51
|
Liu Z, Wu X, Tian Y, Zhang W, Qiao S, Xu W, Liu Y, Wang S. H. pylori infection induces CXCL8 expression and promotes gastric cancer progress through downregulating KLF4. Mol Carcinog 2021; 60:524-537. [PMID: 34038586 DOI: 10.1002/mc.23309] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/07/2021] [Accepted: 05/09/2021] [Indexed: 12/24/2022]
Abstract
Tumour-derived CXCL8 facilitates the movement of myeloid-derived suppressor cells, which are able to restrain antitumour immune responses to the tumour microenvironment. Kruppel-like factor 4 (KLF4) is a potential tumour suppressor in gastric cancer (GC). However, knowledge regarding correlations between KLF4 and CXCL8 in GC is limited. We use cellular and molecular biological methods to assess whether these two factors interact in GC. Expression CXCL8 and KLF4 was altered in human GC tissues compared to normal gastric tissues in opposite ways. Additionally, cytotoxin-associated gene A protein (CagA) gene transduction or Helicobacter pylori (H. pylori) infection upregulated CXCL8 expression. Knockdown of KLF4 expression increased CXCL8 protein and RNA expression, whereas its overexpression had the opposite effect. CXCL8-mediated enhancement of GC cell migration and proliferation was reversed by upregulation of KLF4 expression. Further mechanistic research revealed that KLF4 binds the CXCL8 promoter, suppressing CXCL8 transcription. Moreover, CXCL8 stimulation reduced KLF4 protein expression and promoted GC cell proliferation and migration, eventually promoting neoplasm growth in vivo. Together, our findings demonstrate that CagA promotes CXCL8 and inhibits KLF4. CXCL8 is a decisive downstream target gene of KLF4, and KLF4 negatively regulates CXCL8 in GC. Furthermore, CXCL8's negative regulation of KLF4 in vivo and in vitro, indicates that CagA may downregulate KLF4 by inducing CXCL8 expression, low expression of KLF4 further promotes that of CXCL8, forming a vicious circle in GC. Targeted KLF4 activation might improve the immunosuppressive microenvironment through direct negative regulation of CXCL8, providing a new potential target to strengthen the efficacy of immunotherapy in GC patients.
Collapse
Affiliation(s)
- Zhengxia Liu
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Xiao Wu
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Yuanyuan Tian
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Wanchun Zhang
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Siyuan Qiao
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Wenting Xu
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Yakun Liu
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Siying Wang
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
52
|
Serum levels of CC chemokine ligands in cutaneous leishmaniasis patients. J Parasit Dis 2021; 45:153-158. [PMID: 33746400 DOI: 10.1007/s12639-020-01290-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 10/05/2020] [Indexed: 10/23/2022] Open
Abstract
The crucial functions of chemokine/receptors in numerous parasitic infections, including leishmaniasis, are well documented. This study aimed to assess the serum levels of CC ligand (CCL) 2, CCL5, and CCL11 in cutaneous leishmaniasis (CL) patients. 64 patients, suffering from CL and 100 healthy people were selected, and their blood serum concentrations of CCL2, CCL5, and CCL11 were measured using enzyme-linked immunosorbent assay. The results demonstrated that while the mean serum levels of CCL5 and CCL11 increased significantly in CL patients, the mean serum levels of CCL2 decreased, compared to the control group. Despite the sufficient production of CCL5 and CCL11 in CL patients, they suffered from CCL2 deficiency, as the defense mechanism against parasitic infection. These findings suggest a mechanism that might partially explain the patients' susceptibility to persistent infection and their inability to clear the parasites.
Collapse
|
53
|
Cevaal PM, Ali A, Czuba-Wojnilowicz E, Symons J, Lewin SR, Cortez-Jugo C, Caruso F. In Vivo T Cell-Targeting Nanoparticle Drug Delivery Systems: Considerations for Rational Design. ACS NANO 2021; 15:3736-3753. [PMID: 33600163 DOI: 10.1021/acsnano.0c09514] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
T cells play an important role in immunity and repair and are implicated in diseases, including blood cancers, viral infections, and inflammation, making them attractive targets for the treatment and prevention of diseases. Over recent years, the advent of nanomedicine has shown an increase in studies that use nanoparticles as carriers to deliver therapeutic cargo to T cells for ex vivo and in vivo applications. Nanoparticle-based delivery has several advantages, including the ability to load and protect a variety of drugs, control drug release, improve drug pharmacokinetics and biodistribution, and site- or cell-specific targeting. However, the delivery of nanoparticles to T cells remains a major technological challenge, which is primarily due to the nonphagocytic nature of T cells. In this review, we discuss the physiological barriers to effective T cell targeting and describe the different approaches used to deliver cargo-loaded nanoparticles to T cells for the treatment of disease such as T cell lymphoma and human immunodeficiency virus (HIV). In particular, engineering strategies that aim to improve nanoparticle internalization by T cells, including ligand-based targeting, will be highlighted. These nanoparticle engineering approaches are expected to inspire the development of effective nanomaterials that can target or manipulate the function of T cells for the treatment of T cell-related diseases.
Collapse
Affiliation(s)
| | | | - Ewa Czuba-Wojnilowicz
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | | | - Sharon R Lewin
- Victorian Infectious Diseases, Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Victoria 3004, Australia
| | - Christina Cortez-Jugo
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
54
|
Monocyte subset redistribution from blood to kidneys in patients with Puumala virus caused hemorrhagic fever with renal syndrome. PLoS Pathog 2021; 17:e1009400. [PMID: 33690725 PMCID: PMC7984619 DOI: 10.1371/journal.ppat.1009400] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 03/22/2021] [Accepted: 02/17/2021] [Indexed: 12/18/2022] Open
Abstract
Innate immune cells like monocytes patrol the vasculature and mucosal surfaces, recognize pathogens, rapidly redistribute to affected tissues and cause inflammation by secretion of cytokines. We previously showed that monocytes are reduced in blood but accumulate in the airways of patients with Puumala virus (PUUV) caused hemorrhagic fever with renal syndrome (HFRS). However, the dynamics of monocyte infiltration to the kidneys during HFRS, and its impact on disease severity are currently unknown. Here, we examined longitudinal peripheral blood samples and renal biopsies from HFRS patients and performed in vitro experiments to investigate the fate of monocytes during HFRS. During the early stages of HFRS, circulating CD14-CD16+ nonclassical monocytes (NCMs) that patrol the vasculature were reduced in most patients. Instead, CD14+CD16- classical (CMs) and CD14+CD16+ intermediate monocytes (IMs) were increased in blood, in particular in HFRS patients with more severe disease. Blood monocytes from patients with acute HFRS expressed higher levels of HLA-DR, the endothelial adhesion marker CD62L and the chemokine receptors CCR7 and CCR2, as compared to convalescence, suggesting monocyte activation and migration to peripheral tissues during acute HFRS. Supporting this hypothesis, increased numbers of HLA-DR+, CD14+, CD16+ and CD68+ cells were observed in the renal tissues of acute HFRS patients compared to controls. In vitro, blood CD16+ monocytes upregulated CD62L after direct exposure to PUUV whereas CD16- monocytes upregulated CCR7 after contact with PUUV-infected endothelial cells, suggesting differential mechanisms of activation and response between monocyte subsets. Together, our findings suggest that NCMs are reduced in blood, potentially via CD62L-mediated attachment to endothelial cells and monocytes are recruited to the kidneys during HFRS. Monocyte mobilization, activation and functional impairment together may influence the severity of disease in acute PUUV-HFRS.
Collapse
|
55
|
Jalalpour S, Mirzaee V, Taheri M, Fathollahi MS, Khorramdelazadeh H, Jafarzadeh A. THE H. PYLORI-RELATED VIRULENCE FACTOR CAGA INFLUENCES THE EXPRESSION OF CHEMOKINES CXCL10, CCL17, CCL20, CCL22, AND THEIR RECEPTORS BY PERIPHERAL BLOOD MONONUCLEAR CELLS FROM PEPTIC ULCER PATIENTS. ARQUIVOS DE GASTROENTEROLOGIA 2021; 57:366-374. [PMID: 33331471 DOI: 10.1590/s0004-2803.202000000-70] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 08/17/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND During the Helicobacter pylori (HP) infection, the infiltration of the leukocytes into stomach mucosa is directed by locally produced chemokines that play a decisive role in infection outcome. The CagA is the most potent virulence factor of HP, so that the infection with CagA + strains is associated with more severe complications than infection with CagA - HP. OBJECTIVE The aim was to determine the expression of chemokines CXCL10, CCL17, CCL20 and CCL22, and their receptors by CagA + HP- and CagA - HP-derived crude extract (HP-CE)-stimulated peripheral blood mononuclear cells (PBMCs) from peptic ulcer (PU) patients. METHODS The serum and the PBMCs were collected from 20 HP-infected PU patients, 20 HP-infected asymptomatic subjects (HIA) and 20 non-infected healthy subjects (NHS). The PBMCs were cultured in absence of stimulator or with 10 µg CagA + HP crude extract (CagA + CE), 10 µg CagA - HP crude extract (CagA - CE). Chemokines and receptors were measured by ELISA and real time-PCR respectively. RESULTS In PU patients, the production of chemokines CXCL10, CCL17, CCL20 and CCL22, and the expression of chemokine receptors CXCR3, CCR4 and CCR6 by CagA + CE-induced PBMCs were significantly higher than non-stimulated and CagA - CE stimulated cultures. The CXCL10 production by CagA + CE stimulated PBMCs from HIA subjects was significantly higher than the equal cultures from PU and NHS groups. The CCL17 and the CCL20 production by non-stimulated, CagA + CE stimulated, and CagA - CE stimulated PBMCs from PU subjects were significantly higher than the equal cultures from NHS and HIA groups. The CCL22 production by non-stimulated, CagA + CE stimulated and CagA - CE stimulated PBMCs from NHS group were significantly higher than the equal cultures from HIA and PU groups. The CagA + CE stimulated PBMCs from HIA subjects expressed lower amounts of CCR6 in comparison with CagA + CE stimulated PBMCs from NHS and PU groups. The serum levels CXCL10 and CCL20 in PU and HIA groups were significantly higher than NHS subjects. NHS and HIA groups displayed higher serum levels of CCL22 in comparison with PU patients. CONCLUSION Results indicated that the CagA status of bacterium influence the expression of chemokines and receptors by HP-CE stimulated PBMCs from PU patients.
Collapse
Affiliation(s)
- Shila Jalalpour
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Vahid Mirzaee
- Department of Internal Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Taheri
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahmood Sheikh Fathollahi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hossain Khorramdelazadeh
- Molecular Medicine 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
| | - Abdollah Jafarzadeh
- Molecular Medicine 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.,Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.,Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
56
|
|
57
|
Sier VQ, de Vries MR, van der Vorst JR, Vahrmeijer AL, van Kooten C, Cruz LJ, de Geus-Oei LF, Ferreira V, Sier CFM, Alves F, Muthana M. Cell-Based Tracers as Trojan Horses for Image-Guided Surgery. Int J Mol Sci 2021; 22:E755. [PMID: 33451116 PMCID: PMC7828607 DOI: 10.3390/ijms22020755] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 12/29/2020] [Accepted: 12/30/2020] [Indexed: 12/14/2022] Open
Abstract
Surgeons rely almost completely on their own vision and palpation to recognize affected tissues during surgery. Consequently, they are often unable to distinguish between different cells and tissue types. This makes accurate and complete resection cumbersome. Targeted image-guided surgery (IGS) provides a solution by enabling real-time tissue recognition. Most current targeting agents (tracers) consist of antibodies or peptides equipped with a radiolabel for Positron Emission Tomography (PET) and Single Photon Emission Computed Tomography (SPECT), magnetic resonance imaging (MRI) labels, or a near-infrared fluorescent (NIRF) dye. These tracers are preoperatively administered to patients, home in on targeted cells or tissues, and are visualized in the operating room via dedicated imaging systems. Instead of using these 'passive' tracers, there are other, more 'active' approaches of probe delivery conceivable by using living cells (macrophages/monocytes, neutrophils, T cells, mesenchymal stromal cells), cell(-derived) fragments (platelets, extracellular vesicles (exosomes)), and microorganisms (bacteria, viruses) or, alternatively, 'humanized' nanoparticles. Compared with current tracers, these active contrast agents might be more efficient for the specific targeting of tumors or other pathological tissues (e.g., atherosclerotic plaques). This review provides an overview of the arsenal of possibilities applicable for the concept of cell-based tracers for IGS.
Collapse
Affiliation(s)
- Vincent Q. Sier
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (M.R.d.V.); (J.R.v.d.V.); (A.L.V.)
| | - Margreet R. de Vries
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (M.R.d.V.); (J.R.v.d.V.); (A.L.V.)
| | - Joost R. van der Vorst
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (M.R.d.V.); (J.R.v.d.V.); (A.L.V.)
| | - Alexander L. Vahrmeijer
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (M.R.d.V.); (J.R.v.d.V.); (A.L.V.)
| | - Cornelis van Kooten
- Department of Nephrology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Luis J. Cruz
- Department of Radiology, Translational Nanomaterials and Imaging Group, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
- Biomedical Photonic Imaging Group, University of Twente, 7522 NB Enschede, The Netherlands
| | - Valerie Ferreira
- Department of Research and Development, UniQure, 1100 DA Amsterdam, The Netherlands;
| | - Cornelis F. M. Sier
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (M.R.d.V.); (J.R.v.d.V.); (A.L.V.)
- Percuros B.V. Leiden, 2333 CL Leiden, The Netherlands
| | - Frauke Alves
- Translational Molecular Imaging, Clinic of Hematology and Medical Oncology, Institute of Diagnostic and Interventional Radiology, University Medicine Center Göttingen and Max-Planck-Institute for Experimental Medicine, 37075 Göttingen, Germany;
| | - Munitta Muthana
- Department of Infection and Immunity, University of Sheffield, Sheffield S10 2RX, UK;
| |
Collapse
|
58
|
Li N, Bai B, Zhang H, Zhang W, Tang S. Adipose stem cell secretion combined with biomaterials facilitates large-area wound healing. Regen Med 2020; 15:2311-2323. [PMID: 33320721 DOI: 10.2217/rme-2020-0086] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Adipose-derived stem cell (ADSC)-based therapeutic strategies are in fast-pace advancement in wound treatment due to their availability and the ability to self-renew, undergo multilineage differentiation and self-renewal. Existing studies have successfully explored ADSCs to facilitate scar-free healing of small wounds, but whether the healing of large-area wounds that exhibit over 50% of skin tissue loss in the entire body could be achieved remains controversial. This study sought to review the mechanism of physiological wound healing, and discuss the roles played by chemokines, biological factors and biomaterial scaffolds. The possibility of applying ADSC-conditioned medium or ADSC-released exosomes as 'off-the-shelf' tissue engineering products, integrated with biomaterial scaffolds to facilitate wound healing, was analyzed.
Collapse
Affiliation(s)
- Nan Li
- Institute of Plastic Surgery, Weifang Medical University, No. 4948, Shenglidong Street, Kuiwen District of Weifang City, Shandong Province, PR China
| | - Baoshuai Bai
- Institute of Plastic Surgery, Weifang Medical University, No. 4948, Shenglidong Street, Kuiwen District of Weifang City, Shandong Province, PR China
| | - Hairong Zhang
- Institute of Plastic Surgery, Weifang Medical University, No. 4948, Shenglidong Street, Kuiwen District of Weifang City, Shandong Province, PR China
| | - Wei Zhang
- Institute of Plastic Surgery, Weifang Medical University, No. 4948, Shenglidong Street, Kuiwen District of Weifang City, Shandong Province, PR China
| | - Shengjian Tang
- Institute of Plastic Surgery, Weifang Medical University, No. 4948, Shenglidong Street, Kuiwen District of Weifang City, Shandong Province, PR China
| |
Collapse
|
59
|
Schioppa T, Sozio F, Barbazza I, Scutera S, Bosisio D, Sozzani S, Del Prete A. Molecular Basis for CCRL2 Regulation of Leukocyte Migration. Front Cell Dev Biol 2020; 8:615031. [PMID: 33363177 PMCID: PMC7758318 DOI: 10.3389/fcell.2020.615031] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/23/2020] [Indexed: 12/27/2022] Open
Abstract
CCRL2 is a seven-transmembrane domain receptor that belongs to the chemokine receptor family. At difference from other members of this family, CCRL2 does not promote chemotaxis and shares structural features with atypical chemokine receptors (ACKRs). However, CCRL2 also differs from ACKRs since it does not bind chemokines and is devoid of scavenging functions. The only commonly recognized CCRL2 ligand is chemerin, a non-chemokine chemotactic protein. CCRL2 is expressed both by leukocytes and non-hematopoietic cells. The genetic ablation of CCRL2 has been instrumental to elucidate the role of this receptor as positive or negative regulator of inflammation. CCRL2 modulates leukocyte migration by two main mechanisms. First, when CCRL2 is expressed by barrier cells, such endothelial, and epithelial cells, it acts as a presenting molecule, contributing to the formation of a non-soluble chemotactic gradient for leukocytes expressing CMKLR1, the functional chemerin receptor. This mechanism was shown to be crucial in the induction of NK cell-dependent immune surveillance in lung cancer progression and metastasis. Second, by forming heterocomplexes with other chemokine receptors. For instance, CCRL2/CXCR2 heterodimers were shown to regulate the activation of β2-integrins in mouse neutrophils. This mini-review summarizes the current understanding of CCRL2 biology, based on experimental evidence obtained by the genetic deletion of this receptor in in vivo experimental models. Further studies are required to highlight the complex functional role of CCRL2 in different organs and pathological conditions.
Collapse
Affiliation(s)
- Tiziana Schioppa
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,Humanitas Clinical and Research Center Rozzano-Milano, Rozzano, Italy
| | - Francesca Sozio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,Humanitas Clinical and Research Center Rozzano-Milano, Rozzano, Italy
| | - Ilaria Barbazza
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sara Scutera
- Microbiology Section, Department of Public Health and Pediatric Sciences, University of Torino, Turin, Italy
| | - Daniela Bosisio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Silvano Sozzani
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Annalisa Del Prete
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,Humanitas Clinical and Research Center Rozzano-Milano, Rozzano, Italy
| |
Collapse
|
60
|
Nayak TK, Tilley DG. Recent Advances in GPCR-Regulated Leukocyte Responses during Acute Cardiac Injury. CURRENT OPINION IN PHYSIOLOGY 2020; 19:55-61. [PMID: 33244505 DOI: 10.1016/j.cophys.2020.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Following acute cardiac injury such as myocardial infarction (MI), the controlled activation and recruitment of various leukocytes to the site of tissue damage significantly impacts chronic changes to cardiac structure and function, and ultimately host survival. While recent research has focused primarily on how leukocytes respond to injury, understanding how to effectively modulate their responsiveness to dampen maladaptive inflammation and promote repair processes is not yet fully understood. The complex spatio-temporal migration and activation of leukocytes are largely controlled by various chemokines and their cognate receptors, belonging to the G protein-coupled receptor (GPCR) family. Beyond chemokine receptors, leukocytes express a host of additional GPCRs that have recently been shown to regulate their responsiveness to cardiac injury. In this minireview, we will briefly discuss the impact of chemokine receptors on leukocyte behaviour, with subsequent focus on the most recent advancements in understanding the impact and therapeutic potential of other GPCR classes on leukocyte responses after acute cardiac injury.
Collapse
Affiliation(s)
- Tapas K Nayak
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Douglas G Tilley
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
61
|
Tweedy L, Thomason PA, Paschke PI, Martin K, Machesky LM, Zagnoni M, Insall RH. Seeing around corners: Cells solve mazes and respond at a distance using attractant breakdown. Science 2020; 369:eaay9792. [PMID: 32855311 DOI: 10.1126/science.aay9792] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 05/12/2020] [Accepted: 07/02/2020] [Indexed: 02/11/2024]
Abstract
During development and metastasis, cells migrate large distances through complex environments. Migration is often guided by chemotaxis, but simple chemoattractant gradients between a source and sink cannot direct cells over such ranges. We describe how self-generated gradients, created by cells locally degrading attractant, allow single cells to navigate long, tortuous paths and make accurate choices between live channels and dead ends. This allows cells to solve complex mazes efficiently. Cells' accuracy at finding live channels was determined by attractant diffusivity, cell speed, and path complexity. Manipulating these parameters directed cells in mathematically predictable ways; specific combinations can even actively misdirect them. We propose that the length and complexity of many long-range migratory processes, including inflammation and germ cell migration, means that self-generated gradients are needed for successful navigation.
Collapse
Affiliation(s)
- Luke Tweedy
- CRUK Beatson Institute, Switchback Road, Glasgow G61 1BD, UK
| | | | - Peggy I Paschke
- CRUK Beatson Institute, Switchback Road, Glasgow G61 1BD, UK
| | - Kirsty Martin
- CRUK Beatson Institute, Switchback Road, Glasgow G61 1BD, UK
| | - Laura M Machesky
- CRUK Beatson Institute, Switchback Road, Glasgow G61 1BD, UK
- Institute for Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Michele Zagnoni
- Centre for Microsystems and Photonics, Electronic and Electrical Engineering, University of Strathclyde, Glasgow G1 1XW, UK
| | - Robert H Insall
- CRUK Beatson Institute, Switchback Road, Glasgow G61 1BD, UK.
- Institute for Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| |
Collapse
|
62
|
Combes F, Meyer E, Sanders NN. Immune cells as tumor drug delivery vehicles. J Control Release 2020; 327:70-87. [PMID: 32735878 DOI: 10.1016/j.jconrel.2020.07.043] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 12/21/2022]
Abstract
This review article describes the use of immune cells as potential candidates to deliver anti-cancer drugs deep within the tumor microenvironment. First, the rationale of using drug carriers to target tumors and potentially decrease drug-related side effects is discussed. We further explain some of the current limitations when using nanoparticles for this purpose. Next, a comprehensive step-by-step description of the migration cascade of immune cells is provided as well as arguments on why immune cells can be used to address some of the limitations associated with nanoparticle-mediated drug delivery. We then describe the benefits and drawbacks of using red blood cells, platelets, granulocytes, monocytes, macrophages, myeloid-derived suppressor cells, T cells and NK cells for tumor-targeted drug delivery. An additional section discusses the versatility of nanoparticles to load anti-cancer drugs into immune cells. Lastly, we propose increasing the circulatory half-life and development of conditional release strategies as the two main future pillars to improve the efficacy of immune cell-mediated drug delivery to tumors.
Collapse
Affiliation(s)
- Francis Combes
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium
| | - Evelyne Meyer
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium.
| |
Collapse
|
63
|
Lu M, Kim WH, Lillehoj HS, Li C. Development and characterization of novel mouse monoclonal antibodies against chicken chemokine CC motif ligand 4. Vet Immunol Immunopathol 2020; 227:110091. [PMID: 32682170 DOI: 10.1016/j.vetimm.2020.110091] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 05/28/2020] [Accepted: 07/07/2020] [Indexed: 11/19/2022]
Abstract
Chemokine (C-C motif) ligand (CCL) 4 is a CC chemokine subfamily member defined by the sequential positioning of conserved cysteine residues. Upon the binding of G-protein-coupled receptors on the cell surface, CCL4 mediates a diverse set of biological processes including chemotaxis, tumorigenesis, homeostasis and thymopoiesis. Although the physiological roles of mammalian CCL4s were elucidated >20 years ago, there is limited information on the biological activities of chicken CCL4 (chCCL4). In the present study, we developed and characterized mouse monoclonal antibodies (mAbs) against chCCL4 to characterize better the immunological properties of chCCL4. Out of initial screening of >400 clones, two mAbs detecting chCCL4, 1A12 and 15D9, were identified and characterized using western blotting and chCCL4-specific antigen-capture enzyme-linked immunosorbent assay, and their neutralizing activity was validated by chCCL4-induced peripheral blood mononuclear cell chemotaxis assay. Furthermore, the intracellular expression of chCCL4 in various chicken cells by immunocytochemistry and flow cytometry was confirmed using 1A12 and 15D9 mAbs. These results collectively indicate that 1A12 and 15D9 mAbs specifically detect chicken CCL4 and they will be valuable immune reagents for basic and applied studies in avian immunology.
Collapse
Affiliation(s)
- Mingmin Lu
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture Beltsville, MD, 20705, USA.
| | - Woo H Kim
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture Beltsville, MD, 20705, USA.
| | - Hyun S Lillehoj
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture Beltsville, MD, 20705, USA.
| | - Charles Li
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture Beltsville, MD, 20705, USA.
| |
Collapse
|
64
|
Yuan HX, Chen CY, Li YQ, Ning DS, Li Y, Chen YT, Li SX, Fu MX, Li XD, Ma J, Jian YP, Liu DH, Mo ZW, Peng YM, Xu KQ, Ou ZJ, Ou JS. Circulating extracellular vesicles from patients with valvular heart disease induce neutrophil chemotaxis via FOXO3a and the inhibiting role of dexmedetomidine. Am J Physiol Endocrinol Metab 2020; 319:E217-E231. [PMID: 32516026 DOI: 10.1152/ajpendo.00062.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We previously demonstrated that circulating extracellular vesicles (EVs) from patients with valvular heart disease (VHD; vEVs) contain inflammatory components and inhibit endothelium-dependent vasodilation. Neutrophil chemotaxis plays a key role in renal dysfunction, and dexmedetomidine (DEX) can reduce renal dysfunction in cardiac surgery. However, the roles of vEVs in neutrophil chemotaxis and effects of DEX on vEVs are unknown. Here, we investigated the impact of vEVs on neutrophil chemotaxis in kidneys and the influence of DEX on vEVs. Circulating EVs were isolated from healthy subjects and patients with VHD. The effects of EVs on chemokine generation, forkhead box protein O3a (FOXO3a) pathway activation and neutrophil chemotaxis on cultured human umbilical vein endothelial cells (HUVECs) and kidneys in mice and the influence of DEX on EVs were detected. vEVs increased FOXO3a expression, decreased phosphorylation of Akt and FOXO3a, promoted FOXO3a nuclear translocation, and activated the FOXO3a signaling pathway in vitro. DEX pretreatment reduced vEV-induced CXCL4 and CCL5 expression and neutrophil chemotaxis in cultured HUVECs via the FOXO3a signaling pathway. vEVs were also found to suppress Akt phosphorylation and activate FOXO3a signaling to increase plasma levels of CXCL4 and CCL5 and neutrophil accumulation in kidney. The overall mechanism was inhibited in vivo with DEX pretreatment. Our data demonstrated that vEVs induced CXCL4-CCL5 to stimulate neutrophil infiltration in kidney, which can be inhibited by DEX via the FOXO3a signaling. Our findings reveal a unique mechanism involving vEVs in inducing neutrophils chemotaxis and may provide a novel basis for using DEX in reducing renal dysfunction in valvular heart surgery.
Collapse
Affiliation(s)
- Hao-Xiang Yuan
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Cai-Yun Chen
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yu-Quan Li
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Da-Sheng Ning
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yan Li
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ya-Ting Chen
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Shang-Xuan Li
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Meng-Xia Fu
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiao-Di Li
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jian Ma
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yu-Peng Jian
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Dong-Hong Liu
- Department of Ultrasound, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhi-Wei Mo
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yue-Ming Peng
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Kang-Qing Xu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhi-Jun Ou
- Division of Hypertension and Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jing-Song Ou
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
65
|
Matsuda M, Terai K. Experimental pathology by intravital microscopy and genetically encoded fluorescent biosensors. Pathol Int 2020; 70:379-390. [PMID: 32270554 PMCID: PMC7383902 DOI: 10.1111/pin.12925] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/13/2020] [Accepted: 03/16/2020] [Indexed: 01/03/2023]
Abstract
The invention of two‐photon excitation microscopes widens the potential application of intravital microscopy (IVM) to the broad field of experimental pathology. Moreover, the recent development of fluorescent protein‐based, genetically encoded biosensors provides an ideal tool to visualize the cell function in live animals. We start from a brief review of IVM with two‐photon excitation microscopes and genetically encoded biosensors based on the principle of Förster resonance energy transfer (FRET). Then, we describe how IVM using biosensors has revealed the pathogenesis of several disease models.
Collapse
Affiliation(s)
- Michiyuki Matsuda
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Kenta Terai
- Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
66
|
Rogers TJ. Bidirectional Regulation of Opioid and Chemokine Function. Front Immunol 2020; 11:94. [PMID: 32076421 PMCID: PMC7006827 DOI: 10.3389/fimmu.2020.00094] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 01/14/2020] [Indexed: 12/15/2022] Open
Abstract
The opioid family of GPCRs consists of the classical opioid receptors, designated μ-, κ-, and δ-opioid receptors, and the orphanin-FQ receptor, and these proteins are expressed on both neuronal and hematopoietic cells. A number of laboratories have reported that an important degree of cross-talk can occur between the opioid receptors and the chemokine and chemokine receptor families. As a part of this, the opioid receptors are known to regulate the expression of certain chemokines and chemokine receptors, including those that possess strong pro-inflammatory activity. At the level of receptor function, it is clear that certain members of the chemokine family can mediate cross-desensitization of the opioid receptors. Conversely, the opioid receptors are all able to induce heterologous desensitization of some of the chemokine receptors. Consequently, activation of one or more of the opioid receptors can selectively cross-desensitize chemokine receptors and regulate chemokine function. These cross-talk processes have significant implications for the inflammatory response, since the regulation of both the recruitment of inflammatory cells, as well as the sensation of pain, can be controlled in this way.
Collapse
Affiliation(s)
- Thomas J Rogers
- Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
67
|
Elevated lymphocyte specific protein 1 expression is involved in the regulation of leukocyte migration and immunosuppressive microenvironment in glioblastoma. Aging (Albany NY) 2020; 12:1656-1684. [PMID: 32003759 PMCID: PMC7053627 DOI: 10.18632/aging.102706] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 01/02/2020] [Indexed: 02/07/2023]
Abstract
Immune cell infiltration mediates therapeutic response to immune therapies. The investigation on the genes regulating leukocyte migration may help us to understand the mechanisms regulating immune cell infiltration in tumor microenvironment. Here, we collected the data from Chinese Glioma Genome Atlas (CGGA) and The Cancer Genome Atlas (TCGA) to analyze the expression of leukocyte migration related genes in glioblastoma (GBM). Lymphocyte specific protein 1 (LSP1) was identified as the only gene in this family which not only has an elevated expression, but also serve as an independent predictive factor for progressive malignancy in glioma. We further confirmed these results in clinical glioma samples by quantitative PCR, immunofluorescence, immunohistochemistry, and western blot. Moreover, LSP1 expression was closely related to the response to radio- and chemotherapy in GBM, and positively correlated with immunosuppressive cell populations, including M2 macrophages, neutrophil, and regulatory T cell. Additionally, elevated LSP-1 expression enhanced the expression of immunosuppression related genes like programmed cell death 1 (PD1) and leukocyte associated immunoglobulin like receptor 1 (LAIR1) in macrophages. LSP1 also promoted the migration of macrophages. Together, our study suggests a novel role of LSP1 contributing to immunosuppressive microenvironment in GBM and serving as a potential therapeutic target for it.
Collapse
|
68
|
Do HTT, Lee CH, Cho J. Chemokines and their Receptors: Multifaceted Roles in Cancer Progression and Potential Value as Cancer Prognostic Markers. Cancers (Basel) 2020; 12:E287. [PMID: 31991604 PMCID: PMC7072521 DOI: 10.3390/cancers12020287] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/14/2020] [Accepted: 01/19/2020] [Indexed: 12/19/2022] Open
Abstract
Chemokines are chemotactic cytokines that mediate immune cell chemotaxis and lymphoid tissue development. Recent advances have indicated that chemokines and their cognate receptors play critical roles in cancer-related inflammation and cancer progression. On the basis of these findings, the chemokine system has become a new potential drug target for cancer immunotherapy. In this review, we summarize the essential roles of the complex network of chemokines and their receptors in cancer progression. Furthermore, we discuss the potential value of the chemokine system as a cancer prognostic marker. The chemokine system regulates the infiltration of immune cells into the tumor microenvironment, which induces both pro- and anti-immunity and promotes or suppresses tumor growth and proliferation, angiogenesis, and metastasis. Increasing evidence indicates the promising prognostic value of the chemokine system in cancer patients. While CCL2, CXCL10, and CX3CL1/CX3CR1 can serve as favorable or unfavorable prognostic factors depending on the cancer types, CCL14 and XCL1 possess good prognostic value. Other chemokines such as CXCL1, CXCL8, and CXCL12 are poor prognostic markers. Despite vast advances in our understanding of the complex nature of the chemokine system in tumor biology, knowledge about the multifaceted roles of the chemokine system in different types of cancers is still limited. Further studies are necessary to decipher distinct roles within the chemokine system in terms of cancer progression and to validate their potential value in cancer prognosis.
Collapse
Affiliation(s)
| | | | - Jungsook Cho
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi 10326, Korea; (H.T.T.D.); (C.H.L.)
| |
Collapse
|
69
|
Glaría E, Valledor AF. Roles of CD38 in the Immune Response to Infection. Cells 2020; 9:cells9010228. [PMID: 31963337 PMCID: PMC7017097 DOI: 10.3390/cells9010228] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/12/2020] [Accepted: 01/14/2020] [Indexed: 12/13/2022] Open
Abstract
CD38 is a multifunctional protein widely expressed in cells from the immune system and as a soluble form in biological fluids. CD38 expression is up-regulated by an array of inflammatory mediators, and it is frequently used as a cell activation marker. Studies in animal models indicate that CD38 functional expression confers protection against infection by several bacterial and parasitic pathogens. In addition, infectious complications are associated with anti-CD38 immunotherapy. Although CD38 displays receptor and enzymatic activities that contribute to the establishment of an effective immune response, recent work raises the possibility that CD38 might also enhance the immunosuppressive potential of regulatory leukocytes. This review integrates the current knowledge on the diversity of functions mediated by CD38 in the host defense to infection.
Collapse
|
70
|
Martin P, Wood W, Franz A. Cell migration by swimming: Drosophila adipocytes as a new in vivo model of adhesion-independent motility. Semin Cell Dev Biol 2019; 100:160-166. [PMID: 31812445 DOI: 10.1016/j.semcdb.2019.11.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/12/2019] [Accepted: 11/15/2019] [Indexed: 11/17/2022]
Abstract
Several cell lineages migrate through the developing and adult tissues of our bodies utilising a variety of modes of motility to suit the different substrates and environments they encounter en route to their destinations. Here we describe a novel adhesion-independent mode of single cell locomotion utilised by Drosophila fat body cells - the equivalent of vertebrate adipocytes. Like their human counterpart, these large cells were previously presumed to be immotile. However, in the Drosophila pupa fat body cells appear to be motile and migrate in a directed way towards wounds by peristaltic swimming through the hemolymph. The propulsive force is generated from a wave of cortical actomyosin that travels rearwards along the length of the cell. We discuss how this swimming mode of motility overcomes the physical constraints of microscopic objects moving in fluids, how fat body cells switch on other "motility machinery" to plug the wound on arrival, and whether other cell lineages in Drosophila and other organisms may, under certain circumstances, also adopt swimming as an effective mode of migration.
Collapse
Affiliation(s)
- Paul Martin
- School of Biochemistry, Biomedical Sciences, University of Bristol, Bristol, BS8 1TD, UK; School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, BS8 1TD, UK; School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Will Wood
- Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Anna Franz
- Department of Cell and Developmental Biology, University College London, 21 University Street, London, WC1E 6DE, UK.
| |
Collapse
|
71
|
Hawerkamp HC, Kislat A, Gerber PA, Pollet M, Rolfes KM, Soshilov AA, Denison MS, Momin AA, Arold ST, Datsi A, Braun SA, Oláh P, Lacouture ME, Krutmann J, Haarmann‐Stemmann T, Homey B, Meller S. Vemurafenib acts as an aryl hydrocarbon receptor antagonist: Implications for inflammatory cutaneous adverse events. Allergy 2019; 74:2437-2448. [PMID: 31269229 DOI: 10.1111/all.13972] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 05/02/2019] [Accepted: 05/21/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND In recent years, the BRAF inhibitor vemurafenib has been successfully established in the therapy of advanced melanoma. Despite its superior efficacy, the use of vemurafenib is limited by frequent inflammatory cutaneous adverse events that affect patients' quality of life and may lead to dose reduction or even cessation of anti-tumor therapy. To date, the molecular and cellular mechanisms of vemurafenib-induced rashes have remained largely elusive. METHODS In this study, we deployed immunohistochemistry, RT-qPCR, flow cytometry, lymphocyte activation tests, and different cell-free protein-interaction assays. RESULTS We here demonstrate that vemurafenib inhibits the downstream signaling of the canonical pathway of aryl hydrocarbon receptor (AhR) in vitro, thereby inducing the expression of proinflammatory cytokines (eg, TNF) and chemokines (eg, CCL5). In line with these results, we observed an impaired expression of AhR-regulated genes (eg, CYP1A1) and an upregulation of the corresponding proinflammatory genes in vivo. Moreover, results of lymphocyte activation tests showed the absence of drug-specific T cells in respective patients. CONCLUSION Taken together, we obtained no hint of an underlying sensitization against vemurafenib but found evidence suggesting that vemurafenib enhances proinflammatory responses by inhibition of canonical AhR signaling. Our findings contribute to our understanding of the central role of the AhR in skin inflammation and may point toward a potential role for topical AhR agonists in supportive cancer care.
Collapse
Affiliation(s)
- Heike C. Hawerkamp
- Department of Dermatology, Medical Faculty Heinrich‐Heine‐University Duesseldorf Germany
| | - Andreas Kislat
- Department of Dermatology, Medical Faculty Heinrich‐Heine‐University Duesseldorf Germany
| | - Peter A. Gerber
- Department of Dermatology, Medical Faculty Heinrich‐Heine‐University Duesseldorf Germany
| | - Marius Pollet
- Leibniz‐Research Institute for Environmental Medicine Duesseldorf Germany
| | | | - Anatoly A. Soshilov
- Department of Environmental Toxicology University of California Davis CA USA
| | - Michael S. Denison
- Department of Environmental Toxicology University of California Davis CA USA
| | - Afaque A. Momin
- King Abdullah University of Science and Technology (KAUST) Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE) Thuwal Saudi Arabia
| | - Stefan T. Arold
- King Abdullah University of Science and Technology (KAUST) Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE) Thuwal Saudi Arabia
| | - Angeliki Datsi
- Department of Dermatology, Medical Faculty Heinrich‐Heine‐University Duesseldorf Germany
| | - Stephan A. Braun
- Department of Dermatology, Medical Faculty Heinrich‐Heine‐University Duesseldorf Germany
| | - Péter Oláh
- Department of Dermatology, Medical Faculty Heinrich‐Heine‐University Duesseldorf Germany
- Department of Dermatology, Venereology and Oncodermatology University of Pécs Pécs Hungary
| | - Mario E. Lacouture
- Dermatology Service, Department of Medicine Memorial Sloan‐Kettering Cancer Center New York NY USA
| | - Jean Krutmann
- Leibniz‐Research Institute for Environmental Medicine Duesseldorf Germany
| | | | - Bernhard Homey
- Department of Dermatology, Medical Faculty Heinrich‐Heine‐University Duesseldorf Germany
| | - Stephan Meller
- Department of Dermatology, Medical Faculty Heinrich‐Heine‐University Duesseldorf Germany
| |
Collapse
|
72
|
Lam KH, Kivanany PB, Grose K, Yonet-Tanyeri N, Alsmadi N, Varner VD, Petroll WM, Schmidtke DW. A high-throughput microfluidic method for fabricating aligned collagen fibrils to study Keratocyte behavior. Biomed Microdevices 2019; 21:99. [PMID: 31741114 PMCID: PMC7228026 DOI: 10.1007/s10544-019-0436-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
In vivo, keratocytes are surrounded by aligned type I collagen fibrils that are organized into lamellae. A growing body of literature suggests that the unique topography of the corneal stroma is an important regulator of keratocyte behavior. In this study we describe a microfluidic method to deposit aligned fibrils of type I collagen onto glass coverslips. This high-throughput method allowed for the simultaneous coating of up to eight substrates with aligned collagen fibrils. When these substrates were integrated into a PDMS microwell culture system they provided a platform for high-resolution imaging of keratocyte behavior. Through the use of wide-field fluorescence and differential interference contrast microscopy, we observed that the density of collagen fibrils deposited was dependent upon both the perfusion shear rate of collagen and the time of perfusion. In contrast, a similar degree of fibril alignment was observed over a range of shear rates. When primary normal rabbit keratocytes (NRK) were seeded on substrates with a high density of aligned collagen fibrils and cultured in the presence of platelet derived growth factor (PDGF) the keratocytes displayed an elongated cell body that was co-aligned with the underlying collagen fibrils. In contrast, when NRK were cultured on substrates with a low density of aligned collagen fibrils, the cells showed no preferential orientation. These results suggest that this simple and inexpensive method can provide a general platform to study how simultaneous exposure to topographical and soluble cues influence cell behavior.
Collapse
Affiliation(s)
- Kevin H Lam
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Rd, Richardson, TX, 75080, USA
| | - Pouriska B Kivanany
- Department of Ophthalmology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9057, USA
| | - Kyle Grose
- Department of Ophthalmology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9057, USA
| | - Nihan Yonet-Tanyeri
- Department of Ophthalmology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9057, USA
| | - Nesreen Alsmadi
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Rd, Richardson, TX, 75080, USA
| | - Victor D Varner
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Rd, Richardson, TX, 75080, USA
- Department of Surgery, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9057, USA
| | - W Matthew Petroll
- Department of Ophthalmology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9057, USA
| | - David W Schmidtke
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Rd, Richardson, TX, 75080, USA.
- Department of Surgery, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9057, USA.
| |
Collapse
|