1
|
Zhao L, Shireman J, Probelsky S, Rigg B, Wang X, Huff WX, Kwon JH, Dey M. CCL21 Induces Plasmacytoid Dendritic Cell Migration and Activation in a Mouse Model of Glioblastoma. Cancers (Basel) 2024; 16:3459. [PMID: 39456552 PMCID: PMC11506458 DOI: 10.3390/cancers16203459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells that are traditionally divided into two distinct subsets: myeloid DCs (mDCs) and plasmacytoid DCs (pDCs). pDCs are known for their ability to secrete large amounts of cytokine type I interferons (IFN- α). In our previous work, we have demonstrated that pDC infiltration promotes glioblastoma (GBM) tumor immunosuppression through decreased IFN-α secretion via TLR-9 signaling and increased suppressive function of regulatory T cells (Tregs) via increased IL-10 secretion, resulting in poor overall outcomes in mouse models of GBM. Further dissecting the overall mechanism of pDC-mediated GBM immunosuppression, in this study, we identified CCL21 as highly upregulated by multiple GBM cell lines, which recruit pDCs to tumor sites via CCL21-CCR7 signaling. Furthermore, pDCs are activated by CCL21 in the GBM microenvironment through intracellular signaling of β-arrestin and CIITA. Finally, we found that CCL21-treated pDCs directly suppress CD8+ T cell proliferation without affecting regulatory T cells (Tregs) differentiation, which is considered the canonical pathway of immunotolerant regulation. Taken together, our results show that pDCs play a multifaced role in GBM immunosuppression, and CCL21 could be a novel therapeutic target in GBM to overcome pDC-mediated immunosuppression.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Jack Shireman
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Samantha Probelsky
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Bailey Rigg
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Xiaohu Wang
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Wei X. Huff
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (W.X.H.); (J.H.K.)
| | - Jae H. Kwon
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (W.X.H.); (J.H.K.)
| | - Mahua Dey
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| |
Collapse
|
2
|
Serrano JC, Gillrie MR, Li R, Ishamuddin SH, Moeendarbary E, Kamm RD. Microfluidic-Based Reconstitution of Functional Lymphatic Microvasculature: Elucidating the Role of Lymphatics in Health and Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302903. [PMID: 38059806 PMCID: PMC10837354 DOI: 10.1002/advs.202302903] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/17/2023] [Indexed: 12/08/2023]
Abstract
The knowledge of the blood microvasculature and its functional role in health and disease has grown significantly attributable to decades of research and numerous advances in cell biology and tissue engineering; however, the lymphatics (the secondary vascular system) has not garnered similar attention, in part due to a lack of relevant in vitro models that mimic its pathophysiological functions. Here, a microfluidic-based approach is adopted to achieve precise control over the biological transport of growth factors and interstitial flow that drive the in vivo growth of lymphatic capillaries (lymphangiogenesis). The engineered on-chip lymphatics with in vivo-like morphology exhibit tissue-scale functionality with drainage rates of interstitial proteins and molecules comparable to in vivo standards. Computational and scaling analyses of the underlying transport phenomena elucidate the critical role of the three-dimensional geometry and lymphatic endothelium in recapitulating physiological drainage. Finally, the engineered on-chip lymphatics enabled studies of lymphatic-immune interactions that revealed inflammation-driven responses by the lymphatics to recruit immune cells via chemotactic signals similar to in vivo, pathological events. This on-chip lymphatics platform permits the interrogation of various lymphatic biological functions, as well as screening of lymphatic-based therapies such as interstitial absorption of protein therapeutics and lymphatic immunomodulation for cancer therapy.
Collapse
Affiliation(s)
- Jean C. Serrano
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Mark R. Gillrie
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Medicine University of CalgaryCalgaryABT2N 1N4Canada
| | - Ran Li
- Center for Systems Biology Massachusetts General Hospital Research InstituteBostonMA02114USA
| | - Sarah H. Ishamuddin
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Emad Moeendarbary
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Mechanical EngineeringUniversity College LondonTorrington PlaceLondonWC1E 7JEUK
- 199 Biotechnologies LtdGloucester RoadLondonW2 6LDUK
| | - Roger D. Kamm
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
3
|
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
|
4
|
ElGindi M, Sapudom J, Garcia Sabate A, Chesney Quartey B, Alatoom A, Al-Sayegh M, Li R, Chen W, Teo J. Effects of an aged tissue niche on the immune potency of dendritic cells using simulated microgravity. NPJ AGING 2023; 9:14. [PMID: 37393393 DOI: 10.1038/s41514-023-00111-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/18/2023] [Indexed: 07/03/2023]
Abstract
Microgravity accelerates the aging of various physiological systems, and it is well acknowledged that aged individuals and astronauts both have increased susceptibility to infections and poor response to vaccination. Immunologically, dendritic cells (DCs) are the key players in linking innate and adaptive immune responses. Their distinct and optimized differentiation and maturation phases play a critical role in presenting antigens and mounting effective lymphocyte responses for long-term immunity. Despite their importance, no studies to date have effectively investigated the effects of microgravity on DCs in their native microenvironment, which is primarily located within tissues. Here, we address a significantly outstanding research gap by examining the effects of simulated microgravity via a random positioning machine on both immature and mature DCs cultured in biomimetic collagen hydrogels, a surrogate for tissue matrices. Furthermore, we explored the effects of loose and dense tissues via differences in collagen concentration. Under these various environmental conditions, the DC phenotype was characterized using surface markers, cytokines, function, and transcriptomic profiles. Our data indicate that aged or loose tissue and exposure to RPM-induced simulated microgravity both independently alter the immunogenicity of immature and mature DCs. Interestingly, cells cultured in denser matrices experience fewer effects of simulated microgravity at the transcriptome level. Our findings are a step forward to better facilitate healthier future space travel and enhance our understanding of the aging immune system on Earth.
Collapse
Affiliation(s)
- Mei ElGindi
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, PO Box 129188, United Arab Emirates
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, PO Box 129188, United Arab Emirates
| | - Anna Garcia Sabate
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, PO Box 129188, United Arab Emirates
| | - Brian Chesney Quartey
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, PO Box 129188, United Arab Emirates
| | - Aseel Alatoom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, PO Box 129188, United Arab Emirates
| | - Mohamed Al-Sayegh
- Biology Division, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Rui Li
- Department of Biomedical Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
| | - Weiqiang Chen
- Department of Biomedical Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
- Department of Mechanical and Aerospace Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA
| | - Jeremy Teo
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, PO Box 129188, United Arab Emirates.
- Department of Biomedical Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA.
- Department of Mechanical and Aerospace Engineering, New York University, 6 MetroTech Center, Brooklyn, NY, 11201, USA.
| |
Collapse
|
5
|
Ozulumba T, Montalbine AN, Ortiz-Cárdenas JE, Pompano RR. New tools for immunologists: models of lymph node function from cells to tissues. Front Immunol 2023; 14:1183286. [PMID: 37234163 PMCID: PMC10206051 DOI: 10.3389/fimmu.2023.1183286] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
The lymph node is a highly structured organ that mediates the body's adaptive immune response to antigens and other foreign particles. Central to its function is the distinct spatial assortment of lymphocytes and stromal cells, as well as chemokines that drive the signaling cascades which underpin immune responses. Investigations of lymph node biology were historically explored in vivo in animal models, using technologies that were breakthroughs in their time such as immunofluorescence with monoclonal antibodies, genetic reporters, in vivo two-photon imaging, and, more recently spatial biology techniques. However, new approaches are needed to enable tests of cell behavior and spatiotemporal dynamics under well controlled experimental perturbation, particularly for human immunity. This review presents a suite of technologies, comprising in vitro, ex vivo and in silico models, developed to study the lymph node or its components. We discuss the use of these tools to model cell behaviors in increasing order of complexity, from cell motility, to cell-cell interactions, to organ-level functions such as vaccination. Next, we identify current challenges regarding cell sourcing and culture, real time measurements of lymph node behavior in vivo and tool development for analysis and control of engineered cultures. Finally, we propose new research directions and offer our perspective on the future of this rapidly growing field. We anticipate that this review will be especially beneficial to immunologists looking to expand their toolkit for probing lymph node structure and function.
Collapse
Affiliation(s)
- Tochukwu Ozulumba
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Alyssa N. Montalbine
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States
| | - Jennifer E. Ortiz-Cárdenas
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Rebecca R. Pompano
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
- Carter Immunology Center and University of Virginia (UVA) Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
6
|
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: 3.0] [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
|
7
|
Liu Y, Ren X, Wu J, Wilkins JA, Lin F. T Cells Chemotaxis Migration Studies with a Multi-Channel Microfluidic Device. MICROMACHINES 2022; 13:1567. [PMID: 36295920 PMCID: PMC9611841 DOI: 10.3390/mi13101567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/07/2022] [Accepted: 09/19/2022] [Indexed: 06/16/2023]
Abstract
Immune surveillance is dependent on lymphocyte migration and targeted recruitment. This can involve different modes of cell motility ranging from random walk to highly directional environment-guided migration driven by chemotaxis. This study protocol describes a flow-based microfluidic device to perform quantitative multiplex cell migration assays with the potential to investigate in real time the migratory response of T cells at the population or single-cell level. The device also allows for subsequent in situ fixation and direct fluorescence analysis of the cells in the microchannel.
Collapse
Affiliation(s)
- Yang Liu
- Department of Physics and Astronomy, University of Manitoba, 30A Sifton Rd, 301 Allen Bldg, Winnipeg, MB R3T 2N2, Canada
| | - Xiaoou Ren
- Department of Physics and Astronomy, University of Manitoba, 30A Sifton Rd, 301 Allen Bldg, Winnipeg, MB R3T 2N2, Canada
| | - Jiandong Wu
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - John A. Wilkins
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba and Health Sciences Centre, 799 JBRC, 715 McDermot Ave, Winnipeg, MB R3E 3P4, Canada
| | - Francis Lin
- Department of Physics and Astronomy, University of Manitoba, 30A Sifton Rd, 301 Allen Bldg, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
8
|
Ren J, Wang N, Guo P, Fan Y, Lin F, Wu J. Recent advances in microfluidics-based cell migration research. LAB ON A CHIP 2022; 22:3361-3376. [PMID: 35993877 DOI: 10.1039/d2lc00397j] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cell migration is crucial for many biological processes, including normal development, immune response, and tissue homeostasis and many pathological processes such as cancer metastasis and wound healing. Microfluidics has revolutionized the research in cell migration since its inception as it reduces the cost of studies and allows precise manipulation of different parameters that affect cell migratory response. Over the past decade, the field has made great strides in many directions, such as techniques for better control of the cellular microenvironment, application-oriented physiological-like models, and machine-assisted cell image analysis methods. Here we review recent developments in the field of microfluidic cell migration through the following aspects: 1) the co-culture models for studying host-pathogen interactions at single-cell resolution; 2) the spatiotemporal manipulation of the chemical gradients guiding cell migration; 3) the organ-on-chip models to study cell transmigration; and 4) the deep learning image processing strategies for cell migration data analysis. We further discuss the challenges, possible improvement and future perspectives of using microfluidic techniques to study cell migration.
Collapse
Affiliation(s)
- Jiaqi Ren
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Ning Wang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Piao Guo
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang University Cancer Center, Hangzhou, 310003, China
| | - Yanping Fan
- School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Francis Lin
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada.
| | - Jiandong Wu
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
9
|
Okuma A, Ishida Y, Kawara T, Hisada S, Araki S. Secretory co-factors in next-generation cellular therapies for cancer. Front Immunol 2022; 13:907022. [PMID: 36059449 PMCID: PMC9433659 DOI: 10.3389/fimmu.2022.907022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Since chimeric antigen receptor (CAR) T-cell therapies for hematologic malignancies were approved by the U.S. Food and Drug Administration, numerous "next-generation" CAR T cells have been developed to improve their safety, efficacy, and applicability. Although some of these novel therapeutic strategies are promising, it remains difficult to apply these therapies to solid tumors and to control adverse effects, such as cytokine release syndrome and neurotoxicity. CAR T cells are generated using highly scalable genetic engineering techniques. One of the major strategies for producing next-generation CAR T cells involves the integration of useful co-factor(s) into the artificial genetic design of the CAR gene, resulting in next-generation CAR T cells that express both CAR and the co-factor(s). Many soluble co-factors have been reported for CAR T cells and their therapeutic effects and toxicity have been tested by systemic injection; therefore, CAR T cells harnessing secretory co-factors could be close to clinical application. Here, we review the various secretory co-factors that have been reported to improve the therapeutic efficacy of CAR T cells and ameliorate adverse events. In addition, we discuss the different co-factor expression systems that have been used to optimize their beneficial effects. Altogether, we demonstrate that combining CAR T cells with secretory co-factors will lead to next-generation CAR T-cell therapies that can be used against broader types of cancers and might provide advanced tools for more complicated synthetic immunotherapies.
Collapse
Affiliation(s)
- Atsushi Okuma
- Center for Exploratory Research, Research and Development Group, Hitachi Ltd., Kobe, Japan
| | | | | | | | | |
Collapse
|
10
|
Gowhari Shabgah A, Al-Obaidi ZMJ, Sulaiman Rahman H, Kamal Abdelbasset W, Suksatan W, Bokov DO, Thangavelu L, Turki Jalil A, Jadidi-Niaragh F, Mohammadi H, Mashayekhi K, Gholizadeh Navashenaq J. Does CCL19 act as a double-edged sword in cancer development? Clin Exp Immunol 2021; 207:164-175. [PMID: 35020885 PMCID: PMC8982982 DOI: 10.1093/cei/uxab039] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 11/08/2021] [Accepted: 12/22/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer is considered a life-threatening disease, and several factors are involved in its development. Chemokines are small proteins that physiologically exert pivotal roles in lymphoid and non-lymphoid tissues. The imbalance or dysregulation of chemokines has contributed to the development of several diseases, especially cancer. CCL19 is one of the homeostatic chemokines that is abundantly expressed in the thymus and lymph nodes. This chemokine, which primarily regulates immune cell trafficking, is involved in cancer development. Through the induction of anti-tumor immune responses and inhibition of angiogenesis, CCL19 exerts tumor-suppressive functions. In contrast, CCL19 also acts as a tumor-supportive factor by inducing inflammation, cell growth, and metastasis. Moreover, CCL19 dysregulation in several cancers, including colorectal, breast, pancreatic, and lung cancers, has been considered a tumor biomarker for diagnosis and prognosis. Using CCL19-based therapeutic approaches has also been proposed to overcome cancer development. This review will shed more light on the multifarious function of CCL19 in cancer and elucidate its application in diagnosis, prognosis, and even therapy. It is expected that the study of CCL19 in cancer might be promising to broaden our knowledge of cancer development and might introduce novel approaches in cancer management.
Collapse
Affiliation(s)
| | - Zaid Mahdi Jaber Al-Obaidi
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Alkafeel, Najaf, Iraq,Department of Chemistry and Biochemistry, College of Medicine, University of Kerbala, Karbala, Iraq
| | - Heshu Sulaiman Rahman
- Department of Physiology, College of Medicine, University of Sulaimani, Sulaimaniyah, Iraq,Department of Medical Laboratory Sciences, Komar University of Science and Technology, Sulaimaniyah, Iraq
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia,Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Dmitry O Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russian Federation,Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, Moscow, Russian Federation
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha institute of medical and Technical Sciences, Saveetha University, Chennai, India
| | - Abduladheem Turki Jalil
- Faculty of Biology and Ecology, Yanka Kupala State University of Grodno, Grodno, Belarus,College of Technical Engineering, The Islamic University, Najaf, Iraq
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran,Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Kazem Mashayekhi
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran,Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Jamshid Gholizadeh Navashenaq
- Noncommunicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran,Correspondence: Jamshid Gholizadeh Navashenaq, Bam University of Medical Sciences, Bam, Kerman, Iran. E-mail: ;
| |
Collapse
|
11
|
Alderfer L, Hall E, Hanjaya-Putra D. Harnessing biomaterials for lymphatic system modulation. Acta Biomater 2021; 133:34-45. [PMID: 34118451 PMCID: PMC9113193 DOI: 10.1016/j.actbio.2021.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/20/2021] [Accepted: 06/01/2021] [Indexed: 12/20/2022]
Abstract
The lymphatic system plays an integral part in regulating immune cell trafficking and the transport of macromolecules. However, its influence on disease progression and drug uptake is understood less than that of the vascular system. To bridge this knowledge gap, biomaterials can be used to investigate the lymphatic system and to provide novel understanding into complex disease processes, including cancer metastasis and inflammation. Insight gained from these mechanistic studies can be further used to design innovative biomaterials to modulate the immune system, improve drug delivery, and promote tissue regeneration. This review article focuses on recent advances in (i) biomaterials used for lymphatic vessel formation, (ii) models for studying lymphatic-immune cells interactions, (iii) pharmaceuticals and their interactions with the lymphatic system, (iv) and strategies for drug delivery via the lymphatic system. Finally, several challenges regarding adopting biomaterials for immunomodulation and future perspectives are discussed. STATEMENT OF SIGNIFICANCE: The lymphatic system plays an integral part in regulating immune cell trafficking and the transport of macromolecules. However, its influence on disease progression and drug uptake is understood less than that of the vascular system. This review article focuses on recent progresses in biomaterials to investigate the lymphatic system and to provide novel understanding into complex disease states. Insight gained from these mechanistic studies can be further used to design innovative biomaterials to modulate the immune system, improve drug delivery, and promote tissue regeneration. Finally, a number of challenges in adopting biomaterials for immunomodulation and future perspectives are discussed.
Collapse
|
12
|
Ando Y, Mariano C, Shen K. Engineered in vitro tumor models for cell-based immunotherapy. Acta Biomater 2021; 132:345-359. [PMID: 33857692 PMCID: PMC8434941 DOI: 10.1016/j.actbio.2021.03.076] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 12/15/2022]
Abstract
Tumor immunotherapy is rapidly evolving as one of the major pillars of cancer treatment. Cell-based immunotherapies, which utilize patient's own immune cells to eliminate cancer cells, have shown great promise in treating a range of malignancies, especially those of hematopoietic origins. However, their performance on a broader spectrum of solid tumor types still fall short of expectations in the clinical stage despite promising preclinical assessments. In this review, we briefly introduce cell-based immunotherapies and the inhibitory mechanisms in tumor microenvironments that may have contributed to this discrepancy. Specifically, a major obstacle to the clinical translation of cell-based immunotherapies is in the lack of preclinical models that can accurately assess the efficacies and mechanisms of these therapies in a (patho-)physiologically relevant manner. Lately, tissue engineering and organ-on-a-chip tools and microphysiological models have allowed for more faithful recapitulation of the tumor microenvironments, by incorporating crucial tumor tissue features such as cellular phenotypes, tissue architecture, extracellular matrix, physical parameters, and their dynamic interactions. This review summarizes the existing engineered tumor models with a focus on tumor immunology and cell-based immunotherapy. We also discuss some key considerations for the future development of engineered tumor models for immunotherapeutics. STATEMENT OF SIGNIFICANCE: Cell-based immunotherapies have shown great promise in treating hematological malignancies and some epithelial tumors. However, their performance on a broader spectrum of solid tumor types still fall short of expectations. Major obstacles include the inhibitory mechanisms in tumor microenvironments (TME) and the lack of preclinical models that can accurately assess the efficacies and mechanisms of cellular therapies in a (patho-)physiologically relevant manner. In this review, we introduce recent progress in tissue engineering and microphysiological models for more faithful recapitulation of TME for cell-based immunotherapies, and some key considerations for the future development of engineered tumor models. This overview will provide a better understanding on the role of engineered models in accelerating immunotherapeutic discoveries and clinical translations.
Collapse
Affiliation(s)
- Yuta Ando
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States
| | - Chelsea Mariano
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States
| | - Keyue Shen
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, United States; Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States; USC Stem Cell, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States.
| |
Collapse
|
13
|
Shanti A, Hallfors N, Petroianu GA, Planelles L, Stefanini C. Lymph Nodes-On-Chip: Promising Immune Platforms for Pharmacological and Toxicological Applications. Front Pharmacol 2021; 12:711307. [PMID: 34483920 PMCID: PMC8415712 DOI: 10.3389/fphar.2021.711307] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/04/2021] [Indexed: 12/14/2022] Open
Abstract
Organs-on-chip are gaining increasing attention as promising platforms for drug screening and testing applications. However, lymph nodes-on-chip options remain limited although the lymph node is one of the main determinants of the immunotoxicity of newly developed pharmacological drugs. In this review, we describe existing biomimetic lymph nodes-on-chip, their design, and their physiological relevance to pharmacology and shed the light on future directions associated with lymph node-on-chip design and implementation in drug discovery and development.
Collapse
Affiliation(s)
- Aya Shanti
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Nicholas Hallfors
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Georg A Petroianu
- College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Lourdes Planelles
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Cesare Stefanini
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| |
Collapse
|
14
|
Ren X, Getschman AE, Hwang S, Volkman BF, Klonisch T, Levin D, Zhao M, Santos S, Liu S, Cheng J, Lin F. Investigations on T cell transmigration in a human skin-on-chip (SoC) model. LAB ON A CHIP 2021; 21:1527-1539. [PMID: 33616124 PMCID: PMC8058301 DOI: 10.1039/d0lc01194k] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
A microfluidics-based three-dimensional skin-on-chip (SoC) model is developed in this study to enable quantitative studies of transendothelial and transepithelial migration of human T lymphocytes in mimicked skin inflammatory microenvironments and to test new drug candidates. The keys results include 1) CCL20-dependent T cell transmigration is significantly inhibited by an engineered CCL20 locked dimer (CCL20LD), supporting the potential immunotherapeutic use of CCL20LD for treating skin diseases such as psoriasis; 2) transepithelial migration of T cells in response to a CXCL12 gradient mimicking T cell egress from the skin is significantly reduced by a sphingosine-1-phosphate (S1P) background, suggesting the role of S1P for T cell retention in inflamed skin tissues; and 3) T cell transmigration is induced by inflammatory cytokine stimulated epithelial cells in the SoC model. Collectively, the developed SoC model recreates a dynamic multi-cellular micro-environment that enables quantitative studies of T cell transmigration at a single cell level in response to physiological cutaneous inflammatory mediators and potential drugs.
Collapse
Affiliation(s)
- Xiaoou Ren
- Department of Physics and Astronomy, University of Manitoba, 30A Sifton Rd, 301 Allen Bldg, Winnipeg, MB R3T 2N2, Canada. and Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Anthony E Getschman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Samuel Hwang
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95816, USA
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Thomas Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - David Levin
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Min Zhao
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95816, USA and Department of Ophthalmology & Vision Science, California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Susy Santos
- Victoria General Hospital, Winnipeg, MB R3T 2E8, Canada
| | - Song Liu
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Jasmine Cheng
- Department of Physics and Astronomy, University of Manitoba, 30A Sifton Rd, 301 Allen Bldg, Winnipeg, MB R3T 2N2, Canada.
| | - Francis Lin
- Department of Physics and Astronomy, University of Manitoba, 30A Sifton Rd, 301 Allen Bldg, Winnipeg, MB R3T 2N2, Canada. and Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada and Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95816, USA and Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| |
Collapse
|
15
|
Loef EJ, Sheppard HM, Birch NP, Dunbar PR. Live-Cell Microscopy Reveals That Human T Cells Primarily Respond Chemokinetically Within a CCL19 Gradient That Induces Chemotaxis in Dendritic Cells. Front Immunol 2021; 12:628090. [PMID: 33841411 PMCID: PMC8033042 DOI: 10.3389/fimmu.2021.628090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/05/2021] [Indexed: 11/17/2022] Open
Abstract
The ability to study migratory behavior of immune cells is crucial to understanding the dynamic control of the immune system. Migration induced by chemokines is often assumed to be directional (chemotaxis), yet commonly used end-point migration assays are confounded by detecting increased cell migration that lacks directionality (chemokinesis). To distinguish between chemotaxis and chemokinesis we used the classic “under-agarose assay” in combination with video-microscopy to monitor migration of CCR7+ human monocyte-derived dendritic cells and T cells in response to a concentration gradient of CCL19. Formation of the gradients was visualized with a fluorescent marker and lasted several hours. Monocyte-derived dendritic cells migrated chemotactically towards the CCL19 gradient. In contrast, T cells exhibited a biased random walk that was largely driven by increased exploratory chemokinesis towards CCL19. This dominance of chemokinesis over chemotaxis in T cells is consistent with CCR7 ligation optimizing T cell scanning of antigen-presenting cells in lymphoid tissues.
Collapse
Affiliation(s)
- Evert J Loef
- School of Biological Science, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Hilary M Sheppard
- School of Biological Science, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Nigel P Birch
- School of Biological Science, University of Auckland, Auckland, New Zealand.,Centre for Brain Research and Brain Research New Zealand, Rangahau Roro Aotearoa, University of Auckland, Auckland, New Zealand
| | - P Rod Dunbar
- School of Biological Science, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| |
Collapse
|
16
|
Luo H, Su J, Sun R, Sun Y, Wang Y, Dong Y, Shi B, Jiang H, Li Z. Coexpression of IL7 and CCL21 Increases Efficacy of CAR-T Cells in Solid Tumors without Requiring Preconditioned Lymphodepletion. Clin Cancer Res 2020; 26:5494-5505. [PMID: 32816947 DOI: 10.1158/1078-0432.ccr-20-0777] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/02/2020] [Accepted: 08/03/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE T-cell recruitment, survival, and proliferation are the important limitations to chimeric antigen receptor (CAR) T cells therapy in the treatment of solid tumors. In this study, we engineered CAR-T cells to coexpress cytokines IL7 and CCL21 (7 × 21 CAR-T), a cytokine combination in order to improve proliferation and chemotaxis of CAR-T cells. EXPERIMENTAL DESIGN CLDN18.2-specific second-generation CAR-T cells coexpressing cytokines were prepared using retroviral vector transduction. The proliferation and migration of genetically engineered CAR-T cells were evaluated in vitro. The antitumor activities of genetically engineered CAR-T cells were evaluated against multiple solid tumors in C57BL/6 mice in vivo. RESULTS In vitro, the proliferation and chemotaxis of 7 × 21 CAR-T cells are significantly improved when compared with those of the conventional CAR-T cells. In vivo, 7 × 21 CAR-T cells revealed superior therapeutic effects to either conventional CAR-T cells or 7 × 19 CAR-T cells which coexpress IL7 and CCL19 as previously reported in three different solid tumors without cyclophosphamide precondition. Interestingly, 7 × 21 CAR-T cells could also suppress the tumor growth with heterogeneous antigen expression and even induce tumor complete remission. Mechanistically, IL7 and CCL21 significantly improved survival and infiltration of CAR-T cells and dendritic cells in tumor. In addition, CCL21 also inhibited the tumor angiogenesis as proved by IHC. CONCLUSIONS Coexpression of IL7 and CCL21 could boost CAR-T cells' antitumor activity, and 7 × 21 CAR-T cells may be served as a promising therapy strategy for solid tumors.
Collapse
Affiliation(s)
- Hong Luo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jingwen Su
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruixin Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yansha Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yiwei Dong
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bizhi Shi
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zonghai Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China. .,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,CARsgen Therapeutics, Shanghai, China
| |
Collapse
|
17
|
Rizeq B, Malki MI. The Role of CCL21/CCR7 Chemokine Axis in Breast Cancer Progression. Cancers (Basel) 2020; 12:E1036. [PMID: 32340161 PMCID: PMC7226115 DOI: 10.3390/cancers12041036] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/01/2020] [Accepted: 04/11/2020] [Indexed: 12/14/2022] Open
Abstract
Breast cancer is a leading cause of cancer-related deaths worldwide, predominantly caused by metastasis. It is generally accepted that the pattern of breast cancer metastasis is largely determined by the interaction between the chemokine receptors on cancer cells and the chemokines expressed at the sites of metastatic disease. Chemokine receptors belong to the G-protein-coupled receptors (GPCRs) family that appear to be implicated in inflammatory diseases, tumor growth and metastasis. One of its members, C-C Chemokine receptor 7 (CCR7), binds chemokines CCL19 and CCL21, which are important for tissue homeostasis, immune surveillance and tumorigenesis. These receptors have been shown to induce the pathobiology of breast cancer due to their ability to induce cellular proliferation and migration upon the binding of the cognate chemokine receptors. The underlying signaling pathways and exact cellular interactions within this biological system are not fully understood and need further insights. Thus, in this review, we summarize the essential roles of CCR7 and its receptors in breast cancer progression. Furthermore, we discuss the mechanisms of regulation that may lead to novel opportunities for therapeutic intervention. Despite the enormous advances in our knowledge of the nature of the chemokines in breast cancer metastasis, research about the involvement of CCR7 in cancer progression is still limited. Therefore, further studies are essential to illustrate the distinct roles of CCR7 in cancer progression and validate its potential as a preventive bio-factor for human breast cancer metastasis by targeting chemokine receptor genes.
Collapse
Affiliation(s)
| | - Mohammed Imad Malki
- College of Medicine, QU Health, Qatar University, P. O. Box. 2713, Doha, Qatar;
| |
Collapse
|
18
|
Chemotactic Responses of Jurkat Cells in Microfluidic Flow-Free Gradient Chambers. MICROMACHINES 2020; 11:mi11040384. [PMID: 32260431 PMCID: PMC7231302 DOI: 10.3390/mi11040384] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/02/2020] [Accepted: 04/02/2020] [Indexed: 12/29/2022]
Abstract
Gradients of soluble molecules coordinate cellular communication in a diverse range of multicellular systems. Chemokine-driven chemotaxis is a key orchestrator of cell movement during organ development, immune response and cancer progression. Chemotaxis assays capable of examining cell responses to different chemokines in the context of various extracellular matrices will be crucial to characterize directed cell motion in conditions which mimic whole tissue conditions. Here, a microfluidic device which can generate different chemokine patterns in flow-free gradient chambers while controlling surface extracellular matrix (ECM) to study chemotaxis either at the population level or at the single cell level with high resolution imaging is presented. The device is produced by combining additive manufacturing (AM) and soft lithography. Generation of concentration gradients in the device were simulated and experimentally validated. Then, stable gradients were applied to modulate chemotaxis and chemokinetic response of Jurkat cells as a model for T lymphocyte motility. Live imaging of the gradient chambers allowed to track and quantify Jurkat cell migration patterns. Using this system, it has been found that the strength of the chemotactic response of Jurkat cells to CXCL12 gradient was reduced by increasing surface fibronectin in a dose-dependent manner. The chemotaxis of the Jurkat cells was also found to be governed not only by the CXCL12 gradient but also by the average CXCL12 concentration. Distinct migratory behaviors in response to chemokine gradients in different contexts may be physiologically relevant for shaping the host immune response and may serve to optimize the targeting and accumulation of immune cells to the inflammation site. Our approach demonstrates the feasibility of using a flow-free gradient chamber for evaluating cross-regulation of cell motility by multiple factors in different biologic processes.
Collapse
|
19
|
Lämmermann T, Kastenmüller W. Concepts of GPCR-controlled navigation in the immune system. Immunol Rev 2020; 289:205-231. [PMID: 30977203 PMCID: PMC6487968 DOI: 10.1111/imr.12752] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/01/2019] [Accepted: 02/03/2019] [Indexed: 12/11/2022]
Abstract
G‐protein–coupled receptor (GPCR) signaling is essential for the spatiotemporal control of leukocyte dynamics during immune responses. For efficient navigation through mammalian tissues, most leukocyte types express more than one GPCR on their surface and sense a wide range of chemokines and chemoattractants, leading to basic forms of leukocyte movement (chemokinesis, haptokinesis, chemotaxis, haptotaxis, and chemorepulsion). How leukocytes integrate multiple GPCR signals and make directional decisions in lymphoid and inflamed tissues is still subject of intense research. Many of our concepts on GPCR‐controlled leukocyte navigation in the presence of multiple GPCR signals derive from in vitro chemotaxis studies and lower vertebrates. In this review, we refer to these concepts and critically contemplate their relevance for the directional movement of several leukocyte subsets (neutrophils, T cells, and dendritic cells) in the complexity of mouse tissues. We discuss how leukocyte navigation can be regulated at the level of only a single GPCR (surface expression, competitive antagonism, oligomerization, homologous desensitization, and receptor internalization) or multiple GPCRs (synergy, hierarchical and non‐hierarchical competition, sequential signaling, heterologous desensitization, and agonist scavenging). In particular, we will highlight recent advances in understanding GPCR‐controlled leukocyte navigation by intravital microscopy of immune cells in mice.
Collapse
Affiliation(s)
- Tim Lämmermann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | | |
Collapse
|
20
|
Na JT, Xue CD, Li YJ, Wang Y, Liu B, Qin KR. Precise generation of dynamic biochemical signals by controlling the programmable pump in a Y-shaped microfluidic chip with a "christmas tree" inlet. Electrophoresis 2020; 41:883-890. [PMID: 31901145 DOI: 10.1002/elps.201900400] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 01/01/2023]
Abstract
The generation of dynamic biochemical signals in a microfluidic control system is of importance for the study of the interaction between biological cells and their niches. However, most of microfluidic control systems are not able to provide dynamic biochemical signals with high precision and stability due to inherent mechanical vibrations caused by the actuators of the programmable pumps. In this paper, we propose a novel microfluidic feedback control system integrating an external feedback control system with a Y-shaped microfluidic chip with a "Christmas tree" inlet. The Proportional Integral Derivative (PID) controller is implemented to reduce the influence of vibrations. In order to regulate the control parameters efficiently, a mathematical model is built to describe the actuator of the programmable pump, in which a fractional-order model is utilized. Both simulation and experimental studies are carried out, confirming that the microfluidic feedback control system can precisely and stably generate desired dynamic biochemical signals.
Collapse
Affiliation(s)
- Jing-Tong Na
- School of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian, P. R. China
| | - Chun-Dong Xue
- School of Optoelectronic Engineering and Instrumentation Science, Dalian University of Technology, Dalian, P. R. China
| | - Yong-Jiang Li
- School of Optoelectronic Engineering and Instrumentation Science, Dalian University of Technology, Dalian, P. R. China
| | - Yu Wang
- School of Optoelectronic Engineering and Instrumentation Science, Dalian University of Technology, Dalian, P. R. China
| | - Bo Liu
- School of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian, P. R. China
| | - Kai-Rong Qin
- School of Optoelectronic Engineering and Instrumentation Science, Dalian University of Technology, Dalian, P. R. China
| |
Collapse
|
21
|
Lee Y, Kim SJ, Park JK. Chips-on-a-plate device for monitoring cellular migration in a microchannel-based intestinal follicle-associated epithelium model. BIOMICROFLUIDICS 2019; 13:064127. [PMID: 31893012 PMCID: PMC6930141 DOI: 10.1063/1.5128640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/04/2019] [Indexed: 05/05/2023]
Abstract
This paper describes a chips-on-a-plate (COP) device for monitoring the migration of Raji cells in the Caco-2/Raji coculture. To generate a model of the human intestinal follicle-associated epithelium (FAE), the coculture method using a conventional Transwell cell culture insert was established. Due to the structural limitations of the Transwell insert, live-cell tracking studies have not been performed previously using the existing FAE model. In this study, we designed a COP device to conduct long-term live-cell tracking of Raji cell migration using a microchannel-based FAE model. The COP device incorporates microfluidic chips integrated on a standard well plate, consistent humidity control to allow live-cell microscopy for 2 days, and microchannels connecting the two cell culture chambers of the COP device, which serve as a monitoring area for cellular migration. Using the COP device, we provide the first analysis of various migratory characteristics of Raji cells, including their chemotactic index in the microchannel-based FAE model. We showed that the migration of Raji cells could be controlled by modulating the geometry of the connecting microchannels. Cellular treatments with cytokines revealed that the cytokines increased the permeability of an FAE model with a detachment of Caco-2 cells. Live-cell monitoring of Raji cells treated with a fluorescent reagent also indicated exocytosis as a key agent of the Caco-2/Raji interaction. The COP device allows live-cell tracking analyses of cocultured cells in the microchannel-based FAE model, providing a promising tool for investigating cellular behavior associated with the recruitment of Raji to Caco-2 cells.
Collapse
Affiliation(s)
- Young Lee
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Soo Jee Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Je-Kyun Park
- Author to whom correspondence should be addressed:. Tel.: +82-42-350-4315. Fax: +82-42-350-4310
| |
Collapse
|
22
|
Grebennikov DS, Donets DO, Orlova OG, Argilaguet J, Meyerhans A, Bocharov GA. Mathematical Modeling of the Intracellular Regulation of Immune Processes. Mol Biol 2019. [DOI: 10.1134/s002689331905008x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
23
|
Ren X, Wu J, Levin D, Santos S, de Faria RL, Zhang M, Lin F. Sputum from chronic obstructive pulmonary disease patients inhibits T cell migration in a microfluidic device. Ann N Y Acad Sci 2019; 1445:52-61. [PMID: 30891781 DOI: 10.1111/nyas.14029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 01/25/2019] [Indexed: 12/29/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a common lung disease characterized by narrowed airways, resulting in serious breathing difficulty. Previous studies have demonstrated that inflammatory infiltration of leukocytes in the airway is associated with the pathogenesis of COPD. In the present study, we employed a microfluidic approach to assess the effect of COPD sputum on activated human peripheral blood T cell migration and chemotaxis under well-controlled gradient conditions. Our results showed considerable basal migration of T cells derived from peripheral blood of COPD patients and healthy controls in the medium control groups. By contrast, the migration of T cells from COPD patients and healthy controls was significantly inhibited in the presence of a gradient of sputum supernatant from COPD patients. Furthermore, chemotaxis of T cells from COPD patients or healthy subjects toward an SDF-1α gradient was clearly inhibited by sputum samples from the COPD patients. The inhibition effect revealed by the microfluidic cell migration experiments provides new information about the complex involvement of T cell trafficking in COPD.
Collapse
Affiliation(s)
- Xiaoou Ren
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Biosystems Engineering, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jiandong Wu
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David Levin
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Susy Santos
- The Victoria Institute of Clinical Research & Evaluation, Victoria General Hospital, Winnipeg, Manitoba, Canada
| | - Ricardo Lobato de Faria
- Department of Emergency and Wellness Institute, Seven Oaks General Hospital, Winnipeg, Manitoba, Canada
| | - Michael Zhang
- Department of Emergency and Wellness Institute, Seven Oaks General Hospital, Winnipeg, Manitoba, Canada
| | - Francis Lin
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Biosystems Engineering, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
24
|
Shanti A, Teo J, Stefanini C. In Vitro Immune Organs-on-Chip for Drug Development: A Review. Pharmaceutics 2018; 10:E278. [PMID: 30558264 PMCID: PMC6320867 DOI: 10.3390/pharmaceutics10040278] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 11/12/2018] [Accepted: 11/21/2018] [Indexed: 12/15/2022] Open
Abstract
The current drug development practice lacks reliable and sensitive techniques to evaluate the immunotoxicity of drug candidates, i.e., their effect on the human immune system. This, in part, has resulted in a high attrition rate for novel drugs candidates. Organ-on-chip devices have emerged as key tools that permit the study of human physiology in controlled in vivo simulating environments. Furthermore, there has been a growing interest in developing the so called "body-on-chip" devices to better predict the systemic effects of drug candidates. This review describes existing biomimetic immune organs-on-chip, highlights their physiological relevance to drug development and discovery and emphasizes the need for developing comprehensive immune system-on-chip models. Such immune models can enhance the performance of novel drug candidates during clinical trials and contribute to reducing the high attrition rate as well as the high cost associated with drug development.
Collapse
Affiliation(s)
- Aya Shanti
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, P.O. Box 127788, UAE.
| | - Jeremy Teo
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi, P.O. Box 129188, UAE.
- Department of Mechanical and Aerospace Engineering, New York University, New York City, P.O. Box 903, NY 10276-0903, USA.
| | - Cesare Stefanini
- Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, P.O. Box 127788, UAE.
| |
Collapse
|
25
|
Zhang JF, Li Y, Zhang AZ, He QQ, Du YC, Cao W. Expression and pathological significance of CC chemokine receptor 7 and its ligands in the airway of asthmatic rats exposed to cigarette smoke. J Thorac Dis 2018; 10:5459-5467. [PMID: 30416795 DOI: 10.21037/jtd.2018.08.124] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background Cigarette smoking aggravates the symptoms of asthma, leading to the rapid decline of lung function. Dendritic cells (DCs) and lymphocytes are considered initiating and promoting factors for the airway inflammation reactions of asthma. In addition, activation of CC chemokine receptor 7 (CCR7) by chemokine (C-C motif) ligand (CCL) 19 and 21 promotes DCs and T cells migration to lymphoid tissues during inflammation. We aimed to examine how cigarette smoke affects the expression of CCR7 in the lungs of asthmatic rats and explore the signaling mechanism linking CCR7 expression to exacerbation of symptoms. Methods Forty Wistar rats were randomized to four groups: control, asthma, smoke exposure, and asthma with smoke exposure groups. A rat asthma model was established by intraperitoneal ovalbumin injection. CCR7 expression was examined with immunohistochemistry and western blotting. The number of airway DCs was determined by OX62 immunohistochemistry. Interferon (INF)-γ, interleukin (IL)-4, CCL19, and CCL21 expression levels in blood and bronchioalveolar lavage fluid (BALF) were determined by enzyme-linked immunosorbent assays (ELISAs). Results Tissue CCR7 expression, peripheral blood and BALF CCL19 and CCL21 concentrations, and the number of airway DCs were significantly higher in the asthma with smoke exposure group than the asthma group (P<0.01). In addition, INF-γ expression was decreased and IL-4 increased in the asthma and asthma with smoke exposure groups compared with the control group (P<0.01), and in the asthma with smoke exposure group compared with the asthma group (P<0.01). Expression of CCR7 correlated negatively with INF-γ expression in peripheral blood and BALF (P<0.01), and positively with the airway DCs and IL-4 expression in the peripheral blood and BALF (P<0.01). Conclusions Cigarette smoking may aggravate asthma symptoms by attenuating immunity, possibly through CCR7-mediated DCs aggregation in lung tissue.
Collapse
Affiliation(s)
- Jun-Feng Zhang
- Department of Health Statistics, Public Health of Shanxi Medical University, Taiyuan 030001, China.,Publishing house, Chinese Journal of Rheumatology, Taiyuan 030001, China
| | - Yi Li
- Department of Respiration Medicine, People's Hospital of Shanxi Province, Taiyuan 030001, China
| | - Ai-Zhen Zhang
- Department of Respiration Medicine, People's Hospital of Shanxi Province, Taiyuan 030001, China
| | - Qian-Qian He
- Department of Health Statistics, Public Health of Shanxi Medical University, Taiyuan 030001, China
| | - Yong-Cheng Du
- Department of Respiration Medicine, People's Hospital of Shanxi Province, Taiyuan 030001, China
| | - Wen Cao
- Department of Respiration Medicine, People's Hospital of Shanxi Province, Taiyuan 030001, China
| |
Collapse
|
26
|
Adriani G, Pavesi A, Kamm RD. Studying TCR T cell anti-tumor activity in a microfluidic intrahepatic tumor model. Methods Cell Biol 2018; 146:199-214. [PMID: 30037462 DOI: 10.1016/bs.mcb.2018.05.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adoptive cell therapy (ACT) is showing promising results in clinical trials but many challenges remain in understanding the key role of the tumor microenvironment. These challenges constitute a major barrier to advancing the field. Therefore, it is crucial to perform preclinical tests of the developed ACT strategies in a fast and reproducible way to assess the potential for patient therapy. Here, we describe the development of an intrahepatic tumor model in a microfluidic device for screening T cell-based immunotherapeutic strategies and the role of monocytes in these therapies. This system can be used to test also the effects of supporting cytokine administration and changes in oxygen level that are typically found in a liver tumor microenvironment. As a result, these 3D microfluidic assays provide a means to quantify T cell anti-tumor activity under different conditions to optimize existing therapeutic strategies or the design of new ones.
Collapse
Affiliation(s)
- Giulia Adriani
- BioSystems and Micromechanics IRG, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Andrea Pavesi
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Roger D Kamm
- BioSystems and Micromechanics IRG, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States.
| |
Collapse
|
27
|
Microfluidic single-cell technology in immunology and antibody screening. Mol Aspects Med 2018; 59:47-61. [DOI: 10.1016/j.mam.2017.09.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 09/06/2017] [Accepted: 09/13/2017] [Indexed: 11/20/2022]
|
28
|
Abstract
Chemokine signaling is essential for coordinated cell migration in health and disease to specifically govern cell positioning in space and time. Typically, chemokines signal through heptahelical, G protein-coupled receptors to orchestrate cell migration. Notably, chemokine receptors are highly dynamic structures and signaling efficiency largely depends on the discrete contact with the ligand. Promiscuity of both chemokines and chemokine receptors, combined with biased signaling and allosteric modulation of receptor activation, guarantees a tightly controlled recruitment and positioning of individual cells within the local environment at a given time. Here, we discuss recent insights in understanding chemokine gradient formation by atypical chemokine receptors and how typical chemokine receptors can transmit distinct signals to translate guidance cues into coordinated cell locomotion in space and time.
Collapse
Affiliation(s)
- Daniel F Legler
- Biotechnology Institute Thurgau (BITg), University of Konstanz, Kreuzlingen, Switzerland
| | - Marcus Thelen
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| |
Collapse
|
29
|
Nandagopal S, Lin F, Kung SKP. Microfluidic-Based Live-Cell Analysis of NK Cell Migration In Vitro. Methods Mol Biol 2017; 1441:75-86. [PMID: 27177658 DOI: 10.1007/978-1-4939-3684-7_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Effector functions and cellular properties of natural killer (NK) cells are regulated by cellular and extracellular factors shaped by the microenvironments. NK cells express specific chemokine and non-chemokine receptors to aid preferential migrations or localizations in tissues. Good understanding of how NK-cell migratory properties are regulated in physiological and pathological microenvironments will provide further insights into the development of NK cell-based therapeutic approaches. In contrast to the commonly used conventional in vitro migration assays such as Trans-well assays that measure movements of a population of the migratory cells, microfluidic-based devices support live-cell imaging of cell migrations under a well-defined chemical gradient(s) at microscale. Subsequent analyses at single-cell level provide quantitative measurements of cell-migration parameters such as speed and Chemotactic Index, and permit distinguishing chemotaxis, chemokinesis, and chemo-repulsion. Our recent work established the use of a Y-shaped microfluidic device to study NK cell migrations in vitro. In this chapter, we described the detailed method of acquiring and analyzing NK cell migration in the microfluidic devices.
Collapse
Affiliation(s)
- Saravanan Nandagopal
- Department of Immunology, University of Manitoba, 417 Apotex Centre, 750 McDermot Avenue, Winnipeg, MB, Canada, R3E 0T5
| | - Francis Lin
- Department of Immunology, University of Manitoba, 417 Apotex Centre, 750 McDermot Avenue, Winnipeg, MB, Canada, R3E 0T5.,Department of Physics and Astronomy, University of Manitoba, Winnipeg, MB, Canada.,Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB, Canada
| | - Sam K P Kung
- Department of Immunology, University of Manitoba, 417 Apotex Centre, 750 McDermot Avenue, Winnipeg, MB, Canada, R3E 0T5.
| |
Collapse
|
30
|
Ren X, Levin D, Lin F. Cell Migration Research Based on Organ-on-Chip-Related Approaches. MICROMACHINES 2017; 8:mi8110324. [PMID: 30400514 PMCID: PMC6190356 DOI: 10.3390/mi8110324] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 10/28/2017] [Accepted: 10/28/2017] [Indexed: 11/16/2022]
Abstract
Microfluidic devices have been widely used for cell migration research over the last two decades, owing to their attractive features in cellular microenvironment control and quantitative single-cell migration analysis. However, the majority of the microfluidic cell migration studies have focused on single cell types and have configured microenvironments that are greatly simplified compared with the in-vivo conditions they aspire to model. In addition, although cell migration is considered an important target for disease diagnosis and therapeutics, very few microfluidic cell migration studies involved clinical samples from patients. Therefore, more sophisticated microfluidic systems are required to model the complex in-vivo microenvironment at the tissue or organ level for cell migration studies and to explore cell migration-related clinical applications. Research in this direction that employs organ-on-chip-related approaches for cell migration analysis has been increasingly reported in recent years. In this paper, we briefly introduce the general background of cell migration and organ-on-chip research, followed by a detailed review of specific cell migration studies using organ-on-chip-related approaches, and conclude by discussing our perspectives of the challenges, opportunities and future directions.
Collapse
Affiliation(s)
- Xiaoou Ren
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
| | - David Levin
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
| | - Francis Lin
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
| |
Collapse
|
31
|
Three-dimensional co-culture microfluidic model and its application for research on cancer stem-like cells inducing migration of endothelial cells. Biotechnol Lett 2017; 39:1425-1432. [DOI: 10.1007/s10529-017-2363-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 05/17/2017] [Indexed: 12/20/2022]
|
32
|
Ma T, Luan SL, Huang H, Sun XK, Yang YM, Zhang H, Han WD, Li H, Han Y. Upregulation of CC Chemokine Receptor 7 (CCR7) Enables Migration of Xenogeneic Human Adipose-Derived Mesenchymal Stem Cells to Rat Secondary Lymphoid Organs. Med Sci Monit 2016; 22:5206-5217. [PMID: 28035134 PMCID: PMC5221418 DOI: 10.12659/msm.902690] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background CC chemokine receptor 7 (CCR7) expression is vital for cell migration to secondary lymphoid organs (SLOs). Our previous work showed that inducing CCR7 expression enabled syngeneic mesenchymal stem cells (MSCs) to migrate into SLOs, resulting in enhanced immunosuppressive performance in mice. Given that human adipose-derived stem cells (hASCs) are widely used in clinical therapy, we further investigated whether upregulation of CCR7 enables xenogeneic hASCs to migrate to rat SLOs. Material/Methods hASCs rarely express CCR7; therefore, hASCs were transfected with lentivirus encoding rat CCR7 (rCCR7) plus green fluorescence protein (GFP) or GFP alone. CCR7 mRNA and cell surface expression of rCCR7-hASCs and GFP-hASCs were examined by reverse transcription-polymerase chain reaction (RT-PCR) and flow cytometry (FCM), respectively. The phenotype, differentiation, and proliferation capacity of each cell type was also determined. To examine migration, rCCR7-hASCs and GFP-hASCs were injected intravenously into Lewis rats, and the proportion of GFP-positive cells in the spleen and lymph nodes was determined with FCM. Results mRNA and cell surface protein expression of CCR7 was essentially undetectable in hASCs and GFP-ASCs; however, CCR7 was highly expressed in rCCR7-ASCs. rCCR7-hASCs, GFP-hASCs, and hASCs shared a similar immunophenotype, and maintained the ability of multilineage differentiation and proliferation. In addition, the average proportion of GFP-positive cells was significantly higher following transplantation of rCCR7-hASCs compared with GFP-hASCs (p<0.01). Conclusions These results suggest that upregulation of rat CCR7 expression does not change the phenotype, differentiation, or proliferation capacity of hASCs, but does enable efficient migration of hASCs to rat SLOs.
Collapse
Affiliation(s)
- Tian Ma
- Department of Plastic and Reconstruction, Chinese PLA General Hospital, Beijing, China (mainland)
| | - Shao-Liang Luan
- Department of Vascular and Endovascular, Chinese PLA General Hospital, Beijing, China (mainland)
| | - Hong Huang
- Institute of Basic Medicine, Chinese PLA General Hospital, Beijing, China (mainland)
| | - Xing-Kun Sun
- Department of Stomatology, General Hospital of Chinese People's Armed Police Forces, Beijing, China (mainland)
| | - Yan-Mei Yang
- Department of Stomatology, Chinese PLA General Hospital, Beijing, China (mainland)
| | - Hui Zhang
- Department of Plastic Surgery, The Second Hospital of ShanXi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Wei-Dong Han
- Department of Molecular Biology, Chinese PLA General Hospital, Beijing, China (mainland)
| | - Hong Li
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Beijing, China (mainland)
| | - Yan Han
- Department of Plastic and Reconstruction, Chinese PLA General Hospital, Beijing, China (mainland)
| |
Collapse
|
33
|
Jain NG, Wong EA, Aranyosi AJ, Boneschansker L, Markmann JF, Briscoe DM, Irimia D. Microfluidic mazes to characterize T-cell exploration patterns following activation in vitro. Integr Biol (Camb) 2016; 7:1423-31. [PMID: 26325525 DOI: 10.1039/c5ib00146c] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The migration of T-cell subsets within peripheral tissues is characteristic of inflammation and immunoregulation. In general, the lymphocyte migratory response is assumed directional and guided by local gradients of chemoattractants and/or chemorepellents. However, little is known about how cells explore their tissue environment, and whether lymphocyte activation may influence speed and exploratory patterns of migration. To probe migration patterns by T-cells we designed a microfluidic maze device that replicates critical features of a tissue-like microenvironment. We quantified the migration patterns of unstimulated and mitogen-activated human T-cells at single cell resolution and found significant differences in exploration within microfluidic mazes. While unstimulated lymphocytes migrated in a directed manner, activated T-cells migrated through large areas of the mazes in an exploratory pattern in response to the chemoattractants RANTES (CCL5) and IP-10 (CXCL10). The analysis of migration enabled by the microfluidic devices help develop new methods for determining how human circulating T-cells function in vivo to seek out antigens in health and disease states.
Collapse
Affiliation(s)
- Namrata G Jain
- BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Boston, MA 02129, USA and Transplant Research Program, Boston Children's Hospital, Boston, MA 02115, USA.
| | - Elisabeth A Wong
- BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Alexander J Aranyosi
- BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Leo Boneschansker
- BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Boston, MA 02129, USA and Transplant Research Program, Boston Children's Hospital, Boston, MA 02115, USA.
| | - James F Markmann
- BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Boston, MA 02129, USA and Division of Nephrology, Department of Medicine, Boston Children's Hospital, 300 Longwood Ave, MA 02139, USA
| | - David M Briscoe
- Transplant Research Program, Boston Children's Hospital, Boston, MA 02115, USA. and Division of Nephrology, Department of Medicine, Boston Children's Hospital, 300 Longwood Ave, MA 02139, USA
| | - Daniel Irimia
- BioMEMS Resource Center, Department of Surgery, Massachusetts General Hospital, Boston, MA 02129, USA and Harvard Medical School, Boston, MA 02129, USA.
| |
Collapse
|
34
|
Adriani G, Pavesi A, Tan AT, Bertoletti A, Thiery JP, Kamm RD. Microfluidic models for adoptive cell-mediated cancer immunotherapies. Drug Discov Today 2016; 21:1472-1478. [PMID: 27185084 PMCID: PMC5035566 DOI: 10.1016/j.drudis.2016.05.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/07/2016] [Accepted: 05/09/2016] [Indexed: 01/02/2023]
Abstract
Current adoptive T cell therapies have shown promising results in clinical trials but need further development as an effective cancer treatment. Here, we discuss how 3D microfluidic tumour models mimicking the tumour microenvironment could help in testing T cell immunotherapies by assessing engineered T cells and identifying combinatorial therapy to improve therapeutic efficacy. We propose that 3D microfluidic systems can be used to screen different patient-specific treatments, thereby reducing the burden of in vivo testing and facilitating the rapid translation of successful T cell cancer immunotherapies to the clinic.
Collapse
Affiliation(s)
- Giulia Adriani
- Singapore-MIT Alliance for Research and Technology, BioSyM IRG, 1 Create Way, 138602, Singapore
| | - Andrea Pavesi
- Singapore-MIT Alliance for Research and Technology, BioSyM IRG, 1 Create Way, 138602, Singapore
| | - Anthony T Tan
- DUKE-NUS Graduate Medical School Singapore, Emerging Infectious Disease Program, 8 College Road, 169857, Singapore
| | - Antonio Bertoletti
- DUKE-NUS Graduate Medical School Singapore, Emerging Infectious Disease Program, 8 College Road, 169857, Singapore
| | - Jean Paul Thiery
- National University of Singapore, Department of Biochemistry, Yong Loo Lin School of Medicine MD7, 8 Medical Drive, 117597, Singapore
| | - Roger D Kamm
- Singapore-MIT Alliance for Research and Technology, BioSyM IRG, 1 Create Way, 138602, Singapore; Massachusetts Institute of Technology, Department of Biological Engineering, 77 Massachusetts Avenue, 02139 Cambridge, MA, USA.
| |
Collapse
|
35
|
Kleist AB, Getschman AE, Ziarek JJ, Nevins AM, Gauthier PA, Chevigné A, Szpakowska M, Volkman BF. New paradigms in chemokine receptor signal transduction: Moving beyond the two-site model. Biochem Pharmacol 2016; 114:53-68. [PMID: 27106080 DOI: 10.1016/j.bcp.2016.04.007] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 04/13/2016] [Indexed: 10/21/2022]
Abstract
Chemokine receptor (CKR) signaling forms the basis of essential immune cellular functions, and dysregulated CKR signaling underpins numerous disease processes of the immune system and beyond. CKRs, which belong to the seven transmembrane domain receptor (7TMR) superfamily, initiate signaling upon binding of endogenous, secreted chemokine ligands. Chemokine-CKR interactions are traditionally described by a two-step/two-site mechanism, in which the CKR N-terminus recognizes the chemokine globular core (i.e. site 1 interaction), followed by activation when the unstructured chemokine N-terminus is inserted into the receptor TM bundle (i.e. site 2 interaction). Several recent studies challenge the structural independence of sites 1 and 2 by demonstrating physical and allosteric links between these supposedly separate sites. Others contest the functional independence of these sites, identifying nuanced roles for site 1 and other interactions in CKR activation. These developments emerge within a rapidly changing landscape in which CKR signaling is influenced by receptor PTMs, chemokine and CKR dimerization, and endogenous non-chemokine ligands. Simultaneous advances in the structural and functional characterization of 7TMR biased signaling have altered how we understand promiscuous chemokine-CKR interactions. In this review, we explore new paradigms in CKR signal transduction by considering studies that depict a more intricate architecture governing the consequences of chemokine-CKR interactions.
Collapse
Affiliation(s)
- Andrew B Kleist
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Anthony E Getschman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Joshua J Ziarek
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Ave, Boston, MA 02115, USA.
| | - Amanda M Nevins
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Pierre-Arnaud Gauthier
- Department of Infection and Immunity, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg.
| | - Andy Chevigné
- Department of Infection and Immunity, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg.
| | - Martyna Szpakowska
- Department of Infection and Immunity, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg.
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
36
|
Levin D, Forrest S, Banerjee S, Clay C, Cannon J, Moses M, Koster F. A spatial model of the efficiency of T cell search in the influenza-infected lung. J Theor Biol 2016; 398:52-63. [PMID: 26920246 DOI: 10.1016/j.jtbi.2016.02.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 02/08/2016] [Accepted: 02/12/2016] [Indexed: 11/16/2022]
Abstract
Emerging strains of influenza, such as avian H5N1 and 2009 pandemic H1N1, are more virulent than seasonal H1N1 influenza, yet the underlying mechanisms for these differences are not well understood. Subtle differences in how a given strain interacts with the immune system are likely a key factor in determining virulence. One aspect of the interaction is the ability of T cells to locate the foci of the infection in time to prevent uncontrolled expansion. Here, we develop an agent based spatial model to focus on T cell migration from lymph nodes through the vascular system to sites of infection. We use our model to investigate whether different strains of influenza modulate this process. We calibrate the model using viral and chemokine secretion rates we measure in vitro together with values taken from literature. The spatial nature of the model reveals unique challenges for T cell recruitment that are not apparent in standard differential equation models. In this model comparing three influenza viruses, plaque expansion is governed primarily by the replication rate of the virus strain, and the efficiency of the T cell search-and-kill is limited by the density of infected epithelial cells in each plaque. Thus for each virus there is a different threshold of T cell search time above which recruited T cells are unable to control further expansion. Future models could use this relationship to more accurately predict control of the infection.
Collapse
Affiliation(s)
- Drew Levin
- Department of Computer Science, University of New Mexico, Albuquerque, NM, USA.
| | - Stephanie Forrest
- Department of Computer Science, University of New Mexico, Albuquerque, NM, USA
| | - Soumya Banerjee
- Department of Computer Science, University of New Mexico, Albuquerque, NM, USA
| | - Candice Clay
- Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| | - Judy Cannon
- Department of Molecular Genetics & Microbiology, Department of Pathology, University of New Mexico, Health Sciences Center, Albuquerque, NM, USA
| | - Melanie Moses
- Department of Computer Science, University of New Mexico, Albuquerque, NM, USA
| | - Frederick Koster
- Department of Computer Science, University of New Mexico, Albuquerque, NM, USA; Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| |
Collapse
|
37
|
Vacchini A, Busnelli M, Chini B, Locati M, Borroni EM. Analysis of G Protein and β-Arrestin Activation in Chemokine Receptors Signaling. Methods Enzymol 2016; 570:421-40. [DOI: 10.1016/bs.mie.2015.09.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
38
|
Karin N, Wildbaum G, Thelen M. Biased signaling pathways via CXCR3 control the development and function of CD4+ T cell subsets. J Leukoc Biol 2015; 99:857-62. [PMID: 26657511 DOI: 10.1189/jlb.2mr0915-441r] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 11/03/2015] [Indexed: 12/17/2022] Open
Abstract
Structurally related chemotactic cytokines (chemokines) regulate cell trafficking through interactions with 7-transmembrane domain, G protein-coupled receptors. Biased signaling or functional selectivity is a concept that describes a situation where a 7-transmembrane domain receptor preferentially activates one of several available cellular signaling pathways. It can be divided into 3 distinct cases: ligand bias, receptor bias, and tissue or cell bias. Many studies, including those coming from our lab, have shown that only a limited number of chemokines are key drivers of inflammation. We have referred to them as "driver chemokines." They include the CXCR3 ligands CXCL9 and CXCL10, the CCR2 ligand CCL2, all 3 CCR5 ligands, and the CCR9 ligand CCL25. As for CXCR3, despite the proinflammatory nature of CXCL10 and CXCL9, transgenic mice lacking CXCR3 display an aggravated manifestation of different autoimmune disease, including Type I diabetes and experimental autoimmune encephalomyelitis. Recently, we showed that whereas CXCL9 and CXCL10 induce effector Th1/Th17 cells to promote inflammation, CXCL11, with a relatively higher binding affinity to CXCR3, drives the development of the forkhead box P3-negative IL-10(high) T regulatory 1 cell subset and hence, dampens inflammation. We also showed that CXCL9/CXCL10 activates a different signaling cascade than CXCL11, despite binding to the same receptor, CXCR3, which results in these diverse biologic activities. This provides new evidence for the role of biased signaling in regulating biologic activities, in which CXCL11 induces ligand bias at CXCR3 and receptor-biased signaling via atypical chemokine receptor 3.
Collapse
Affiliation(s)
- Nathan Karin
- Department of Immunology, Rappaport Family Institute for Research in the Medical Sciences and Bruce Rappaport Faculty of Medicine, Haifa, Israel; and
| | - Gizi Wildbaum
- Department of Immunology, Rappaport Family Institute for Research in the Medical Sciences and Bruce Rappaport Faculty of Medicine, Haifa, Israel; and
| | - Marcus Thelen
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| |
Collapse
|
39
|
Amarandi RM, Hjortø GM, Rosenkilde MM, Karlshøj S. Probing Biased Signaling in Chemokine Receptors. Methods Enzymol 2015; 570:155-86. [PMID: 26921946 DOI: 10.1016/bs.mie.2015.09.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The chemokine system mediates leukocyte migration during homeostatic and inflammatory processes. Traditionally, it is described as redundant and promiscuous, with a single chemokine ligand binding to different receptors and a single receptor having several ligands. Signaling of chemokine receptors occurs via two major routes, G protein- and β-arrestin-dependent, which can be preferentially modulated depending on the ligands or receptors involved, as well as the cell types or tissues in which the signaling event occurs. The preferential activation of a certain signaling pathway to the detriment of others has been termed signaling bias and can accordingly be grouped into ligand bias, receptor bias, and tissue bias. Bias has so far been broadly overlooked in the process of drug development. The low number of currently approved drugs targeting the chemokine system, as well as the broad range of failed clinical trials, reflects the need for a better understanding of the chemokine system. Thus, understanding the character, direction, and consequence of biased signaling in the chemokine system may aid the development of new therapeutics. This review describes experiments to assess G protein-dependent and -independent signaling in order to quantify chemokine system bias.
Collapse
Affiliation(s)
- Roxana-Maria Amarandi
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark; Faculty of Chemistry, Alexandru Ioan Cuza University of Iaşi, Iaşi, Romania
| | - Gertrud Malene Hjortø
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Mette Marie Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Stefanie Karlshøj
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
40
|
Dominguez GA, Hammer DA. Effect of adhesion and chemokine presentation on T-lymphocyte haptokinesis. Integr Biol (Camb) 2015; 6:862-73. [PMID: 25012074 DOI: 10.1039/c4ib00094c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Motility is critical for the function of T-lymphocytes. Motility in T-lymphocytes is driven by the occupancy of chemokine receptors by chemokines, and modulated by adhesive interactions. However, it is not well understood how the combination of adhesion and chemokine binding affects T-lymphocyte migration. We used microcontact printing on polymeric substrates to measure how lymphocyte migration is quantitatively controlled by adhesion and chemokine ligation. Focusing only on random motion, we found that T-lymphocytes exhibit biphasic motility in response to the substrate concentration of either ICAM-1 or VCAM-1, and generally display more active motion on ICAM-1 surfaces. Furthermore, we examined how the combination of the homeostatic chemokines CCL19 and CCL21 contribute to motility. By themselves, CCL19 and CCL21, ligands for CCR7, elicit biphasic motility, but their combination synergistically increases CCR7 mediated chemokinesis on ICAM-1. By presenting CCL21 with ICAM-1 on the surface with soluble CCL19, we observed random motion that is greater than what is observed with soluble chemokines alone. These data suggest that ICAM-1 has a greater contribution to motility than VCAM-1 and that both adhesive interactions and chemokine ligation work in concert to control T-lymphocyte motility.
Collapse
Affiliation(s)
- George A Dominguez
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd St, Philadelphia, PA 19104, USA
| | | |
Collapse
|
41
|
Mehling M, Frank T, Albayrak C, Tay S. Real-time tracking, retrieval and gene expression analysis of migrating human T cells. LAB ON A CHIP 2015; 15:1276-83. [PMID: 25512266 DOI: 10.1039/c4lc01038h] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Dynamical analysis of single-cells allows assessment of the extent and role of cell-to-cell variability, however traditional dish-and-pipette techniques have hindered single-cell analysis in quantitative biology. We developed an automated microfluidic cell culture system that generates stable diffusion-based chemokine gradients, where cells can be placed in predetermined positions, monitored via single-cell time-lapse microscopy, and subsequently be retrieved based on their migration speed and directionality for further off-chip gene expression analysis, constituting a powerful platform for multiparameter quantitative studies of single-cell chemotaxis. Using this system we studied CXCL12-directed migration of individual human primary T cells. Spatiotemporally deterministic retrieval of T cell subsets in relation to their migration speed, and subsequent analysis with microfluidic droplet digital-PCR showed that the expression level of CXCR4 – the receptor of CXCL12 – underlies enhanced human T cell chemotaxis.
Collapse
Affiliation(s)
- Matthias Mehling
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, 4058 Basel, Switzerland.
| | | | | | | |
Collapse
|
42
|
Analysis of CCR7 mediated T cell transfectant migration using a microfluidic gradient generator. J Immunol Methods 2015; 419:9-17. [PMID: 25733353 DOI: 10.1016/j.jim.2015.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 02/07/2015] [Accepted: 02/18/2015] [Indexed: 12/11/2022]
Abstract
T lymphocyte migration is crucial for adaptive immunity. Manipulation of signaling molecules controlling cell migration combined with in-vitro cell migration analysis provides a powerful research approach. Microfluidic devices, which can precisely configure chemoattractant gradients and allow quantitative single cell analysis, have been increasingly applied to cell migration and chemotaxis studies. However, there are a very limited number of published studies involving microfluidic migration analysis of genetically manipulated immune cells. In this study, we describe a simple microfluidic method for quantitative analysis of T cells expressing transfected chemokine receptors and other cell migration signaling probes. Using this method, we demonstrated chemotaxis of Jurkat transfectants expressing wild-type or C-terminus mutated CCR7 within a gradient of chemokine CCL19, and characterized the difference in transfectant migration mediated by wild-type and mutant CCR7. The EGFP-tagged CCR7 allows identification of CCR7-expressing transfectants in cell migration analysis and microscopy assessment of CCR7 dynamics. Collectively, our study demonstrated the effective use of the microfluidic method for studying CCR7 mediated T cell transfectant migration. We envision this developed method will provide a useful platform to functionally test various signaling mechanisms at the cell migration level.
Collapse
|
43
|
Mahmood S, Nandagopal S, Sow I, Lin F, Kung SKP. Microfluidic-based, live-cell analysis allows assessment of NK-cell migration in response to crosstalk with dendritic cells. Eur J Immunol 2014; 44:2737-48. [DOI: 10.1002/eji.201344244] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 05/02/2014] [Accepted: 06/13/2014] [Indexed: 12/13/2022]
Affiliation(s)
- Sajid Mahmood
- Department of Immunology; University of Manitoba; Winnipeg MB Canada
| | - Saravanan Nandagopal
- Department of Immunology; University of Manitoba; Winnipeg MB Canada
- Department of Physics and Astronomy; University of Manitoba; Winnipeg MB Canada
- Department of Biosystems Engineering; University of Manitoba; Winnipeg MB Canada
| | - Ibrahim Sow
- Department of Immunology; University of Manitoba; Winnipeg MB Canada
| | - Francis Lin
- Department of Immunology; University of Manitoba; Winnipeg MB Canada
- Department of Physics and Astronomy; University of Manitoba; Winnipeg MB Canada
- Department of Biosystems Engineering; University of Manitoba; Winnipeg MB Canada
| | - Sam K. P. Kung
- Department of Immunology; University of Manitoba; Winnipeg MB Canada
| |
Collapse
|
44
|
Steen A, Larsen O, Thiele S, Rosenkilde MM. Biased and g protein-independent signaling of chemokine receptors. Front Immunol 2014; 5:277. [PMID: 25002861 PMCID: PMC4066200 DOI: 10.3389/fimmu.2014.00277] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 05/28/2014] [Indexed: 01/14/2023] Open
Abstract
Biased signaling or functional selectivity occurs when a 7TM-receptor preferentially activates one of several available pathways. It can be divided into three distinct forms: ligand bias, receptor bias, and tissue or cell bias, where it is mediated by different ligands (on the same receptor), different receptors (with the same ligand), or different tissues or cells (for the same ligand–receptor pair). Most often biased signaling is differentiated into G protein-dependent and β-arrestin-dependent signaling. Yet, it may also cover signaling differences within these groups. Moreover, it may not be absolute, i.e., full versus no activation. Here we discuss biased signaling in the chemokine system, including the structural basis for biased signaling in chemokine receptors, as well as in class A 7TM receptors in general. This includes overall helical movements and the contributions of micro-switches based on recently published 7TM crystals and molecular dynamics studies. All three forms of biased signaling are abundant in the chemokine system. This challenges our understanding of “classic” redundancy inevitably ascribed to this system, where multiple chemokines bind to the same receptor and where a single chemokine may bind to several receptors – in both cases with the same functional outcome. The ubiquitous biased signaling confers a hitherto unknown specificity to the chemokine system with a complex interaction pattern that is better described as promiscuous with context-defined roles and different functional outcomes in a ligand-, receptor-, or cell/tissue-defined manner. As the low number of successful drug development plans implies, there are great difficulties in targeting chemokine receptors; in particular with regard to receptor antagonists as anti-inflammatory drugs. Un-defined and putative non-selective targeting of the complete cellular signaling system could be the underlying cause of lack of success. Therefore, biased ligands could be the solution.
Collapse
Affiliation(s)
- Anne Steen
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Olav Larsen
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Stefanie Thiele
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Mette M Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
45
|
Junkin M, Tay S. Microfluidic single-cell analysis for systems immunology. LAB ON A CHIP 2014; 14:1246-60. [PMID: 24503696 DOI: 10.1039/c3lc51182k] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
The immune system constantly battles infection and tissue damage, but exaggerated immune responses lead to allergies, autoimmunity and cancer. Discrimination of self from foreign and the fine-tuning of immunity are achieved by information processing pathways, whose regulatory mechanisms are little understood. Cell-to-cell variability and stochastic molecular interactions result in diverse cellular responses to identical signaling inputs, casting doubt on the reliability of traditional population-averaged analyses. Furthermore, dynamic molecular and cellular interactions create emergent properties that change over multiple time scales. Understanding immunity in the face of complexity and noisy dynamics requires time-dependent analysis of single-cells in a proper context. Microfluidic systems create precisely defined microenvironments by controlling fluidic and surface chemistries, feature sizes, geometries and signal input timing, and thus enable quantitative multi-parameter analysis of single cells. Such qualities allow observable dynamic environments approaching in vivo levels of biological complexity. Seamless parallelization of functional units in microfluidic devices allows high-throughput measurements, an essential feature for statistically meaningful analysis of naturally variable biological systems. These abilities recapitulate diverse scenarios such as cell-cell signaling, migration, differentiation, antibody and cytokine production, clonal selection, and cell lysis, thereby enabling accurate and meaningful study of immune behaviors in vitro.
Collapse
Affiliation(s)
- Michael Junkin
- Department of Biosystems Science and Engineering, ETH Zürich, Switzerland.
| | | |
Collapse
|
46
|
Microfluidics and its applications in quantitative biology. QUANTITATIVE BIOLOGY 2013. [DOI: 10.1007/s40484-014-0024-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
47
|
Miller FW, Cooper RG, Vencovsky J, Rider LG, Danko K, Wedderburn LR, Lundberg IE, Pachman LM, Reed AM, Ytterberg SR, Padyukov L, Selva-O’Callaghan A, Radstake T, Isenberg DA, Chinoy H, Ollier WER, O’Hanlon TP, Peng B, Lee A, Lamb JA, Chen W, Amos CI, Gregersen PK. Genome-wide association study of dermatomyositis reveals genetic overlap with other autoimmune disorders. ARTHRITIS AND RHEUMATISM 2013; 65:3239-47. [PMID: 23983088 PMCID: PMC3934004 DOI: 10.1002/art.38137] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 08/13/2013] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To identify new genetic associations with juvenile and adult dermatomyositis (DM). METHODS We performed a genome-wide association study (GWAS) of adult and juvenile DM patients of European ancestry (n = 1,178) and controls (n = 4,724). To assess genetic overlap with other autoimmune disorders, we examined whether 141 single-nucleotide polymorphisms (SNPs) outside the major histocompatibility complex (MHC) locus, and previously associated with autoimmune diseases, predispose to DM. RESULTS Compared to controls, patients with DM had a strong signal in the MHC region consisting of GWAS-level significance (P < 5 × 10(-8)) at 80 genotyped SNPs. An analysis of 141 non-MHC SNPs previously associated with autoimmune diseases showed that 3 SNPs linked with 3 genes were associated with DM, with a false discovery rate (FDR) of <0.05. These genes were phospholipase C-like 1 (PLCL1; rs6738825, FDR = 0.00089), B lymphoid tyrosine kinase (BLK; rs2736340, FDR = 0.0031), and chemokine (C-C motif) ligand 21 (CCL21; rs951005, FDR = 0.0076). None of these genes was previously reported to be associated with DM. CONCLUSION Our findings confirm the MHC as the major genetic region associated with DM and indicate that DM shares non-MHC genetic features with other autoimmune diseases, suggesting the presence of additional novel risk loci. This first identification of autoimmune disease genetic predispositions shared with DM may lead to enhanced understanding of pathogenesis and novel diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Frederick W. Miller
- National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, Maryland
| | - Robert G. Cooper
- The University of Manchester Rheumatic Diseases Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom
| | | | - Lisa G. Rider
- National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, Maryland
| | | | | | - Ingrid E. Lundberg
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Lauren M. Pachman
- Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | | | | | - Leonid Padyukov
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| | | | - Timothy Radstake
- Utrecht University Medical Center, Dept. of Rheumatology and Clinical Immunology; Laboratory for Translational Immunology; and Nijmegen Center for Molecular Life Sciences, Nijmegen, The Netherlands
| | | | - Hector Chinoy
- Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - William E. R. Ollier
- Centre for Integrated Genomic Medical Research, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Terrance P. O’Hanlon
- National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, Maryland
| | - Bo Peng
- M.D. Anderson Cancer Center, Houston, Texas
| | - Annette Lee
- Feinstein Institute for Medical Research, Manhasset, New York
| | - Janine A. Lamb
- Centre for Integrated Genomic Medical Research, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Wei Chen
- M.D. Anderson Cancer Center, Houston, Texas
| | | | | | | |
Collapse
|
48
|
Lara Rodriguez L, Schneider IC. Directed cell migration in multi-cue environments. Integr Biol (Camb) 2013; 5:1306-23. [DOI: 10.1039/c3ib40137e] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
| | - Ian C. Schneider
- Department of Chemical and Biological Engineering, Iowa State University, USA
- Department of Genetics, Development and Cell Biology, Iowa State University, USA
| |
Collapse
|
49
|
Wu J, Wu X, Lin F. Recent developments in microfluidics-based chemotaxis studies. LAB ON A CHIP 2013; 13:2484-99. [PMID: 23712326 DOI: 10.1039/c3lc50415h] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Microfluidic devices can better control cellular microenvironments compared to conventional cell migration assays. Over the past few years, microfluidics-based chemotaxis studies showed a rapid growth. New strategies were developed to explore cell migration in manipulated chemical gradients. In addition to expanding the use of microfluidic devices for a broader range of cell types, microfluidic devices were used to study cell migration and chemotaxis in complex environments. Furthermore, high-throughput microfluidic chemotaxis devices and integrated microfluidic chemotaxis systems were developed for medical and commercial applications. In this article, we review recent developments in microfluidics-based chemotaxis studies and discuss the new trends in this field observed over the past few years.
Collapse
Affiliation(s)
- Jiandong Wu
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | | | | |
Collapse
|
50
|
Wu D, Joyee AG, Nandagopal S, Lopez M, Ma X, Berry J, Lin F. Effects of Clostridium difficile toxin A and B on human T lymphocyte migration. Toxins (Basel) 2013; 5:926-38. [PMID: 23645153 PMCID: PMC3709270 DOI: 10.3390/toxins5050926] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Revised: 03/27/2013] [Accepted: 04/23/2013] [Indexed: 01/24/2023] Open
Abstract
Bacterial products such as toxins can interfere with a variety of cellular processes, leading to severe human diseases. Clostridium difficile toxins, TcdA and TcdB are the primary contributing factors to the pathogenesis of C. difficile-associated diseases (CDAD). While the mechanisms for TcdA and TcdB mediated cellular responses are complex, it has been shown that these toxins can alter chemotactic responses of neutrophils and intestinal epithelial cells leading to innate immune responses and tissue damages. The effects of C. difficile toxins on the migration and trafficking of other leukocyte subsets, such as T lymphocytes, are not clear and may have potential implications for adaptive immunity. We investigated here the direct and indirect effects of TcdA and TcdB on the migration of human blood T cells using conventional cell migration assays and microfluidic devices. It has been found that, although both toxins decrease T cell motility, only TcdA but not TcdB decreases T cell chemotaxis. Similar effects are observed in T cell migration toward the TcdA- or TcdB-treated human epithelial cells. Our study demonstrated the primary role of TcdA (compared to TcdB) in altering T cell migration and chemotaxis, suggesting possible implications for C. difficile toxin mediated adaptive immune responses in CDAD.
Collapse
Affiliation(s)
- Dan Wu
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; E-Mails: (D.W.); (S.N.); (X.M.)
| | - Antony George Joyee
- Cangene Corporation, Winnipeg, MB R3T 2N2, Canada; E-Mails: (A.G.J.); (J.B.)
- Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Saravanan Nandagopal
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; E-Mails: (D.W.); (S.N.); (X.M.)
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Marianela Lopez
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; E-Mail:
| | - Xiuli Ma
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; E-Mails: (D.W.); (S.N.); (X.M.)
| | - Jody Berry
- Cangene Corporation, Winnipeg, MB R3T 2N2, Canada; E-Mails: (A.G.J.); (J.B.)
- BD Biosciences, La Jolla, CA 92121, USA
| | - Francis Lin
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; E-Mails: (D.W.); (S.N.); (X.M.)
- Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-204-474-9895; Fax: +1-204-474-7622
| |
Collapse
|