [PMID: 39378897 DOI: 10.1088/1758-5090/ad847f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 10/08/2024] [Indexed: 10/10/2024]
Abstract
Tumors in patients non-responsive to immunotherapy harbor a series of barriers that impede the efficacy of effector T-cells. Consequently, therapeutically modulating the chemotaxis machinery to enable effector T cell infiltration and function in the tumor could result in more successful therapeutic outcomes. Complexin-vitromodels allow re-creation ofin-vivotumor complexities in anin-vitrosetting, allowing improved translatability to patient biology at the laboratory scale. We identified a gap in available industrial scale microphysiological (MPS) assays for faster validation of targets and strategies that enable T-cell chemotaxis and effector function within tumor microenvironments. Using a commercially available, 96-chip 2-lane microfluidic assay system, we present a novel, scalable, complexin vitroMPS assay to study 3D T-cell chemotaxis and function within native, extracellular matrix (ECM)-rich multicellular tumor environments. Activated or naïve CD3+ T-cells stained with far-red nuclear stain responded to the chemokine gradients generated within the matrigel-collagen ECM by migrating into the microfluidic channel (∼5 mm horizontal window), in a concentration- and cell type-dependent manner. Furthermore, we observed and tracked chemotaxis and cancer cell killing function of antigen-specific CD4.CD8. chimeric antigen receptor (CAR)-T cells that responded to CXCR3 agonist gradient built through the expansive 5 mm of cancer cell colony containing stroma. The 2-lane assay system yielded useful information regarding donor and dose-dependent differences in CAR-T cell chemotaxis and tumor killing. The scalable assay system allows a granular window into immune cell migration and function in tissue spaces beyond endothelium, addressing a missing gap in studying tissue-specific immune cell chemotaxis and function to bring forward advancements in cancer immunotherapy.
Collapse
Affiliation(s)
| | - Makda Mebrahtu
- Genome Biology, GSK, Collegeville, PA, United States of America
| | - Ryan Musso
- Genome Biology, GSK, Collegeville, PA, United States of America
| | - Alexis Fullman
- Genome Biology, GSK, Collegeville, PA, United States of America
| | - Brady Nifong
- Research Statistics, GSK, Collegeville, PA, United States of America
| | - Katrina Wisdom
- Complex In-Vitro Models, GSK, Collegeville, PA, United States of America
| | - Terrence T Roh
- Complex In-Vitro Models, GSK, Collegeville, PA, United States of America
| | - Matthew Sender
- Chemical Biology, GSK, Collegeville, PA, United States of America
| | - Derek Poore
- Immuno-Oncology and Combinations (IOC), GSK, Collegeville, PA, United States of America
| | | | - Ravit Oren
- Oncology Cell Therapy, GSK, Stevenage, United Kingdom
| | - Sue Griffin
- Oncology Translational Research, GSK, Stevenage, United Kingdom
| | - Aaron T Cheng
- Genome Biology, GSK, Collegeville, PA, United States of America
| | - Jason E Ekert
- Complex In-Vitro Models, GSK, Collegeville, PA, United States of America
| |
Collapse
|
7
|
Schrope JH, Horn A, Lazorchak K, Tinnen CW, Stevens JJ, Farooqui M, Li J, Bennin D, Robertson T, Juang TD, Ahmed A, Li C, Huttenlocher A, Beebe D. Confinement by liquid-liquid interface replicates in vivo neutrophil deformations and elicits bleb based migration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.14.544898. [PMID: 38106211 PMCID: PMC10723256 DOI: 10.1101/2023.06.14.544898] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Leukocytes navigate through interstitial spaces resulting in deformation of both the motile leukocytes and surrounding cells. Creating an in vitro system that models the deformable cellular environment encountered in vivo has been challenging. Here, we engineer microchannels with a liquid-liquid interface that exerts confining pressures (200-3000 Pa) similar to cells in tissues, and, thus, is deformable by cell generated forces. Consequently, the balance between migratory cell-generated and interfacial pressures determines the degree of confinement. Pioneer cells that first contact the interfacial barrier require greater deformation forces to forge a path for migration, and as a result migrate slower than trailing cells. Critically, resistive pressures are tunable by controlling the curvature of the liquid interface, which regulates motility. By granting cells autonomy in determining their confinement, and tuning environmental resistance, interfacial deformations are made to match those of surrounding cells in vivo during interstitial neutrophil migration in a larval zebrafish model. We discover that, in this context, neutrophils employ a bleb-based mechanism of force generation to deform a barrier exerting cell-scale confining pressures.
Collapse
|
8
|
Camacho-Gomez D, Movilla N, Borau C, Martin A, Oñate Salafranca C, Pardo J, Gomez-Benito MJ, Garcia-Aznar JM. An agent-based method to estimate 3D cell migration trajectories from 2D measurements: Quantifying and comparing T vs CAR-T 3D cell migration. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 255:108331. [PMID: 39068872 DOI: 10.1016/j.cmpb.2024.108331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/05/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND AND OBJECTIVE Immune cell migration is one of the key features that enable immune cells to find invading pathogens, control tissue damage, and eliminate primary developing tumors. Chimeric antigen receptor (CAR) T-cell therapy is a novel strategy in the battle against various cancers. It has been successful in treating hematological tumors, yet it still faces many challenges in the case of solid tumors. In this work, we evaluate the three-dimensional (3D) migration capacity of T and CAR-T cells within dense collagen-based hydrogels. Quantifying three-dimensional (3D) cell migration requires microscopy techniques that may not be readily accessible. Thus, we introduce a straightforward mathematical model designed to infer 3D trajectories of cells from two-dimensional (2D) cell trajectories. METHODS We develop a 3D agent-based model (ABM) that simulates the temporal changes in the direction of migration with an inverse transform sampling method. Then, we propose an optimization procedure to accurately orient cell migration over time to reproduce cell migration from 2D experimental cell trajectories. With this model, we simulate cell migration assays of T and CAR-T cells in microfluidic devices conducted under hydrogels with different concentrations of type I collagen and validate our 3D cell migration predictions with light-sheet microscopy. RESULTS Our findings indicate that CAR-T cell migration is more sensitive to collagen concentration increases than T cells, resulting in a more pronounced reduction in their invasiveness. Moreover, our computational model reveals significant differences in 3D movement patterns between T and CAR-T cells. T cells exhibit migratory behavior in 3D whereas that CAR-T cells predominantly move within the XY plane, with limited movement in the Z direction. However, upon the introduction of a CXCL12 chemical gradient, CAR-T cells present migration patterns that closely resemble those of T cells. CONCLUSIONS This framework demonstrates that 2D projections of 3D trajectories may not accurately represent real migration patterns. Moreover, it offers a tool to estimate 3D migration patterns from 2D experimental data, which can be easily obtained with automatic quantification algorithms. This approach helps reduce the need for sophisticated and expensive microscopy equipment required in laboratories, as well as the computational burden involved in producing and analyzing 3D experimental data.
Collapse
Affiliation(s)
- Daniel Camacho-Gomez
- Department of Mechanical Engineering, Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
| | - Nieves Movilla
- Department of Mechanical Engineering, Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
| | - Carlos Borau
- Department of Mechanical Engineering, Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain; Centro Universitario de la Defensa de Zaragoza, Zaragoza, 50090, Spain
| | - Alejandro Martin
- Department of Mechanical Engineering, Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
| | | | - Julian Pardo
- Faculty of Medicine, University of Zaragoza/IIS Aragon, Spain; CIBER of Infectious diseases, IS Carlos III, Madrid, Spain
| | - Maria Jose Gomez-Benito
- Department of Mechanical Engineering, Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
| | - Jose Manuel Garcia-Aznar
- Department of Mechanical Engineering, Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain.
| |
Collapse
|
9
|
Hall CK, Barr OM, Delamare A, Burkholder A, Tsai A, Tian Y, Felix E Ellett, Li BM, Tanzi RE, Jorfi M. Profiling migration of human monocytes in response to chemotactic and barotactic guidance cues. CELL REPORTS METHODS 2024; 4:100846. [PMID: 39241776 PMCID: PMC11440068 DOI: 10.1016/j.crmeth.2024.100846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/10/2024] [Accepted: 08/09/2024] [Indexed: 09/09/2024]
Abstract
Monocytes are critical to innate immunity, participating in chemotaxis during tissue injury, infection, and inflammatory conditions. However, the migration dynamics of human monocytes under different guidance cues are not well characterized. Here, we developed a microfluidic device to profile the migration characteristics of human monocytes under chemotactic and barotactic guidance cues while also assessing the effects of age and cytokine stimulation. Human monocytes preferentially migrated toward the CCL2 gradient through confined microchannels, regardless of donor age and migration pathway. Stimulation with interferon (IFN)-γ, but not granulocyte-macrophage colony-stimulating factor (GM-CSF), disrupted monocyte navigation through complex paths and decreased monocyte CCL2 chemotaxis, velocity, and CCR2 expression. Additionally, monocytes exhibited a bias toward low-hydraulic-resistance pathways in asymmetric environments, which remained consistent across donor ages, cytokine stimulation, and chemoattractants. This microfluidic system provides insights into the unique migratory behaviors of human monocytes and is a valuable tool for studying peripheral immune cell migration in health and disease.
Collapse
Affiliation(s)
- Clare K Hall
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Olivia M Barr
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Antoine Delamare
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Alex Burkholder
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Alice Tsai
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Yuyao Tian
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Felix E Ellett
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Charlestown, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Brent M Li
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Mehdi Jorfi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA; Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Charlestown, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Liu Y, Zhao W, Hodgson J, Egan M, Cooper Pope CN, Hicks G, Nikolinakos PG, Mao L. CTC-Race: Single-Cell Motility Assay of Circulating Tumor Cells from Metastatic Lung Cancer Patients. ACS NANO 2024; 18:8683-8693. [PMID: 38465942 PMCID: PMC10976960 DOI: 10.1021/acsnano.3c09450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/26/2024] [Accepted: 03/04/2024] [Indexed: 03/12/2024]
Abstract
Distinctive subpopulations of circulating tumor cells (CTCs) with increased motility are considered to possess enhanced tumor-initiating potential and contribute to metastasis. Single-cell analysis of the migratory CTCs may increase our understanding of the metastatic process, yet most studies are limited by technical challenges associated with the isolation and characterization of these cells due to their extreme scarcity and heterogeneity. We report a microfluidic method based on CTCs' chemotactic motility, termed as CTC-Race assay, that can analyze migrating CTCs from metastatic non-small-cell lung cancer (NSCLC) patients with advanced tumor stages and enable concurrent biophysical and biochemical characterization of them with single-cell resolution. Analyses of motile CTCs in the CTC-Race assay, in synergy with other single cell characterization techniques, could provide insights into cancer metastasis.
Collapse
Affiliation(s)
- Yang Liu
- School
of Chemical, Materials and Biomedical Engineering, College of Engineering, The University of Georgia, Athens, Georgia 30602, United States
| | - Wujun Zhao
- FCS
Technology, LLC, Athens, Georgia 30602, United States
| | - Jamie Hodgson
- University
Cancer and Blood Center, LLC, Athens, Georgia 30607, United States
| | - Mary Egan
- University
Cancer and Blood Center, LLC, Athens, Georgia 30607, United States
| | | | - Glenda Hicks
- University
Cancer and Blood Center, LLC, Athens, Georgia 30607, United States
| | | | - Leidong Mao
- School
of Electrical and Computer Engineering, College of Engineering, The University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
11
|
Ramadan Q, Hazaymeh R, Zourob M. Immunity-on-a-Chip: Integration of Immune Components into the Scheme of Organ-on-a-Chip Systems. Adv Biol (Weinh) 2023; 7:e2200312. [PMID: 36866511 DOI: 10.1002/adbi.202200312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/16/2023] [Indexed: 03/04/2023]
Abstract
Studying the immune system in vitro aims to understand how, when, and where the immune cells migrate/differentiate and respond to the various triggering events and the decision points along the immune response journey. It becomes evident that organ-on-a-chip (OOC) technology has a superior capability to recapitulate the cell-cell and tissue-tissue interaction in the body, with a great potential to provide tools for tracking the paracrine signaling with high spatial-temporal precision and implementing in situ real-time, non-destructive detection assays, therefore, enabling extraction of mechanistic information rather than phenotypic information. However, despite the rapid development in this technology, integration of the immune system into OOC devices stays among the least navigated tasks, with immune cells still the major missing components in the developed models. This is mainly due to the complexity of the immune system and the reductionist methodology of the OOC modules. Dedicated research in this field is demanded to establish the understanding of mechanism-based disease endotypes rather than phenotypes. Herein, we systemically present a synthesis of the state-of-the-art of immune-cantered OOC technology. We comprehensively outlined what is achieved and identified the technology gaps emphasizing the missing components required to establish immune-competent OOCs and bridge these gaps.
Collapse
Affiliation(s)
- Qasem Ramadan
- Alfaisal University, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Rana Hazaymeh
- Almaarefa University, Diriyah, 13713, Kingdom of Saudi Arabia
| | | |
Collapse
|
12
|
Kim S, Lee SK, Son A, Lee J, Kim HG. A Comparative Inflammation-on-a-Chip with a Complete 3D Interface: Pharmacological Applications in COPD-Induced Neutrophil Migration. Adv Healthc Mater 2023; 12:e2301673. [PMID: 37505448 PMCID: PMC11469264 DOI: 10.1002/adhm.202301673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/06/2023] [Indexed: 07/29/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a slow-progressing inflammatory lung disease that is associated with high mortality and disability. There is a lack of appropriate preclinical models of COPD, which hampers drug discovery efforts. Herein, a comparative inflammation-on-a-chip (IoC) is developed with a complete 3D interface without the formation of any micropillar and phaseguide structures that replicated chemoattractant-induced neutrophil transendothelial migration (NTEM), a key feature of COPD. The IoC model is used to evaluate the pharmacological effects of CXCR2 inhibitors (MK-7123, AZD5069, and SB225002) on the migration of neutrophil-like cells in the presence of plasma samples from patients with COPD. This is the first study to evaluate inhibitors of CXCR2-dependent NTEM in a comparative IoC model that mimics the physiological 3D microenvironment, consisting of an endothelial barrier, extracellular compartment, and inflammatory conditions. This IoC model will be useful to investigate COPD severity using patient samples, and will aid basic and translational research involving NTEM.
Collapse
Affiliation(s)
- Soohyun Kim
- Center for Infectious Disease Vaccine and Diagnosis InnovationKorea Research Institute of Chemical TechnologyDaejeon34114Republic of Korea
| | - Sung Kyun Lee
- Center for Infectious Disease Vaccine and Diagnosis InnovationKorea Research Institute of Chemical TechnologyDaejeon34114Republic of Korea
| | - Ahryeong Son
- Center for Infectious Disease Vaccine and Diagnosis InnovationKorea Research Institute of Chemical TechnologyDaejeon34114Republic of Korea
| | - Jong‐Hwan Lee
- Center for Infectious Disease Vaccine and Diagnosis InnovationKorea Research Institute of Chemical TechnologyDaejeon34114Republic of Korea
| | - Hong Gi Kim
- Center for Infectious Disease Vaccine and Diagnosis InnovationKorea Research Institute of Chemical TechnologyDaejeon34114Republic of Korea
| |
Collapse
|
13
|
Fay ME, Oshinowo O, Iffrig E, Fibben KS, Caruso C, Hansen S, Musick JO, Valdez JM, Azer SS, Mannino RG, Choi H, Zhang DY, Williams EK, Evans EN, Kanne CK, Kemp ML, Sheehan VA, Carden MA, Bennett CM, Wood DK, Lam WA. iCLOTS: open-source, artificial intelligence-enabled software for analyses of blood cells in microfluidic and microscopy-based assays. Nat Commun 2023; 14:5022. [PMID: 37596311 PMCID: PMC10439163 DOI: 10.1038/s41467-023-40522-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 07/28/2023] [Indexed: 08/20/2023] Open
Abstract
While microscopy-based cellular assays, including microfluidics, have significantly advanced over the last several decades, there has not been concurrent development of widely-accessible techniques to analyze time-dependent microscopy data incorporating phenomena such as fluid flow and dynamic cell adhesion. As such, experimentalists typically rely on error-prone and time-consuming manual analysis, resulting in lost resolution and missed opportunities for innovative metrics. We present a user-adaptable toolkit packaged into the open-source, standalone Interactive Cellular assay Labeled Observation and Tracking Software (iCLOTS). We benchmark cell adhesion, single-cell tracking, velocity profile, and multiscale microfluidic-centric applications with blood samples, the prototypical biofluid specimen. Moreover, machine learning algorithms characterize previously imperceptible data groupings from numerical outputs. Free to download/use, iCLOTS addresses a need for a field stymied by a lack of analytical tools for innovative, physiologically-relevant assays of any design, democratizing use of well-validated algorithms for all end-user biomedical researchers who would benefit from advanced computational methods.
Collapse
Affiliation(s)
- Meredith E Fay
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Oluwamayokun Oshinowo
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Elizabeth Iffrig
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Kirby S Fibben
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Christina Caruso
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Scott Hansen
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Jamie O Musick
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - José M Valdez
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Sally S Azer
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Robert G Mannino
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hyoann Choi
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Dan Y Zhang
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- The George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Evelyn K Williams
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Erica N Evans
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Celeste K Kanne
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Melissa L Kemp
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Vivien A Sheehan
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Marcus A Carden
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Carolyn M Bennett
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - David K Wood
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Wilbur A Lam
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
14
|
Natalia A, Zhang L, Sundah NR, Zhang Y, Shao H. Analytical device miniaturization for the detection of circulating biomarkers. NATURE REVIEWS BIOENGINEERING 2023; 1:1-18. [PMID: 37359772 PMCID: PMC10064972 DOI: 10.1038/s44222-023-00050-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 06/28/2023]
Abstract
Diverse (sub)cellular materials are secreted by cells into the systemic circulation at different stages of disease progression. These circulating biomarkers include whole cells, such as circulating tumour cells, subcellular extracellular vesicles and cell-free factors such as DNA, RNA and proteins. The biophysical and biomolecular state of circulating biomarkers carry a rich repertoire of molecular information that can be captured in the form of liquid biopsies for disease detection and monitoring. In this Review, we discuss miniaturized platforms that allow the minimally invasive and rapid detection and analysis of circulating biomarkers, accounting for their differences in size, concentration and molecular composition. We examine differently scaled materials and devices that can enrich, measure and analyse specific circulating biomarkers, outlining their distinct detection challenges. Finally, we highlight emerging opportunities in biomarker and device integration and provide key future milestones for their clinical translation.
Collapse
Affiliation(s)
- Auginia Natalia
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Li Zhang
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Noah R. Sundah
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
| | - Yan Zhang
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
| | - Huilin Shao
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
15
|
Shao N, Zhou Y, Yao J, Zhang P, Song Y, Zhang K, Han X, Wang B, Liu X. A Bidirectional Single-Cell Migration and Retrieval Chip for Quantitative Study of Dendritic Cell Migration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204544. [PMID: 36658690 PMCID: PMC10015900 DOI: 10.1002/advs.202204544] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 12/22/2022] [Indexed: 06/17/2023]
Abstract
Dendritic cell (DC) migration is a fundamental step during execution of its adaptive immunity functions. Studying DC migration characteristics is critical for development of DC-dependent allergy treatments, vaccines, and cancer immunotherapies. Here, a microfluidics-based single-cell migration platform is described that enables high-throughput and precise bidirectional cell migration assays. It also allows selective retrieval of cell subpopulations that have different migratory potentials. Using this microfluidic platform, DC migration is investigated in response to different chemoattractants and inhibitors, quantitatively describe DC migration patterns and retrieve DC subpopulations of different migratory potentials for differential gene expression analysis. This platform opens an avenue for precise characterization of cell migration and potential discovery of therapeutic modulators.
Collapse
Affiliation(s)
- Ning Shao
- Department of NanomedicineHouston Methodist Research InstituteHoustonTX77030USA
| | - Yufu Zhou
- Department of NanomedicineHouston Methodist Research InstituteHoustonTX77030USA
- The Third Xiangya HospitalCentral South UniversityChangsha410008P. R. China
| | - Jun Yao
- Department of Molecular and Cellular OncologyThe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Pengchao Zhang
- Department of NanomedicineHouston Methodist Research InstituteHoustonTX77030USA
- Present address:
Key Laboratory of Advanced Technology for Materials Synthesis and ProcessingSchool of Materials Science and EngineeringWuhan University of TechnologyWuhan430070P. R. China
| | - Yanni Song
- Department of NanomedicineHouston Methodist Research InstituteHoustonTX77030USA
- Department of Breast SurgeryHarbin Medical University Cancer HospitalHarbin150081P. R. China
| | - Kai Zhang
- Department of NanomedicineHouston Methodist Research InstituteHoustonTX77030USA
| | - Xin Han
- Department of NanomedicineHouston Methodist Research InstituteHoustonTX77030USA
- Present address:
School of Medicine and Holistic Integrative MedicineNanjing University of Chinese MedicineNanjing210023P. R. China
| | - Bin Wang
- Department of GeneticsThe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Xuewu Liu
- Department of NanomedicineHouston Methodist Research InstituteHoustonTX77030USA
| |
Collapse
|
16
|
Rodríguez-Fernández JL, Criado-García O. A meta-analysis indicates that the regulation of cell motility is a non-intrinsic function of chemoattractant receptors that is governed independently of directional sensing. Front Immunol 2022; 13:1001086. [PMID: 36341452 PMCID: PMC9630654 DOI: 10.3389/fimmu.2022.1001086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/03/2022] [Indexed: 11/30/2022] Open
Abstract
Chemoattraction, defined as the migration of a cell toward a source of a chemical gradient, is controlled by chemoattractant receptors. Chemoattraction involves two basic activities, namely, directional sensing, a molecular mechanism that detects the direction of a source of chemoattractant, and actin-based motility, which allows the migration of a cell towards it. Current models assume first, that chemoattractant receptors govern both directional sensing and motility (most commonly inducing an increase in the migratory speed of the cells, i.e. chemokinesis), and, second, that the signaling pathways controlling both activities are intertwined. We performed a meta-analysis to reassess these two points. From this study emerge two main findings. First, although many chemoattractant receptors govern directional sensing, there are also receptors that do not regulate cell motility, suggesting that is the ability to control directional sensing, not motility, that best defines a chemoattractant receptor. Second, multiple experimental data suggest that receptor-controlled directional sensing and motility can be controlled independently. We hypothesize that this independence may be based on the existence of separated signalling modules that selectively govern directional sensing and motility in chemotactic cells. Together, the information gathered can be useful to update current models representing the signalling from chemoattractant receptors. The new models may facilitate the development of strategies for a more effective pharmacological modulation of chemoattractant receptor-controlled chemoattraction in health and disease.
Collapse
|
17
|
Chen C, Li P, Guo T, Chen S, Xu D, Chen H. Generation of Dynamic Concentration Profile Using A Microfluidic Device Integrating Pneumatic Microvalves. BIOSENSORS 2022; 12:bios12100868. [PMID: 36291005 PMCID: PMC9599525 DOI: 10.3390/bios12100868] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/08/2022] [Accepted: 10/09/2022] [Indexed: 06/12/2023]
Abstract
Generating and maintaining the concentration dilutions of diffusible molecules in microchannels is critical for high-throughput chemical and biological analysis. Conventional serial network microfluidic technologies can generate high orders of arbitrary concentrations by a predefined microchannel network. However, a previous design requires a large occupancy area and is unable to dynamically generate different profiles in the same chip, limiting its applications. This study developed a microfluidic device enabling dynamic variations of both the concentration in the same channel and the concentration distribution in multiple channels by adjusting the flow resistance using programmable pneumatic microvalves. The key component (the pneumatic microvalve) allowed dynamic adjustment of the concentration profile but occupied a tiny space. Additionally, a Matlab program was developed to calculate the flow rates and flow resistance of various sections of the device, which provided theoretical guidance for dimension design. In silico investigations were conducted to evaluate the microvalve deformation with widths from 100 to 300 µm and membrane thicknesses of 20 and 30 µm under the activation pressures between 0 and 2000 mbar. The flow resistance of the deformed valve was studied both numerically and experimentally and an empirical model for valve flow resistance with the form of Rh=aebP was proposed. Afterward, the fluid flow in the valve region was characterized using Micro PIV to further demonstrate the adjustment mechanism of the flow resistance. Then, the herringbone structures were employed for fast mixing to allow both quick variation of concentration and minor space usage of the channel network. Finally, an empirical formula-supported computational program was developed to provide the activation pressures required for the specific concentration profile. Both linear (Ck = -0.2k + 1) and nonlinear (Ck = (110)k) concentration distribution in four channels were varied using the same device by adjusting microvalves. The device demonstrated the capability to control the concentration profile dynamically in a small space, offering superior application potentials in analytical chemistry, drug screening, and cell biology research.
Collapse
Affiliation(s)
- Chang Chen
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen 518055, China
| | - Panpan Li
- School of Science, Harbin Institute of Technology, Shenzhen 518055, China
| | - Tianruo Guo
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Siyuan Chen
- School of Science, Harbin Institute of Technology, Shenzhen 518055, China
| | - Dong Xu
- School of Science, Harbin Institute of Technology, Shenzhen 518055, China
| | - Huaying Chen
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen 518055, China
| |
Collapse
|
18
|
Ashley BK, Sui J, Javanmard M, Hassan U. Antibody-functionalized aluminum oxide-coated particles targeting neutrophil receptors in a multifrequency microfluidic impedance cytometer. LAB ON A CHIP 2022; 22:3055-3066. [PMID: 35851596 PMCID: PMC9378602 DOI: 10.1039/d2lc00563h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Personalized diagnostics of infectious diseases require monitoring disease progression due to their ever-changing physiological conditions and the multi-faceted organ system mechanisms involved in disease pathogenesis. In such instances, the recommended clinical strategies involve multiplexing data collection from critical biomarkers related to a patient's conditions along with longitudinal frequent patient monitoring. Numerous detection technologies exist both in research and commercial settings to monitor these conditions, however, they fail to provide biomarker multiplexing ability with design and data processing simplicity. For a recently conceived multiplexing biomarker modality, this work demonstrates the use of electrically sensitive microparticles targeting and identifying membrane receptors on leukocytes using a single detection source, with a high potential for multiplexing greater than any existing impedance-based single-detection scheme. Here, polystyrene microparticles are coated with varying thicknesses of metal oxides, which generate quantifiable impedance shifts when exposed to multifrequency electric fields depending on the metal oxide thickness. Using multifrequency impedance cytometry, these particles can be measured and differentiated rapidly across one coplanar electrode scheme. After surface-functionalizing particles with antibodies targeting CD11b and CD66b receptors, the particles are combined with isolated neutrophils to measure receptor expression. A combination of data analysis techniques including multivariate analysis, supervised machine learning, and unsupervised machine learning was able to accurately differentiate samples with up to 91% accuracy. This proof-of-concept study demonstrates the potential for these oxide-coated particles for enumerating specific leukocytes enabling multiplexing. Further, additional coating thicknesses or different metal oxide materials can enable a compendium of multiplexing targeting resource to be used to develop a high-multiplexing sensor for targeting membrane receptor expression.
Collapse
Affiliation(s)
- Brandon K Ashley
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA.
| | - Jianye Sui
- Department of Electrical and Computer Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Mehdi Javanmard
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA.
- Department of Electrical and Computer Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Umer Hassan
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA.
- Department of Electrical and Computer Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
- Global Health Institute, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| |
Collapse
|
19
|
Sala F, Ficorella C, Osellame R, Käs JA, Martínez Vázquez R. Microfluidic Lab-on-a-Chip for Studies of Cell Migration under Spatial Confinement. BIOSENSORS 2022; 12:bios12080604. [PMID: 36004998 PMCID: PMC9405557 DOI: 10.3390/bios12080604] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/27/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022]
Abstract
Understanding cell migration is a key step in unraveling many physiological phenomena and predicting several pathologies, such as cancer metastasis. In particular, confinement has been proven to be a key factor in the cellular migration strategy choice. As our insight in the field improves, new tools are needed in order to empower biologists’ analysis capabilities. In this framework, microfluidic devices have been used to engineer the mechanical and spatial stimuli and to investigate cellular migration response in a more controlled way. In this work, we will review the existing technologies employed in the realization of microfluidic cellular migration assays, namely the soft lithography of PDMS and hydrogels and femtosecond laser micromachining. We will give an overview of the state of the art of these devices, focusing on the different geometrical configurations that have been exploited to study specific aspects of cellular migration. Our scope is to highlight the advantages and possibilities given by each approach and to envisage the future developments in in vitro migration studies under spatial confinement in microfluidic devices.
Collapse
Affiliation(s)
- Federico Sala
- Institute for Photonics and Nanotechnologies, CNR, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| | - Carlotta Ficorella
- Peter Debye Institute for Soft Matter Physics, University of Leipzig, 04109 Leipzig, Germany
| | - Roberto Osellame
- Institute for Photonics and Nanotechnologies, CNR, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| | - Josef A. Käs
- Peter Debye Institute for Soft Matter Physics, University of Leipzig, 04109 Leipzig, Germany
| | - Rebeca Martínez Vázquez
- Institute for Photonics and Nanotechnologies, CNR, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
- Correspondence:
| |
Collapse
|
20
|
Heydarian M, Rühl E, Rawal R, Kozjak-Pavlovic V. Tissue Models for Neisseria gonorrhoeae Research—From 2D to 3D. Front Cell Infect Microbiol 2022; 12:840122. [PMID: 35223556 PMCID: PMC8873371 DOI: 10.3389/fcimb.2022.840122] [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: 12/20/2021] [Accepted: 01/24/2022] [Indexed: 12/02/2022] Open
Abstract
Neisseria gonorrhoeae is a human-specific pathogen that causes gonorrhea, the second most common sexually transmitted infection worldwide. Disease progression, drug discovery, and basic host-pathogen interactions are studied using different approaches, which rely on models ranging from 2D cell culture to complex 3D tissues and animals. In this review, we discuss the models used in N. gonorrhoeae research. We address both in vivo (animal) and in vitro cell culture models, discussing the pros and cons of each and outlining the recent advancements in the field of three-dimensional tissue models. From simple 2D monoculture to complex advanced 3D tissue models, we provide an overview of the relevant methodology and its application. Finally, we discuss future directions in the exciting field of 3D tissue models and how they can be applied for studying the interaction of N. gonorrhoeae with host cells under conditions closely resembling those found at the native sites of infection.
Collapse
|
21
|
Paduthol G, Korma TS, Agrawal A, Paul D. Dynamic generation of power function gradient profiles in a universal microfluidic gradient generator by controlling the inlet flow rates. LAB ON A CHIP 2022; 22:592-604. [PMID: 34985077 DOI: 10.1039/d1lc00938a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
We report a two-inlet universal microfluidic gradient generator capable of generating gradient profiles of the functional form xp in the same device by controlling only the inlet flow rates. We have developed an analytical model to predict the inlet flow rates needed to generate a user-specified gradient profile at the outlet. We have validated this model by performing both COMSOL simulations and experiments. Our experiments show an excellent match between the target functions (x0.33, x1, x2 and x3) and the gradient profiles generated in this device. Unlike the universal gradient generators reported earlier, our device does not require changing the positions of the internal barriers for each new gradient profile, thereby making it easier for the user to operate this device.
Collapse
Affiliation(s)
- Gauri Paduthol
- Department of Bioscience and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
- Department of Mechanical Engineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Teji Shenne Korma
- Department of Bioscience and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| | - Amit Agrawal
- Department of Mechanical Engineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Debjani Paul
- Department of Bioscience and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| |
Collapse
|
22
|
Choi Y, Sunkara V, Lee Y, Cho YK. Exhausted mature dendritic cells exhibit a slower and less persistent random motility but retain chemotaxis against CCL19. LAB ON A CHIP 2022; 22:377-386. [PMID: 34927189 DOI: 10.1039/d1lc00876e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Dendritic cells (DCs), which are immune sentinels in the peripheral tissues, play a number of roles, including patrolling for pathogens, internalising antigens, transporting antigens to the lymph nodes (LNs), interacting with T cells, and secreting cytokines. The well-coordinated migration of DCs under various immunological or inflammatory conditions is therefore essential to ensure an effective immune response. Upon maturation, DCs migrate faster and more persistently than immature DCs (iDCs), which is believed to facilitate CCR7-dependent chemotaxis. It has been reported that lipopolysaccharide-activated DCs produce IL-12 only transiently, and become resistant to further stimulation through exhaustion. However, little is known about the influence of DC exhaustion on cellular motility. Here, we studied the cellular migration of exhausted DCs in tissue-mimicked confined environments. We found that the speed of exhausted matured DCs (xmDCs) decreased significantly compared to active matured DCs (amDCs) and iDCs. In contrast, the speed fluctuation increased compared to that of amDCs and was similar to that of iDCs. In addition, the diffusivity of the xmDCs was significantly lower than that of the amDCs, which implies that DC exhaustion reduces the space exploration ability. Interestingly, CCR7-dependent chemotaxis against CCL19 in xmDCs was not considerably different from that observed in amDCs. Taken together, we report a unique intrinsic cell migration behaviour of xmDCs, which exhibit a slower, less persistent, and less diffusive random motility, which results in the DCs remaining at the site of infection, although a well-preserved CCR7-dependent chemotactic motility is maintained.
Collapse
Affiliation(s)
- Yongjun Choi
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Vijaya Sunkara
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Yeojin Lee
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Yoon-Kyoung Cho
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| |
Collapse
|
23
|
Babatunde KA, Ayuso JM, Kerr SC, Huttenlocher A, Beebe DJ. Microfluidic Systems to Study Neutrophil Forward and Reverse Migration. Front Immunol 2021; 12:781535. [PMID: 34899746 PMCID: PMC8653704 DOI: 10.3389/fimmu.2021.781535] [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: 09/22/2021] [Accepted: 11/09/2021] [Indexed: 12/26/2022] Open
Abstract
During infection, neutrophils are the most abundantly recruited innate immune cells at sites of infection, playing critical roles in the elimination of local infection and healing of the injury. Neutrophils are considered to be short-lived effector cells that undergo cell death at infection sites and in damaged tissues. However, recent in vitro and in vivo evidence suggests that neutrophil behavior is more complex and that they can migrate away from the inflammatory site back into the vasculature following the resolution of inflammation. Microfluidic devices have contributed to an improved understanding of the interaction and behavior of neutrophils ex vivo in 2D and 3D microenvironments. The role of reverse migration and its contribution to the resolution of inflammation remains unclear. In this review, we will provide a summary of the current applications of microfluidic devices to investigate neutrophil behavior and interactions with other immune cells with a focus on forward and reverse migration in neutrophils.
Collapse
Affiliation(s)
| | - Jose M Ayuso
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, United States
| | - Sheena C Kerr
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, United States.,Carbone Cancer Center, University of Wisconsin, Madison, WI, United States
| | - Anna Huttenlocher
- Departments of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United States
| | - David J Beebe
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, United States.,Carbone Cancer Center, University of Wisconsin, Madison, WI, United States.,Department of Biomedical Engineering, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
24
|
Muldur S, Vadysirisack DD, Ragunathan S, Tang Y, Ricardo A, Sayegh CE, Irimia D. Human Neutrophils Respond to Complement Activation and Inhibition in Microfluidic Devices. Front Immunol 2021; 12:777932. [PMID: 34899737 PMCID: PMC8653703 DOI: 10.3389/fimmu.2021.777932] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 10/29/2021] [Indexed: 12/30/2022] Open
Abstract
Complement activation is key to anti-microbial defenses by directly acting on microbes and indirectly by triggering cellular immune responses. Complement activation may also contribute to the pathogenesis of numerous inflammatory and immunological diseases. Consequently, intense research focuses on developing therapeutics that block pathology-causing complement activation while preserving anti-microbial complement activities. However, the pace of research is slowed down significantly by the limitations of current tools for evaluating complement-targeting therapeutics. Moreover, the effects of potential therapeutic agents on innate immune cells, like neutrophils, are not fully understood. Here, we employ microfluidic assays and measure chemotaxis, phagocytosis, and swarming changes in human neutrophils ex vivo in response to various complement-targeting agents. We show that whereas complement factor 5 (C5) cleavage inhibitor eculizumab blocks all neutrophil anti-microbial functions, newer compounds like the C5 cleavage inhibitor RA101295 and C5a receptor antagonist avacopan inhibit chemotaxis and swarming while preserving neutrophil phagocytosis. These results highlight the utility of microfluidic neutrophil assays in evaluating potential complement-targeting therapeutics.
Collapse
Affiliation(s)
- Sinan Muldur
- BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Shriners Burns Hospital, Boston, MA, United States
| | | | | | - Yalan Tang
- Ra Pharmaceuticals, Inc., Cambridge, MA, United States
| | | | | | - Daniel Irimia
- BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Shriners Burns Hospital, Boston, MA, United States
| |
Collapse
|
25
|
Hadjitheodorou A, Bell GRR, Ellett F, Shastry S, Irimia D, Collins SR, Theriot JA. Directional reorientation of migrating neutrophils is limited by suppression of receptor input signaling at the cell rear through myosin II activity. Nat Commun 2021; 12:6619. [PMID: 34785640 PMCID: PMC8595366 DOI: 10.1038/s41467-021-26622-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/29/2021] [Indexed: 11/09/2022] Open
Abstract
To migrate efficiently to target locations, cells must integrate receptor inputs while maintaining polarity: a distinct front that leads and a rear that follows. Here we investigate what is necessary to overwrite pre-existing front-rear polarity in neutrophil-like HL60 cells migrating inside straight microfluidic channels. Using subcellular optogenetic receptor activation, we show that receptor inputs can reorient weakly polarized cells, but the rear of strongly polarized cells is refractory to new inputs. Transient stimulation reveals a multi-step repolarization process, confirming that cell rear sensitivity to receptor input is the primary determinant of large-scale directional reversal. We demonstrate that the RhoA/ROCK/myosin II pathway limits the ability of receptor inputs to signal to Cdc42 and reorient migrating neutrophils. We discover that by tuning the phosphorylation of myosin regulatory light chain we can modulate the activity and localization of myosin II and thus the amenability of the cell rear to 'listen' to receptor inputs and respond to directional reprogramming.
Collapse
Affiliation(s)
- Amalia Hadjitheodorou
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - George R R Bell
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, USA
| | - Felix Ellett
- Department of Surgery, BioMEMS Resource Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shashank Shastry
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, USA
| | - Daniel Irimia
- Department of Surgery, BioMEMS Resource Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sean R Collins
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA, USA.
| | - Julie A Theriot
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
26
|
Liu Y, Zhao W, Cheng R, Hodgson J, Egan M, Pope CNC, Nikolinakos PG, Mao L. Simultaneous biochemical and functional phenotyping of single circulating tumor cells using ultrahigh throughput and recovery microfluidic devices. LAB ON A CHIP 2021; 21:3583-3597. [PMID: 34346469 DOI: 10.1039/d1lc00454a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Profiling circulating tumour cells (CTCs) in cancer patients' blood samples is critical to understand the complex and dynamic nature of metastasis. This task is challenged by the fact that CTCs are not only extremely rare in circulation but also highly heterogeneous in their molecular programs and cellular functions. Here we report a combinational approach for the simultaneous biochemical and functional phenotyping of patient-derived CTCs, using an integrated inertial ferrohydrodynamic cell separation (i2FCS) method and a single-cell microfluidic migration assay. This combinatorial approach offers unique capability to profile CTCs on the basis of their surface expression and migratory characteristics. We achieve this using the i2FCS method that successfully processes whole blood samples in a tumor cell marker and size agnostic manner. The i2FCS method enables an ultrahigh blood sample processing throughput of up to 2 × 105 cells s-1 with a blood sample flow rate of 60 mL h-1. Its short processing time (10 minutes for a 10 mL sample), together with a close-to-complete CTC recovery (99.70% recovery rate) and a low WBC contamination (4.07-log depletion rate by removing 99.992% of leukocytes), results in adequate and functional CTCs for subsequent studies in the single-cell migration device. For the first time, we employ this new approach to query CTCs with single-cell resolution in accordance with their expression of phenotypic surface markers and migration properties, revealing the dynamic phenotypes and the existence of a high-motility subpopulation of CTCs in blood samples from metastatic lung cancer patients. This method could be adopted to study the biological and clinical value of invasive CTC phenotypes.
Collapse
Affiliation(s)
- Yang Liu
- Department of Chemistry, The University of Georgia, Athens, Georgia, USA
| | - Wujun Zhao
- FCS Technology, LLC, Athens, GA, 30606, USA
| | - Rui Cheng
- School of Electrical and Computer Engineering, College of Engineering, The University of Georgia, Athens, Georgia, USA.
| | - Jamie Hodgson
- University Cancer & Blood Center, LLC, Athens, GA, 30607, USA
| | - Mary Egan
- University Cancer & Blood Center, LLC, Athens, GA, 30607, USA
| | | | | | - Leidong Mao
- School of Electrical and Computer Engineering, College of Engineering, The University of Georgia, Athens, Georgia, USA.
| |
Collapse
|
27
|
Ashley BK, Hassan U. Point-of-critical-care diagnostics for sepsis enabled by multiplexed micro and nanosensing technologies. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1701. [PMID: 33650293 PMCID: PMC8447248 DOI: 10.1002/wnan.1701] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 12/14/2020] [Accepted: 01/08/2021] [Indexed: 11/12/2022]
Abstract
Sepsis is responsible for the highest economic and mortality burden in critical care settings around the world, prompting the World Health Organization in 2018 to designate it as a global health priority. Despite its high universal prevalence and mortality rate, a disproportionately low amount of sponsored research funding is directed toward diagnosis and treatment of sepsis, when early treatment has been shown to significantly improve survival. Additionally, current technologies and methods are inadequate to provide an accurate and timely diagnosis of septic patients in multiple clinical environments. For improved patient outcomes, a comprehensive immunological evaluation is critical which is comprised of both traditional testing and quantifying recently proposed biomarkers for sepsis. There is an urgent need to develop novel point-of-care, low-cost systems which can accurately stratify patients. These point-of-critical-care sensors should adopt a multiplexed approach utilizing multimodal sensing for heterogenous biomarker detection. For effective multiplexing, the sensors must satisfy criteria including rapid sample to result delivery, low sample volumes for clinical sample sparring, and reduced costs per test. A compendium of currently developed multiplexed micro and nano (M/N)-based diagnostic technologies for potential applications toward sepsis are presented. We have also explored the various biomarkers targeted for sepsis including immune cell morphology changes, circulating proteins, small molecules, and presence of infectious pathogens. An overview of different M/N detection mechanisms are also provided, along with recent advances in related nanotechnologies which have shown improved patient outcomes and perspectives on what future successful technologies may encompass. This article is categorized under: Diagnostic Tools > Biosensing.
Collapse
Affiliation(s)
- Brandon K. Ashley
- Department of Biomedical Engineering, Rutgers, State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Umer Hassan
- Department of Biomedical Engineering, Rutgers, State University of New Jersey, Piscataway, NJ, 08854, USA
- Department of Electrical Engineering, Rutgers, State University of New Jersey, Piscataway, NJ, 08854, USA
- Global Health Institute, Rutgers, State University of New Jersey. Piscataway, NJ, 08854, USA
| |
Collapse
|
28
|
Höving AL, Schmitz J, Schmidt KE, Greiner JFW, Knabbe C, Kaltschmidt B, Grünberger A, Kaltschmidt C. Human Blood Serum Induces p38-MAPK- and Hsp27-Dependent Migration Dynamics of Adult Human Cardiac Stem Cells: Single-Cell Analysis via a Microfluidic-Based Cultivation Platform. BIOLOGY 2021; 10:biology10080708. [PMID: 34439941 PMCID: PMC8389316 DOI: 10.3390/biology10080708] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/16/2021] [Accepted: 07/21/2021] [Indexed: 12/14/2022]
Abstract
Simple Summary Adult human stem cells possess the ability to contribute to endogenous regeneration processes of injured tissue by migrating to specific locations. For stem cell-based clinical applications it is highly important to gain knowledge about the migration behavior of adult human stem cells and the underlying molecular mechanisms of this ability. Human blood serum has been shown to have beneficial effects on other regenerative capacities of adult human stem cells. Within this study we tested the effect of human blood serum on the migration behavior of stem cells from the human heart. We used a microfluidic cultivation device, which allowed us to monitor the living cells and their movement behavior in real time. After addition of human blood serum, the heart stem cells increased their speed of movement and covered distance. Further, we observed that this effect could be diminished by inhibition of a specific kinase, p38-MAPK. Thus, our data suggest beneficial effects of human blood serum on adult human heart stem cells dependent on p38-MAPK. Our study contributes to a deeper understanding of the dynamics of stem cell migration and introduces a new platform to monitor stem cell movement in real time. Abstract Migratory capabilities of adult human stem cells are vital for assuring endogenous tissue regeneration and stem cell-based clinical applications. Although human blood serum has been shown to be beneficial for cell migration and proliferation, little is known about its impact on the migratory behavior of cardiac stem cells and underlying signaling pathways. Within this study, we investigated the effects of human blood serum on primary human cardiac stem cells (hCSCs) from the adult heart auricle. On a technical level, we took advantage of a microfluidic cultivation platform, which allowed us to characterize cell morphologies and track migration of single hCSCs via live cell imaging over a period of up to 48 h. Our findings showed a significantly increased migration distance and speed of hCSCs after treatment with human serum compared to control. Exposure of blood serum-stimulated hCSCs to the p38 mitogen-activated protein kinase (p38-MAPK) inhibitor SB239063 resulted in significantly decreased migration. Moreover, we revealed increased phosphorylation of heat shock protein 27 (Hsp27) upon serum treatment, which was diminished by p38-MAPK-inhibition. In summary, we demonstrate human blood serum as a strong inducer of adult human cardiac stem cell migration dependent on p38-MAPK/Hsp27-signalling. Our findings further emphasize the great potential of microfluidic cultivation devices for assessing spatio-temporal migration dynamics of adult human stem cells on a single-cell level.
Collapse
Affiliation(s)
- Anna L. Höving
- Department of Cell Biology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany; (K.E.S.); (J.F.W.G.); (B.K.); (C.K.)
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany;
- Correspondence:
| | - Julian Schmitz
- Multiscale Bioengineering, Faculty of Technology, Bielefeld University, 33615 Bielefeld, Germany; (J.S.); (A.G.)
| | - Kazuko E. Schmidt
- Department of Cell Biology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany; (K.E.S.); (J.F.W.G.); (B.K.); (C.K.)
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany;
| | - Johannes F. W. Greiner
- Department of Cell Biology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany; (K.E.S.); (J.F.W.G.); (B.K.); (C.K.)
| | - Cornelius Knabbe
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany;
| | - Barbara Kaltschmidt
- Department of Cell Biology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany; (K.E.S.); (J.F.W.G.); (B.K.); (C.K.)
- Molecular Neurobiology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany
| | - Alexander Grünberger
- Multiscale Bioengineering, Faculty of Technology, Bielefeld University, 33615 Bielefeld, Germany; (J.S.); (A.G.)
| | - Christian Kaltschmidt
- Department of Cell Biology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany; (K.E.S.); (J.F.W.G.); (B.K.); (C.K.)
| |
Collapse
|
29
|
Jyoti A, Kumar S, Kumar Srivastava V, Kaushik S, Govind Singh S. Neonatal sepsis at point of care. Clin Chim Acta 2021; 521:45-58. [PMID: 34153274 DOI: 10.1016/j.cca.2021.06.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/14/2021] [Accepted: 06/14/2021] [Indexed: 01/03/2023]
Abstract
Sepsis, which includes infection followed by inflammation, is one of the leading causes of death among neonates worldwide. The major attribute of this disease process is dysregulated host response to infection leading to organ dysfunction and potentially death. A comprehensive understanding of the host response as well as the pathogen itself are important factors contributing to outcome. Early diagnosis is paramount, as it leads to accurate assessment and improved clinical management. Accordingly, a number of diagnostic platforms have been introduced to assess the presence of blood stream pathogens in septic neonates. Unfortunately, current point-of-care (POC) methods rely on a single parameter/biomarker and thus lack a comprehensive evaluation. The emerging field of biosensing has, however, resulted in the development of a wide range of analytical devices that may be useful at POC. This review discusses currently available methods to screen the inflammatory process in neonatal sepsis. We describe POC sensor-based methods for single platform multi-analyte detection and highlight the latest advances in this evolving technology. Finally, we critically evaluate the applicability of these POC devices clinically for early diagnosis of sepsis in neonates.
Collapse
Affiliation(s)
- Anupam Jyoti
- Amity Institute of Biotechnology, Amity University, Jaipur, Rajasthan 303002, India; Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Bajhol, Solan, Himachal Pradesh, 173229, India.
| | - Sanni Kumar
- Department of Electrical Engineering, Indian Institute of Technology, Hyderabad, Telangana 502285, India.
| | | | - Sanket Kaushik
- Amity Institute of Biotechnology, Amity University, Jaipur, Rajasthan 303002, India.
| | - Shiv Govind Singh
- Department of Electrical Engineering, Indian Institute of Technology, Hyderabad, Telangana 502285, India.
| |
Collapse
|
30
|
Walters N, Zhang J, Rima XY, Nguyen LTH, Germain RN, Lämmermann T, Reátegui E. Analyzing Inter-Leukocyte Communication and Migration In Vitro: Neutrophils Play an Essential Role in Monocyte Activation During Swarming. Front Immunol 2021; 12:671546. [PMID: 34054848 PMCID: PMC8152805 DOI: 10.3389/fimmu.2021.671546] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/16/2021] [Indexed: 11/13/2022] Open
Abstract
Neutrophils are known to be the first responders to infection or injury. However, as inflammation progresses, other leukocytes become increasingly important in inflammation propagation, tissue reconstruction, and inflammation resolution. In recent years, there has been an increase in publications that analyze neutrophil behavior in vitro, but there remains a gap in the literature for in vitro technologies that enable quantitatively measuring interactions between different types of human leukocytes. Here, we used an in vitro platform that mimics inflammation by inducing neutrophil swarming to analyze the behavior of various leukocytes in a swarming setting. Using human peripheral blood leukocytes isolated directly from whole blood, we found that myeloid cells and lymphoid cells had different migratory behaviors. Myeloid cells, which are predominately neutrophils, exhibited swarming behavior. This behavior was not seen with lymphoid cells. We perturbed the peripheral blood leukocyte system by adding exogenous leukotriene B4 (LTB4) to the medium. Notably, only the myeloid cell compartment was significantly changed by the addition of LTB4. Additionally, LTB4 had no significant impact on myeloid cell migration during the recruitment phase of swarming. To further investigate the myeloid cell compartment, we isolated neutrophils and monocytes to analyze their interaction on the platform. We found that neutrophils increase monocyte migration toward the bioparticle clusters, as measured through speed, chemotactic index, track straightness, and swarm size. These results were confirmed with in vivo mouse experiments, where monocyte accumulation only occurred when neutrophils were present. Additionally, we found that both neutrophils and monocytes release the monocyte chemoattractant proteins CCL2 and CCL3 in the presence of Staphylococcus aureus bioparticles. Furthermore, extracellular vesicles from swarming neutrophils caused monocyte activation. These findings suggest that neutrophils play an essential role in the onset of inflammation not only by sealing off the site of infection or injury, but also by recruiting additional leukocytes to the site.
Collapse
Affiliation(s)
- Nicole Walters
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States
| | - Jingjing Zhang
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States
| | - Xilal Y Rima
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States
| | - Luong T H Nguyen
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States
| | - Ronald N Germain
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Tim Lämmermann
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.,Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Eduardo Reátegui
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States.,Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
31
|
Medapati MR, Singh N, Bhagirath AY, Duan K, Triggs-Raine B, Batista EL, Chelikani P. Bitter taste receptor T2R14 detects quorum sensing molecules from cariogenic Streptococcus mutans and mediates innate immune responses in gingival epithelial cells. FASEB J 2021; 35:e21375. [PMID: 33559200 DOI: 10.1096/fj.202000208r] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 12/18/2020] [Accepted: 01/04/2021] [Indexed: 12/26/2022]
Abstract
Host-pathogen interactions play an important role in defining the outcome of a disease. Recent studies have shown that the bacterial quorum sensing molecules (QSM) can interact with host cell membrane proteins, mainly G protein-coupled receptors (GPCRs), and induce innate immune responses. However, few studies have examined QSM-GPCR interactions and their influence on oral innate immune responses. In this study, we examined the role of bitter taste receptor T2R14 in sensing competence stimulating peptides (CSPs) secreted by cariogenic bacterium Streptococcus mutans and in mediating innate immune responses in gingival epithelial cells (GECs). Transcriptomic and western blot analyses identify T2R14 to be highly expressed in GECs. Our data show that only CSP-1 from S. mutans induces robust intracellular calcium mobilization compared to CSP-2 and CSP-3. By using CRISPR-Cas9, we demonstrate that CSP-1 induced calcium signaling and secretion of cytokines CXCL-8/IL-8, TNF-α, and IL-6 is mediated through T2R14 in GECs. Interestingly, the NF-kB signaling activated by CSP-1 in GECs was independent of T2R14. CSP-1-primed GECs attracted differentiated HL-60 immune cells (dHL-60) and this effect was abolished in T2R14 knock down GECs and also in cells primed with T2R14 antagonist 6-Methoxyflavone (6-MF). Our findings identify S. mutans CSP-1 as a peptide ligand for the T2R family. Our study establishes a novel host-pathogen interaction between cariogenic S. mutans CSP-1 and T2R14 in GECs leading to an innate immune response. Collectively, these findings suggest T2Rs as potential therapeutic targets to modulate innate immune responses upon oral bacterial infections.
Collapse
Affiliation(s)
- Manoj Reddy Medapati
- Manitoba Chemosensory Biology Research Group and Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Nisha Singh
- Manitoba Chemosensory Biology Research Group and Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Anjali Yadav Bhagirath
- Manitoba Chemosensory Biology Research Group and Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Children's Hospital Research Institute of Manitoba (CHRIM), University of Manitoba, Winnipeg, Canada
| | - Kangmin Duan
- Manitoba Chemosensory Biology Research Group and Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Children's Hospital Research Institute of Manitoba (CHRIM), University of Manitoba, Winnipeg, Canada
| | - Barbara Triggs-Raine
- Children's Hospital Research Institute of Manitoba (CHRIM), University of Manitoba, Winnipeg, Canada.,Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Eraldo L Batista
- Department of Dental Diagnostic and Clinical Sciences, Dr. Gerald Niznick College of Dentistry, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Prashen Chelikani
- Manitoba Chemosensory Biology Research Group and Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Children's Hospital Research Institute of Manitoba (CHRIM), University of Manitoba, Winnipeg, Canada
| |
Collapse
|
32
|
Kilb MF, Engemann VI, Siddique A, Stark RW, Schmitz K. Immobilisation of CXCL8 gradients in microfluidic devices for migration experiments. Colloids Surf B Biointerfaces 2020; 198:111498. [PMID: 33302150 DOI: 10.1016/j.colsurfb.2020.111498] [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: 10/05/2020] [Revised: 11/23/2020] [Accepted: 11/27/2020] [Indexed: 11/29/2022]
Abstract
The release of inflammatory chemokines leads to the formation of chemokine gradients that result in the directed migration of immune cells to the site of injury. In this process, cells respond to soluble gradients (chemotaxis) as well as to immobilised gradients (haptotaxis). Surface-bound chemokine gradients are mostly presented by endothelial cells and supported by glycosaminoglycans (GAGs), such as heparan sulfate, involving the GAG binding site of chemokines. Microfluidic devices have been used to analyse cell migration along soluble chemokine gradients, as these devices allow the generation of stable gradients with resolutions in the range of microns. To immobilise well-controlled soluble gradients of interleukin-8 (CXCL8), an inflammatory chemokine, we developed a simple procedure using a heparin-coated PDMS-microfluidic device. We used these immobilised gradients for migration experiments with CXCL8-responsive THP-1 cells and confirmed directed cell migration. This setup might be useful for the examination of factors that may alter chemotaxis and haptotaxis as well as synergistic and antagonistic effects of other soluble and immobilised chemokines.
Collapse
Affiliation(s)
- Michelle F Kilb
- Technical University of Darmstadt, Clemens-Schöpf-Institute of Organic Chemistry and Biochemistry, Alarich-Weiss-Straße 8, 64287, Darmstadt, Germany
| | - Victoria I Engemann
- Technical University of Darmstadt, Clemens-Schöpf-Institute of Organic Chemistry and Biochemistry, Alarich-Weiss-Straße 8, 64287, Darmstadt, Germany
| | - Asma Siddique
- Technical University of Darmstadt, Institute of Materials Science, Physics of Surfaces, Alarich-Weiss-Straße 16, 64287. Darmstadt, Germany
| | - Robert W Stark
- Technical University of Darmstadt, Institute of Materials Science, Physics of Surfaces, Alarich-Weiss-Straße 16, 64287. Darmstadt, Germany
| | - Katja Schmitz
- Technical University of Darmstadt, Clemens-Schöpf-Institute of Organic Chemistry and Biochemistry, Alarich-Weiss-Straße 8, 64287, Darmstadt, Germany.
| |
Collapse
|
33
|
Satti S, Deng P, Matthews K, Duffy SP, Ma H. Multiplexed end-point microfluidic chemotaxis assay using centrifugal alignment. LAB ON A CHIP 2020; 20:3096-3103. [PMID: 32748936 DOI: 10.1039/d0lc00311e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
A fundamental challenge to multiplexing microfluidic chemotaxis assays at scale is the requirement for time-lapse imaging to continuously track migrating cells. Drug testing and drug screening applications require the ability to perform hundreds of experiments in parallel, which is not feasible for assays that require continuous imaging. To address this limitation, end-point chemotaxis assays have been developed using fluid flow to align cells in traps or sieves prior to cell migration. However, these methods require precisely controlled fluid flow to transport cells to the correct location without undesirable mechanical stress, which introduce significant set up time and design complexity. Here, we describe a microfluidic device that eliminates the need for precise flow control by using centrifugation to align cells at a common starting point. A chemoattractant gradient is then formed using passive diffusion prior to chemotaxis in an incubated environment. This approach provides a simple and scalable approach to multiplexed chemotaxis assays. Centrifugal alignment is also insensitive to cell geometry, enabling this approach to be compatible with primary cell samples that are often heterogeneous. We demonstrate the capability of this approach by assessing chemotaxis of primary neutrophils in response to an fMLP (N-formyl-met-leu-phe) gradient. Our results show that cell alignment by centrifugation offers a potential avenue to develop scalable end-point multiplexed microfluidic chemotaxis assays.
Collapse
Affiliation(s)
- Sampath Satti
- School of Biomedical Engineering, University of British Columbia, Canada. and Centre for Blood Research, University of British Columbia, Canada
| | - Pan Deng
- Centre for Blood Research, University of British Columbia, Canada and Department of Mechanical Engineering, University of British Columbia, Canada
| | - Kerryn Matthews
- Centre for Blood Research, University of British Columbia, Canada and Department of Mechanical Engineering, University of British Columbia, Canada
| | - Simon P Duffy
- Centre for Blood Research, University of British Columbia, Canada and Department of Mechanical Engineering, University of British Columbia, Canada and British Columbia Institute of Technology, Canada
| | - Hongshen Ma
- School of Biomedical Engineering, University of British Columbia, Canada. and Centre for Blood Research, University of British Columbia, Canada and Department of Mechanical Engineering, University of British Columbia, Canada and Department of Urologic Sciences, University of British Columbia, Canada
| |
Collapse
|
34
|
Selenoprotein S attenuates endothelial dysfunction in a diabetic vascular chip. Exp Gerontol 2020; 137:110963. [DOI: 10.1016/j.exger.2020.110963] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 04/18/2020] [Accepted: 04/21/2020] [Indexed: 12/18/2022]
|
35
|
Grigolato F, Egholm C, Impellizzieri D, Arosio P, Boyman O. Establishment of a scalable microfluidic assay for characterization of population-based neutrophil chemotaxis. Allergy 2020; 75:1382-1393. [PMID: 31971608 DOI: 10.1111/all.14195] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/06/2019] [Accepted: 12/10/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Regulation of neutrophil chemotaxis and activation plays crucial roles in immunity, and dysregulated neutrophil responses can lead to pathology as seen in neutrophilic asthma. Neutrophil recruitment is key for initiating immune defense and inflammation, and its modulation is a promising therapeutic target. Microfluidic technology is an attractive tool for characterization of neutrophil migration. Compared to transwell assays, microfluidic approaches could offer several advantages, including precis e control of defined chemokine gradients in space and time, automated quantitative analysis of chemotaxis, and high throughput. METHODS We established a microfluidic device for fully automated, quantitative assessment of neutrophil chemotaxis. Freshly isolated mouse neutrophils from bone marrow or human neutrophils from peripheral blood were assessed in real time using an epifluorescence microscope for their migration toward the potent chemoattractants C-X-C-motif ligand 2 (CXCL2) and CXCL8, without or with interleukin-4 (IL-4) pre-incubation. RESULTS Our microfluidic device allowed the precise and reproducible determination of the optimal CXCL2 and CXCL8 concentrations for mouse and human neutrophil chemotaxis, respectively. Furthermore, our microfluidic assay was able to measure the equilibrium and real-time dynamic effects of specific modulators of neutrophil chemotaxis. We demonstrated this concept by showing that IL-4 receptor signaling in mouse and human neutrophils inhibited their migration toward CXCL2 and CXCL8, respectively, and this inhibition was time-dependent. CONCLUSION Collectively, our microfluidic device shows several advantages over traditional transwell migration assays and its design is amenable to future integration into multiplexed high-throughput platforms for screening of molecules that modulate the chemotaxis of different immune cells.
Collapse
Affiliation(s)
- Fulvio Grigolato
- Department of Chemistry and Applied Biosciences Swiss Federal Institute of Technology, Zurich Zurich Switzerland
| | - Cecilie Egholm
- Department of Immunology University Hospital Zurich Zurich Switzerland
| | | | - Paolo Arosio
- Department of Chemistry and Applied Biosciences Swiss Federal Institute of Technology, Zurich Zurich Switzerland
| | - Onur Boyman
- Department of Immunology University Hospital Zurich Zurich Switzerland
- Faculty of Medicine University of Zurich Zurich Switzerland
| |
Collapse
|
36
|
Garner RM, Skariah G, Hadjitheodorou A, Belliveau NM, Savinov A, Footer MJ, Theriot JA. Neutrophil-like HL-60 cells expressing only GFP-tagged β-actin exhibit nearly normal motility. Cytoskeleton (Hoboken) 2020; 77:181-196. [PMID: 32072765 PMCID: PMC7383899 DOI: 10.1002/cm.21603] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 12/21/2019] [Accepted: 01/27/2020] [Indexed: 12/30/2022]
Abstract
Observations of actin dynamics in living cells using fluorescence microscopy have been foundational in the exploration of the mechanisms underlying cell migration. We used CRISPR/Cas9 gene editing to generate neutrophil‐like HL‐60 cell lines expressing GFP‐β‐actin from the endogenous locus (ACTB). In light of many previous reports outlining functional deficiencies of labeled actin, we anticipated that HL‐60 cells would only tolerate a monoallelic edit, as biallelic edited cells would produce no normal β‐actin. Surprisingly, we recovered viable monoallelic GFP‐β‐actin cells as well as biallelic edited GFP‐β‐actin cells, in which one copy of the ACTB gene is silenced and the other contains the GFP tag. Furthermore, the edited cells migrate with similar speeds and persistence as unmodified cells in a variety of motility assays, and have nearly normal cell shapes. These results might partially be explained by our observation that GFP‐β‐actin incorporates into the F‐actin network in biallelic edited cells at similar efficiencies as normal β‐actin in unedited cells. Additionally, the edited cells significantly upregulate γ‐actin, perhaps helping to compensate for the loss of normal β‐actin. Interestingly, biallelic edited cells have only modest changes in global gene expression relative to the monoallelic line, as measured by RNA sequencing. While monoallelic edited cells downregulate expression of the tagged allele and are thus only weakly fluorescent, biallelic edited cells are quite bright and well‐suited for live cell microscopy. The nondisruptive phenotype and direct interpretability of this fluorescent tagging approach make it a promising tool for studying actin dynamics in these rapidly migrating and highly phagocytic cells.
Collapse
Affiliation(s)
- Rikki M Garner
- Biophysics Program, Stanford University School of Medicine, Stanford, CA.,Department of Biology, Howard Hughes Medical Institute, University of Washington, Seattle, WA
| | - Gemini Skariah
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA
| | - Amalia Hadjitheodorou
- Department of Bioengineering, Stanford University Schools of Medicine and Engineering, Stanford, CA
| | - Nathan M Belliveau
- Department of Biology, Howard Hughes Medical Institute, University of Washington, Seattle, WA
| | - Andrew Savinov
- Department of Genome Sciences, University of Washington, Seattle, WA
| | - Matthew J Footer
- Department of Biology, Howard Hughes Medical Institute, University of Washington, Seattle, WA
| | - Julie A Theriot
- Department of Biology, Howard Hughes Medical Institute, University of Washington, Seattle, WA
| |
Collapse
|
37
|
Tsai TYC, Collins SR, Chan CK, Hadjitheodorou A, Lam PY, Lou SS, Yang HW, Jorgensen J, Ellett F, Irimia D, Davidson MW, Fischer RS, Huttenlocher A, Meyer T, Ferrell JE, Theriot JA. Efficient Front-Rear Coupling in Neutrophil Chemotaxis by Dynamic Myosin II Localization. Dev Cell 2020; 49:189-205.e6. [PMID: 31014479 DOI: 10.1016/j.devcel.2019.03.025] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 02/01/2019] [Accepted: 03/26/2019] [Indexed: 12/23/2022]
Abstract
Efficient chemotaxis requires rapid coordination between different parts of the cell in response to changing directional cues. Here, we investigate the mechanism of front-rear coordination in chemotactic neutrophils. We find that changes in the protrusion rate at the cell front are instantaneously coupled to changes in retraction at the cell rear, while myosin II accumulation at the rear exhibits a reproducible 9-15-s lag. In turning cells, myosin II exhibits dynamic side-to-side relocalization at the cell rear in response to turning of the leading edge and facilitates efficient turning by rapidly re-orienting the rear. These manifestations of front-rear coupling can be explained by a simple quantitative model incorporating reversible actin-myosin interactions with a rearward-flowing actin network. Finally, the system can be tuned by the degree of myosin regulatory light chain (MRLC) phosphorylation, which appears to be set in an optimal range to balance persistence of movement and turning ability.
Collapse
Affiliation(s)
- Tony Y-C Tsai
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA; Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sean R Collins
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Caleb K Chan
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amalia Hadjitheodorou
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA; Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Pui-Ying Lam
- Department of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706, USA; Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sunny S Lou
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA; Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hee Won Yang
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Julianne Jorgensen
- Department of Surgery, BioMEMS Resource Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Felix Ellett
- Department of Surgery, BioMEMS Resource Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Daniel Irimia
- Department of Surgery, BioMEMS Resource Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Michael W Davidson
- National High Magnetic Field Laboratory, Department of Biological Science, The Florida State University, Tallahassee, FL 32304, USA
| | - Robert S Fischer
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anna Huttenlocher
- Department of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Tobias Meyer
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - James E Ferrell
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA; Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Julie A Theriot
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
38
|
Abstract
Neutrophils often communicate with each other and coordinate their actions to seal off large sites of injury and infection that individual neutrophils could not cover. The concerted actions of neutrophils are essential for the expeditious protection of healthy tissues from wounds and microbes. These processes, collectively known as swarming, are typically studied in vivo in mice. However, these studies are low throughput and their relevance to human disease is limited. Recently, new tools have been developed for the study of human neutrophil swarming ex vivo. The emergent microscale swarming assays are providing significant insights into the molecular mediators of swarming. By enabling the direct study of human cells, these assays also shed new light on human diseases and host responses against infections. Here, we describe a robust technique for manufacturing microscale swarming arrays with live microbial targets (e.g., clusters of Candida albicans). These arrays allow for the direct, precise, and efficient interrogation of the antimicrobial functions of human swarming against a variety of targets.
Collapse
Affiliation(s)
- Alex Hopke
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Shriners Hospital for Children, Boston, MA, USA
| | - Daniel Irimia
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Shriners Hospital for Children, Boston, MA, USA.
| |
Collapse
|
39
|
Martínez-Burgo B, Cobb SL, Pohl E, Kashanin D, Paul T, Kirby JA, Sheerin NS, Ali S. A C-terminal CXCL8 peptide based on chemokine-glycosaminoglycan interactions reduces neutrophil adhesion and migration during inflammation. Immunology 2019; 157:173-184. [PMID: 31013364 DOI: 10.1111/imm.13063] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/27/2019] [Accepted: 04/05/2019] [Indexed: 12/15/2022] Open
Abstract
Leucocyte recruitment is critical during many acute and chronic inflammatory diseases. Chemokines are key mediators of leucocyte recruitment during the inflammatory response, by signalling through specific chemokine G-protein-coupled receptors (GPCRs). In addition, chemokines interact with cell-surface glycosaminoglycans (GAGs) to generate a chemotactic gradient. The chemokine interleukin-8/CXCL8, a prototypical neutrophil chemoattractant, is characterized by a long, highly positively charged GAG-binding C-terminal region, absent in most other chemokines. To examine whether the CXCL8 C-terminal peptide has a modulatory role in GAG binding during neutrophil recruitment, we synthesized the wild-type CXCL8 C-terminal [CXCL8 (54-72)] (Peptide 1), a peptide with a substitution of glutamic acid (E) 70 with lysine (K) (Peptide 2) to increase positive charge; and also, a scrambled sequence peptide (Peptide 3). Surface plasmon resonance showed that Peptide 1, corresponding to the core CXCL8 GAG-binding region, binds to GAG but Peptide 2 binding was detected at lower concentrations. In the absence of cellular GAG, the peptides did not affect CXCL8-induced calcium signalling or neutrophil chemotaxis along a diffusion gradient, suggesting no effect on GPCR binding. All peptides equally inhibited neutrophil adhesion to endothelial cells under physiological flow conditions. Peptide 2, with its greater positive charge and binding to polyanionic GAG, inhibited CXCL8-induced neutrophil transendothelial migration. Our studies suggest that the E70K CXCL8 peptide, may serve as a lead molecule for further development of therapeutic inhibitors of neutrophil-mediated inflammation based on modulation of chemokine-GAG binding.
Collapse
Affiliation(s)
- Beatriz Martínez-Burgo
- Applied Immunobiology and Transplantation Research Group, Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne, UK.,Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne, UK
| | | | - Ehmke Pohl
- Chemistry Department, Durham University, Durham, UK
| | | | | | - John A Kirby
- Applied Immunobiology and Transplantation Research Group, Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne, UK.,Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne, UK
| | - Neil S Sheerin
- Applied Immunobiology and Transplantation Research Group, Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne, UK.,Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne, UK
| | - Simi Ali
- Applied Immunobiology and Transplantation Research Group, Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne, UK.,Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne, UK
| |
Collapse
|
40
|
Park J, Baik SH, Mook-Jung I, Irimia D, Cho H. Mimicry of Central-Peripheral Immunity in Alzheimer's Disease and Discovery of Neurodegenerative Roles in Neutrophil. Front Immunol 2019; 10:2231. [PMID: 31611872 PMCID: PMC6776120 DOI: 10.3389/fimmu.2019.02231] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/03/2019] [Indexed: 12/21/2022] Open
Abstract
Neuroinflammatory roles of central innate immunity in brain parenchyma are well-regarded in the progression of neurodegenerative disorders including Alzheimer's disease (AD), however, the roles of peripheral immunity in central nervous system (CNS) diseases are less clear. Here, we created a microfluidic environment of human AD brains: microglial neuroinflammation induced by soluble amyloid-beta (Abeta), a signature molecule in AD and employed the environment to investigate the roles of neutrophils through the central-peripheral innate immunity crosstalk. We observed that soluble Abeta-activated human microglial cells produced chemoattractants for neutrophils including IL6, IL8, CCL2, CCL3/4, CCL5 and consequently induced reliable recruitment of human neutrophils. Particularly, we validated the discernable chemo-attractive roles of IL6, IL8, and CCL2 for neutrophils by interrupting the recruitment with neutralizing antibodies. Upon recruitment, microglia-neutrophils interaction results in the production of inflammatory mediators such as MIF and IL2, which are known to up-regulate neuroinflammation in AD. We envision that targeting the crosstalk between central-peripheral immune community is a potential strategy to reduce immunological burdens in other neuroinflammatory CNS diseases.
Collapse
Affiliation(s)
- Joseph Park
- The Nanoscale Science Program, Department of Mechanical Engineering and Engineering Science, Department of Biological Sciences, Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Sung Hoon Baik
- Department of Biochemistry and Biomedical Sciences, Seoul National University, Seoul, South Korea
| | - Inhee Mook-Jung
- Department of Biochemistry and Biomedical Sciences, Seoul National University, Seoul, South Korea
| | - Daniel Irimia
- Department of Surgery, BioMEMS Resource Center, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, United States
| | - Hansang Cho
- The Nanoscale Science Program, Department of Mechanical Engineering and Engineering Science, Department of Biological Sciences, Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, NC, United States.,Department of Surgery, BioMEMS Resource Center, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, United States.,Department of Biophysics, Institute of Quantum Biology, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
41
|
Rinschen MM, Ivanisevic J, Giera M, Siuzdak G. Identification of bioactive metabolites using activity metabolomics. Nat Rev Mol Cell Biol 2019; 20:353-367. [PMID: 30814649 PMCID: PMC6613555 DOI: 10.1038/s41580-019-0108-4] [Citation(s) in RCA: 655] [Impact Index Per Article: 109.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The metabolome, the collection of small-molecule chemical entities involved in metabolism, has traditionally been studied with the aim of identifying biomarkers in the diagnosis and prediction of disease. However, the value of metabolome analysis (metabolomics) has been redefined from a simple biomarker identification tool to a technology for the discovery of active drivers of biological processes. It is now clear that the metabolome affects cellular physiology through modulation of other 'omics' levels, including the genome, epigenome, transcriptome and proteome. In this Review, we focus on recent progress in using metabolomics to understand how the metabolome influences other omics and, by extension, to reveal the active role of metabolites in physiology and disease. This concept of utilizing metabolomics to perform activity screens to identify biologically active metabolites - which we term activity metabolomics - is already having a broad impact on biology.
Collapse
Affiliation(s)
- Markus M Rinschen
- The Scripps Research Institute, Center for Metabolomics and Mass Spectrometry, La Jolla, CA, USA
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Martin Giera
- Leiden University Medical Center, Center for Proteomics & Metabolomics, Leiden, Netherlands.
| | - Gary Siuzdak
- The Scripps Research Institute, Center for Metabolomics and Mass Spectrometry, La Jolla, CA, USA.
| |
Collapse
|
42
|
Park H, Doh J. T cell migration in microchannels densely packed with T cells. Sci Rep 2019; 9:7198. [PMID: 31076592 PMCID: PMC6510777 DOI: 10.1038/s41598-019-43569-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 04/23/2019] [Indexed: 01/22/2023] Open
Abstract
T cells migrate diverse microenvironments of the body to mount antigen-specific immune responses. T cell activation, a key initial process for antigen-specific immune responses, occur in secondary lymphoid organs such as spleens and lymph nodes where high density of T cells migrates rapidly through the reticular networks formed by stromal cells. In vitro model system recapitulating key characteristics of secondary lymphoid organs, confined spaces densely packed with rapidly migrating cells, would be useful to investigate mechanisms of T cell migration. In this study, we devised a method to fabricate microchannels densely packed with T cells. Microchannel arrays with fixed height (4 μm) and length (1.5 mm) and various widths (15~80 μm) were fabricated in between trapezoid-shaped reservoirs that facilitated T cell sedimentation near microchannel entries. Microchannel surface chemistry and filling time were optimized to achieve high packing density (0.89) of T cell filling within microchannels. Particle image velocimetry (PIV) analysis method was employed to extract velocity field of microchannels densely packed with T cells. Using velocity field information, various motility parameters were further evaluated to quantitatively assess the effects of microchannel width and media tonicity on T cell motility within cell dense microenvironments.
Collapse
Affiliation(s)
- HyoungJun Park
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH) San 31, Hyoja-dong, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea
| | - Junsang Doh
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH) San 31, Hyoja-dong, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea. .,School of Interdisciplinary Bioscience and Bioengineering (I-Bio), Pohang University of Science and Technology (POSTECH) San 31, Hyoja-dong, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea. .,Department of Materials Science and Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea.
| |
Collapse
|
43
|
Boribong BP, Lenzi MJ, Li L, Jones CN. Super-Low Dose Lipopolysaccharide Dysregulates Neutrophil Migratory Decision-Making. Front Immunol 2019; 10:359. [PMID: 30915068 PMCID: PMC6422936 DOI: 10.3389/fimmu.2019.00359] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 02/12/2019] [Indexed: 12/30/2022] Open
Abstract
Neutrophils are the first responders to infection and play a pivotal role in many inflammatory diseases, including sepsis. Recent studies have shown that lipopolysaccharide (LPS), a classical pattern recognition molecule, dynamically programs innate immune responses. In this study, we show that pre-treatment with super-low levels of LPS [1 ng/mL] significantly dysregulate neutrophil migratory phenotypes, including spontaneous migration and altering neutrophil decision-making. To quantify neutrophil migratory decision-making with single-cell resolution, we developed a novel microfluidic competitive chemotaxis-chip (μC3) that exposes cells in a central channel to competing chemoattractant gradients. In this reductionist approach, we use two chemoattractants: a pro-resolution (N-Formyl-Met-Leu-Phe, fMLP) and pro-inflammatory (Leukotriene B4, LTB4) chemoattractant to model how a neutrophil makes a decision to move toward an end target chemoattractant (e.g., bacterial infection) vs. an intermediary chemoattractant (e.g., inflammatory signal). We demonstrate that naïve neutrophils migrate toward the primary end target signal in higher percentages than toward the secondary intermediary signal. As expected, we found that training with high dose LPS [100 ng/mL] influences a higher percentage of neutrophils to migrate toward the end target signal, while reducing the percentage of neutrophils that migrate toward the intermediary signal. Surprisingly, super-low dose LPS [1 ng/mL] significantly changes the ratios of migrating cells and an increased percentage of cells migrate toward the intermediary signal. Significantly, there was also an increase in the numbers of spontaneously migrating neutrophils after treatment with super-low dose LPS. These results shed light onto the directional migratory decision-making of neutrophils exposed to inflammatory training signals. Understanding these mechanisms may lead to the development of pro-resolution therapies that correct the neutrophil compass and reduce off-target organ damage.
Collapse
Affiliation(s)
- Brittany P Boribong
- Genetics, Bioinformatics, and Computational Biology, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Mark J Lenzi
- Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Liwu Li
- Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Caroline N Jones
- Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| |
Collapse
|
44
|
Liu Y, Yang Q, Cao L, Xu F. Analysis of Leukocyte Behaviors on Microfluidic Chips. Adv Healthc Mater 2019; 8:e1801406. [PMID: 30672149 DOI: 10.1002/adhm.201801406] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/05/2019] [Indexed: 01/17/2023]
Abstract
The orchestration of massive leukocytes in the immune system protects humans from invading pathogens and abnormal cells in the body. So far, researches focusing on leukocyte behaviors are performed based on both in vivo and in vitro models. The in vivo animal models are usually less controllable due to their extreme complexity and nonignorable species issue. Therefore, many researchers turn to in vitro models. With the advances in micro/nanofabrication, the microfluidic chip has emerged as a novel platform for model construction in multiple biomedical research fields. Specifically, the microfluidic chip is used to study leukocyte behaviors, due to its incomparable advantages in high throughput, precise control, and flexible integration. Moreover, the small size of the microstructures on the microfluidic chip can better mimic the native microenvironment of leukocytes, which contributes to a more reliable recapitulation. Herein are reviewed the recent advances in microfluidic chip-based leukocyte behavior analysis to provide an overview of this field. Detailed discussions are specifically focused on host defense against pathogens, immunodiagnosis, and immunotherapy studies on microfluidic chips. Finally, the current technical challenges are discussed, as well as possible innovations in this field to improve the related applications.
Collapse
Affiliation(s)
- Yan Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education; School of Life Science and Technology; Xi'an Jiaotong University; Xi'an Shaanxi 710049 China
- Bioinspired Engineering and Biomechanics Center (BEBC); Xi'an Jiaotong University; Xi'an Shaanxi 710049 China
| | - Qingzhen Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education; School of Life Science and Technology; Xi'an Jiaotong University; Xi'an Shaanxi 710049 China
- Bioinspired Engineering and Biomechanics Center (BEBC); Xi'an Jiaotong University; Xi'an Shaanxi 710049 China
| | - Lei Cao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education; School of Life Science and Technology; Xi'an Jiaotong University; Xi'an Shaanxi 710049 China
- Bioinspired Engineering and Biomechanics Center (BEBC); Xi'an Jiaotong University; Xi'an Shaanxi 710049 China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education; School of Life Science and Technology; Xi'an Jiaotong University; Xi'an Shaanxi 710049 China
- Bioinspired Engineering and Biomechanics Center (BEBC); Xi'an Jiaotong University; Xi'an Shaanxi 710049 China
| |
Collapse
|
45
|
Boribong BP, Rahimi A, Jones CN. Microfluidic Platform to Quantify Neutrophil Migratory Decision-Making. Methods Mol Biol 2019; 1960:113-122. [PMID: 30798526 DOI: 10.1007/978-1-4939-9167-9_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
Abstract
Neutrophils are the most abundant leukocytes in blood, serving as the first line of host defense in tissue damage and infections. Upon activation by chemokines released from pathogens or injured tissues, neutrophils migrate through complex tissue microenvironments toward sites of infections along the chemokine gradients, in a process named chemotaxis. However, current methods for measuring neutrophil chemotaxis require large volumes of blood and are often bulk, endpoint measurements. To address the need for rapid and robust assays, we engineered a novel dual gradient microfluidic platform that precisely quantifies neutrophil migratory decision-making with high temporal resolution. Here, we present a protocol to measure neutrophil migratory phenotypes (velocity, directionality) with single-cell resolution.
Collapse
Affiliation(s)
- Brittany P Boribong
- Genetics, Bioinformatics, and Computational Biology, Virginia Tech, Blacksburg, VA, USA
| | - Amina Rahimi
- Department of Biochemistry, Virginia Tech, Blacksburg, VA, USA
| | - Caroline N Jones
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
46
|
Nakayama H, Kusumoto C, Nakahara M, Fujiwara A, Higashiyama S. Semaphorin 3F and Netrin-1: The Novel Function as a Regulator of Tumor Microenvironment. Front Physiol 2018; 9:1662. [PMID: 30532711 PMCID: PMC6265511 DOI: 10.3389/fphys.2018.01662] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 11/05/2018] [Indexed: 01/13/2023] Open
Abstract
Axon guidance molecules play an important role in regulating proper neuronal networking during neuronal development. They also have non-neuronal properties, which include angiogenesis, inflammation, and tumor development. Semaphorin 3F (SEMA3F), a member of the class 3 semaphorins, was initially identified as an axon guidance factor, that repels axons and collapses growth cones. However, SEMA3F has similar effects on endothelial cells (ECs) and tumor cells. In this review, we discuss the novel molecular mechanisms underlying SEMA3F activity in vascular and tumor biology. Recent evidence suggests that SEMA3F functions as a PI3K-Akt-mTOR inhibitor in mammalian cells, including T cells, ECs, and tumor cells. Therefore, SEMA3F may have broad therapeutic implications. We also discuss the key role of axon guidance molecules as regulators of the tumor microenvironment. Netrin-1, a chemoattractant factor in the neuronal system, promotes tumor progression by enhancing angiogenesis and metastasis. Moreover, our recent studies demonstrate that netrin-1/neogenin interactions augment CD4+ T cell chemokinesis and elicit pro-inflammatory responses, suggesting that netrin-1 plays a key role in modulating the function of a tumor and its surrounding cells in the tumor microenvironment. Overall, this review focuses on SEMA3F and netrin-1 signaling mechanisms to understand the diverse biological functions of axon guidance molecules.
Collapse
Affiliation(s)
- Hironao Nakayama
- Department of Medical Science and Technology, Hiroshima International University, Higashihiroshima, Japan.,Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Toon, Japan
| | - Chiaki Kusumoto
- Department of Medical Science and Technology, Hiroshima International University, Higashihiroshima, Japan
| | - Masako Nakahara
- Department of Medical Science and Technology, Hiroshima International University, Higashihiroshima, Japan
| | - Akira Fujiwara
- Department of Medical Science and Technology, Hiroshima International University, Higashihiroshima, Japan
| | - Shigeki Higashiyama
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Ehime University, Toon, Japan
| |
Collapse
|
47
|
Wang X, Jodoin E, Jorgensen J, Lee J, Markmann JJ, Cataltepe S, Irimia D. Progressive mechanical confinement of chemotactic neutrophils induces arrest, oscillations, and retrotaxis. J Leukoc Biol 2018; 104:1253-1261. [PMID: 30129679 DOI: 10.1002/jlb.5ta0318-110rrr] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 07/18/2018] [Accepted: 07/19/2018] [Indexed: 12/25/2022] Open
Abstract
Neutrophils reach the sites of inflammation and infection in a timely manner by navigating efficiently through mechanically complex interstitial spaces, following the guidance of chemical gradients. However, our understanding of how neutrophils that follow chemical cues overcome mechanical obstacles in their path is restricted by the limitations of current experimental systems. Observations in vivo provide limited insights due to the complexity of the tissue environment. Here, we developed microfluidic devices to study the effect of progressive mechanical confinement on the migration patterns of human neutrophils toward chemical attractants. Using these devices, we identified four migration patterns: arrest, oscillation, retrotaxis, and persistent migration. The proportion of these migration patterns is different in patients receiving immunosuppressant treatments after kidney transplant, patients in critical care, and neonatal patients with infections and is distinct from that in healthy donors. The occurrence of these migration patterns is independent of the nuclear lobe number of the neutrophils and depends on the integrity of their cytoskeletal components. Our study highlights the important role of mechanical cues in moving neutrophils and suggests the mechanical constriction-induced migration patterns as potential markers for infection and inflammation.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Surgery, BioMEMS Resource Center, Massachusetts General Hospital, Harvard Medical School, Shriners Burns Hospital, Boston, Massachusetts, USA
| | - Emily Jodoin
- Department of Surgery, BioMEMS Resource Center, Massachusetts General Hospital, Harvard Medical School, Shriners Burns Hospital, Boston, Massachusetts, USA
| | - Julianne Jorgensen
- Department of Surgery, BioMEMS Resource Center, Massachusetts General Hospital, Harvard Medical School, Shriners Burns Hospital, Boston, Massachusetts, USA
| | - Jarone Lee
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - James J Markmann
- Department of Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sule Cataltepe
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel Irimia
- Department of Surgery, BioMEMS Resource Center, Massachusetts General Hospital, Harvard Medical School, Shriners Burns Hospital, Boston, Massachusetts, USA
| |
Collapse
|
48
|
Kim JJ, Reátegui E, Hopke A, Jalali F, Roushan M, Doyle PS, Irimia D. Large-scale patterning of living colloids for dynamic studies of neutrophil-microbe interactions. LAB ON A CHIP 2018; 18:1514-1520. [PMID: 29770423 PMCID: PMC5995581 DOI: 10.1039/c8lc00228b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Neutrophils are the first white blood cells to respond to microbes and to limit their invasion of the body. However, the growth of the microbes into colonies often challenges the neutrophils ability to contain them. To study the interactions between neutrophils and microbial colonies, we designed an assay for arranging microbes in clusters of controlled size (i.e. living colloids). The patterned microbes in the living colloid are mechanically trapped inside the wells and fully accessible to neutrophils. Using the assay, we studied the interactions between human neutrophils and Candida albicans and Staphylococcus aureus, two common human pathogens. We also probed the susceptibility of C. albicans colloids to caspofungin, a common antifungal drug.
Collapse
Affiliation(s)
- Jae Jung Kim
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | | | | | | | | | | | | |
Collapse
|
49
|
Saxena N, Mogha P, Dash S, Majumder A, Jadhav S, Sen S. Matrix elasticity regulates mesenchymal stem cell chemotaxis. J Cell Sci 2018. [PMID: 29535208 DOI: 10.1242/jcs.211391] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Efficient homing of human mesenchymal stem cells (hMSCs) is likely to be dictated by a combination of physical and chemical factors present in the microenvironment. However, crosstalk between the physical and chemical cues remains incompletely understood. Here, we address this question by probing the efficiency of epidermal growth factor (EGF)-induced hMSC chemotaxis on substrates of varying stiffness (3, 30 and 600 kPa) inside a polydimethylsiloxane (PDMS) microfluidic device. Chemotactic speed was found to be the sum of a stiffness-dependent component and a chemokine concentration-dependent component. While the stiffness-dependent component scaled inversely with stiffness, the chemotactic component was independent of stiffness. Faster chemotaxis on the softest 3 kPa substrates is attributed to a combination of weaker adhesions and higher protrusion rate. While chemotaxis was mildly sensitive to contractility inhibitors, suppression of chemotaxis upon actin depolymerization demonstrates the role of actin-mediated protrusions in driving chemotaxis. In addition to highlighting the collective influence of physical and chemical cues in chemotactic migration, our results suggest that hMSC homing is more efficient on softer substrates.
Collapse
Affiliation(s)
- Neha Saxena
- Department of Chemical Engineering, IIT, Bombay, Maharashtra 400076, India
| | - Pankaj Mogha
- Department of Chemical Engineering, IIT, Bombay, Maharashtra 400076, India
| | - Silalipi Dash
- Department of Chemical Engineering, IIT, Bombay, Maharashtra 400076, India
| | - Abhijit Majumder
- Department of Chemical Engineering, IIT, Bombay, Maharashtra 400076, India
| | - Sameer Jadhav
- Department of Chemical Engineering, IIT, Bombay, Maharashtra 400076, India
| | - Shamik Sen
- Department of Bioscience and Bioengineering, IIT, Bombay, Maharashtra 400076, India
| |
Collapse
|
50
|
Tay HM, Dalan R, Li KHH, Boehm BO, Hou HW. A Novel Microdevice for Rapid Neutrophil Purification and Phenotyping in Type 2 Diabetes Mellitus. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:1702832. [PMID: 29168915 DOI: 10.1002/smll.201702832] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 09/21/2017] [Indexed: 06/07/2023]
Abstract
Neutrophil dysfunction is strongly linked to type 2 diabetes mellitus (T2DM) pathophysiology, but the prognostic potential of neutrophil biomarkers remains largely unexplored due to arduous leukocyte isolation methods. Herein, a novel integrated microdevice is reported for single-step neutrophil sorting and phenotyping (chemotaxis and formation of neutrophil extracellular traps (NETosis)) using small blood volumes (fingerprick). Untouched neutrophils are purified on-chip from whole blood directly using biomimetic cell margination and affinity-based capture, and are exposed to preloaded chemoattractant or NETosis stimulant to initiate chemotaxis or NETosis, respectively. Device performance is first characterized using healthy and in vitro inflamed blood samples (tumor necrosis factor alpha, high glucose), followed by clinical risk stratification in a cohort of subjects with T2DM. Interestingly, "high-risk" T2DM patients characterized by severe chemotaxis impairment reveal significantly higher C-reactive protein levels and poor lipid metabolism characteristics as compared to "low-risk" subjects, and their neutrophil chemotaxis responses can be mitigated after in vitro metformin treatment. Overall, this unique and user-friendly microfluidics immune health profiling strategy can significantly aid the quantification of chemotaxis and NETosis in clinical settings, and be further translated into a tool for risk stratification and precision medicine methods in subjects with metabolic diseases such as T2DM.
Collapse
Affiliation(s)
- Hui Min Tay
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Clinical Sciences Building Level 11, Singapore, 308232, Singapore
| | - Rinkoo Dalan
- Endocrine and Diabetes, Tan Tock Seng Hospital, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
| | - King Ho Holden Li
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Block N3, Singapore, 639798, Singapore
| | - Bernhard O Boehm
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Clinical Sciences Building Level 11, Singapore, 308232, Singapore
- Endocrine and Diabetes, Tan Tock Seng Hospital, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
| | - Han Wei Hou
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Clinical Sciences Building Level 11, Singapore, 308232, Singapore
| |
Collapse
|