1
|
Huang J, Michaud E, Shinde-Jadhav S, Fehric S, Marcq G, Mansure JJ, Cury F, Brimo F, Piccirillo CA, Kassouf W. Effects of combined radiotherapy with immune checkpoint blockade on immunological memory in luminal-like subtype murine bladder cancer model. Cancer Biol Ther 2024; 25:2365452. [PMID: 38860746 PMCID: PMC11174127 DOI: 10.1080/15384047.2024.2365452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024] Open
Abstract
MIBC is a highly lethal disease, and the patient survival rate has not improved significantly over the last decades. UPPL is a cell line that can be used to recapitulate the luminal-like molecular subtype of bladder cancer and to discover effective treatments to be translated in patients. Here, we investigate the effects of combinational treatments of radiotherapy and immunotherapy in this recently characterized UPPL tumor-bearing mice. We first characterized the baseline tumor microenvironment and the effect of radiation, anti-PD-L1, and combinatorial treatments. Then, the mice were re-challenged with a second tumor (rechallenged tumor) in the contralateral flank of the first tumor to assess the immunological memory. Radiation slowed down the tumor growth. All treatments also decreased the neutrophil population and increased the T cell population. Anti-PD-L1 therapy was not able to synergize with radiation to further delay tumor growth. Furthermore, none of the treatments were able to generate immune memory. The treatments were not sufficient to induce a significant and lasting pool of memory cells. We show here that anti-PD-L1 treatment added to radiotherapy was not enough to achieve T cell-mediated memory in UPPL tumors. Stronger T cell activation signals may be required to enhance radiation efficacy in luminal-like bladder cancer.
Collapse
Affiliation(s)
- JiaMin Huang
- Cancer Research Program, Research Institute of McGill University Health Center, Montréal, QC, Canada
| | - Eva Michaud
- Cancer Research Program, Research Institute of McGill University Health Center, Montréal, QC, Canada
| | - Surashri Shinde-Jadhav
- Cancer Research Program, Research Institute of McGill University Health Center, Montréal, QC, Canada
| | - Sabina Fehric
- Cancer Research Program, Research Institute of McGill University Health Center, Montréal, QC, Canada
| | - Gautier Marcq
- Division of Urology, Department of Surgery, McGill University Health Center, Montréal, QC, Canada
| | - Jose Joao Mansure
- Cancer Research Program, Research Institute of McGill University Health Center, Montréal, QC, Canada
| | - Fabio Cury
- Department of Radiation Oncology, McGill University Health Center, Montréal, QC, Canada
| | - Fadi Brimo
- Department of Pathology, McGill University Health Center, Montréal, QC, Canada
| | - Ciriaco A. Piccirillo
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Infectious Diseases and Immunology in Global Health Program, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, QC, Canada
- Centre of Excellence in Translational Immunology, Montréal, QC, Canada
| | - Wassim Kassouf
- Cancer Research Program, Research Institute of McGill University Health Center, Montréal, QC, Canada
- Division of Urology, Department of Surgery, McGill University Health Center, Montréal, QC, Canada
- Centre of Excellence in Translational Immunology, Montréal, QC, Canada
| |
Collapse
|
2
|
Conceição JA, Carneiro PP, Dórea AS, Oliveira WN, Muniz AC, Carvalho EM, Wilson ME, Bacellar O. Contrasting Phenotypes of Neutrophils During Asymptomatic Versus Symptomatic Leishmania braziliensis Infection. J Infect Dis 2024; 230:e1385-e1393. [PMID: 38912968 PMCID: PMC11646601 DOI: 10.1093/infdis/jiae317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/08/2024] [Accepted: 06/19/2024] [Indexed: 06/25/2024] Open
Abstract
BACKGROUND The mechanisms that mediate immune protection in individuals with subclinical (SC) or asymptomatic infection with Leishmania braziliensis are largely unknown. Neutrophils (polymorphonuclear leukocytes [PMNs]) have been implicated in progressive symptomatic cutaneous leishmaniasis (CL), but their potential participation in maintenance of subclinical infection is unexplored. The aim of this study was to compare the phenotypic and functional profiles of PMNs in individuals with SC infection versus patients with symptomatic CL due to L braziliensis. METHODS Subjects were recruited in the endemic region of Corte de Pedra, Bahia, Brazil. Surface markers to define activation status were characterized by flow cytometry. Functional responses of PMNs including phagocytic capacity, production of oxidative species, and oxidative killing of intracellular parasites were studied in vitro. RESULTS PMNs from individuals with SC infection displayed a more activated phenotype and greater ability to control the infection than PMNs from patients with CL. In contrast, PMNs from patients with CL exhibited higher expression of HLA-DR and higher production of oxidative species than PMNs from subjects with SC infection. CONCLUSIONS PMNs from individuals with SC infection can control the infection more efficiently than PMNs from patients with CL, despite the lower production of oxidants. Our observations suggest that L braziliensis may evade microbicidal mechanisms of PMNs from patients with CL, contributing to parasite dissemination and the establishment of disease.
Collapse
Affiliation(s)
- Jacilara A Conceição
- Department of Internal Medicine, University of Iowa, Iowa City
- Serviço de Imunologia, Hospital Universitário Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Medical Service, Veterans’ Affairs Medical Center, Iowa City
| | - Pedro P Carneiro
- Serviço de Imunologia, Hospital Universitário Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Andreza S Dórea
- Serviço de Imunologia, Hospital Universitário Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Walker N Oliveira
- Serviço de Imunologia, Hospital Universitário Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Aline C Muniz
- Serviço de Imunologia, Hospital Universitário Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Edgar M Carvalho
- Instituto Gonçalo Moniz, Fiocruz Bahia
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais, The National Council for Scientific and Technological Development/Ministry of Science and Technology, Salvador, Brazil
| | - Mary E Wilson
- Department of Internal Medicine, University of Iowa, Iowa City
- Medical Service, Veterans’ Affairs Medical Center, Iowa City
- Department of Microbiology and Immunology, University of Iowa, Iowa City
| | - Olívia Bacellar
- Serviço de Imunologia, Hospital Universitário Prof. Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais, The National Council for Scientific and Technological Development/Ministry of Science and Technology, Salvador, Brazil
| |
Collapse
|
3
|
Vats D, Rani G, Arora A, Sharma V, Rathore I, Mubeen SA, Singh A. Tuberculosis and T cells: Impact of T cell diversity in tuberculosis infection. Tuberculosis (Edinb) 2024; 149:102567. [PMID: 39305817 DOI: 10.1016/j.tube.2024.102567] [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: 06/11/2024] [Revised: 09/07/2024] [Accepted: 09/11/2024] [Indexed: 11/30/2024]
Abstract
Tuberculosis is a global threat and is still a leading cause of death due to an infectious agent. The infection is spread through inhalation of M. tb containing aerosol droplets. Bacteria after reaching the lung alveoli are engulfed by alveolar macrophages, leading to an immune response. Then, pro-inflammatory cytokines are released by these macrophages, recruiting other antigen-presenting cells like dendritic cells. These cells phagocytose the bacteria and present mycobacterial antigens to naïve T cells. After activation by DCs, T cells differentiate into various T cells subsets, viz. CD4+, CD8+, Th17, Treg, Tfh cells and others display enormous diversification in their characteristics and functions. This review comprises a comprehensive literature on conventional and unconventional T cells, highlighting the polyfunctional T cells as well, their role in controlling TB infection, and their implications in the spectrum of TB infection. While some subsets such as CD4+ T cells are extensively studied, some T cell subsets such as gamma delta T cells and Tfh cells remain poorly understood in the pathophysiology of tuberculosis, despite having significant potential implications. The goal of TB eradication can be assisted by development of better vaccines against TB, which can effectively induce a robust and long-term T cells memory. The same has been discussed in the latter part of this review. BCG being the standalone commercialised TB vaccine so far has its limitations. Strategies for the enhancement of BCG along with novel studies in vaccine development, has also been discussed in great detail. Lastly, T cells display a complex interplay of an adaptive immune response against TB, with activation and enhancement of the innate immune responses. Therefore, it is critical to fully understand the role of various T cells subsets in pathophysiology of tuberculosis to provide better therapeutic inventions and improve patient care.
Collapse
Affiliation(s)
- Deepak Vats
- All India Institute of Medical Sciences, New Delhi, India
| | - Geeta Rani
- All India Institute of Medical Sciences, New Delhi, India
| | - Alisha Arora
- All India Institute of Medical Sciences, New Delhi, India
| | - Vidushi Sharma
- All India Institute of Medical Sciences, New Delhi, India
| | - Isha Rathore
- All India Institute of Medical Sciences, New Delhi, India
| | | | - Archana Singh
- All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
4
|
Ramarapu R, Wulcan JM, Chang H, Moore PF, Vernau W, Keller SM. Single cell RNA-sequencing of feline peripheral immune cells with V(D)J repertoire and cross species analysis of T lymphocytes. Front Immunol 2024; 15:1438004. [PMID: 39620216 PMCID: PMC11604454 DOI: 10.3389/fimmu.2024.1438004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/23/2024] [Indexed: 12/11/2024] Open
Abstract
Introduction The domestic cat (Felis catus) is a valued companion animal and a model for virally induced cancers and immunodeficiencies. However, species-specific limitations such as a scarcity of immune cell markers constrain our ability to resolve immune cell subsets at sufficient detail. The goal of this study was to characterize circulating feline T cells and other leukocytes based on their transcriptomic landscape and T-cell receptor repertoire using single cell RNA-sequencing. Methods Peripheral blood from 4 healthy cats was enriched for T cells by flow cytometry cell sorting using a mouse anti-feline CD5 monoclonal antibody. Libraries for whole transcriptome, αβ T cell receptor transcripts and γδ T cell receptor transcripts were constructed using the 10x Genomics Chromium Next GEM Single Cell 5' reagent kit and the Chromium Single Cell V(D)J Enrichment Kit with custom reverse primers for the feline orthologs. Results Unsupervised clustering of whole transcriptome data revealed 7 major cell populations - T cells, neutrophils, monocytic cells, B cells, plasmacytoid dendritic cells, mast cells and platelets. Sub cluster analysis of T cells resolved naive (CD4+ and CD8+), CD4+ effector T cells, CD8+ cytotoxic T cells and γδ T cells. Cross species analysis revealed a high conservation of T cell subsets along an effector gradient with equitable representation of veterinary species (horse, dog, pig) and humans with the cat. Our V(D)J repertoire analysis identified a subset of CD8+ cytotoxic T cells with skewed TRA and TRB gene usage, conserved TRA and TRB junctional motifs, restricted TRA/TRB pairing and reduced diversity in TRG junctional length. We also identified a public γδ T cell subset with invariant TRD and TRG chains and a CD4+ TEM-like phenotype. Among monocytic cells, we resolved three clusters of classical monocytes with polarization into pro- and anti-inflammatory phenotypes in addition to a cluster of conventional dendritic cells. Lastly, our neutrophil sub clustering revealed a larger mature neutrophil cluster and a smaller exhausted/activated cluster. Discussion Our study is the first to characterize subsets of circulating T cells utilizing an integrative approach of single cell RNA-sequencing, V(D)J repertoire analysis and cross species analysis. In addition, we characterize the transcriptome of several myeloid cell subsets and demonstrate immune cell relatedness across different species.
Collapse
MESH Headings
- Animals
- Cats
- Single-Cell Analysis
- Transcriptome
- Species Specificity
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Dogs
- Sequence Analysis, RNA
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- RNA-Seq
- V(D)J Recombination/genetics
Collapse
Affiliation(s)
- Raneesh Ramarapu
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Judit M. Wulcan
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Haiyang Chang
- Department of Mathematics and Statistics, University of Guelph, Guelph, ON, Canada
| | - Peter F. Moore
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - William Vernau
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Stefan M. Keller
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
5
|
Xiong H, Shen Z. Tissue-resident memory T cells in immunotherapy and immune-related adverse events by immune checkpoint inhibitor. Int J Cancer 2024; 155:193-202. [PMID: 38554117 DOI: 10.1002/ijc.34940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 04/01/2024]
Abstract
Tissue-resident memory T cells (TRM) are a specialized subset of T cells that reside in tissues and provide long-term protective immunity against pathogens that enter the body through that specific tissue. TRM cells have specific phenotype and reside preferentially in barrier tissues. Recent studies have revealed that TRM cells are the main target of immune checkpoint inhibitor immunotherapy since their role in cancer immunosurveillance. Furthermore, TRM cells also play a crucial part in pathogenesis of immune-related adverse events (irAEs). Here, we provide a concise review of biological characteristics of TRM cells, and the major advances and recent findings regarding their involvement in immune checkpoint inhibitor immunotherapy and the corresponding irAEs.
Collapse
Affiliation(s)
- Hao Xiong
- Department of Dermatology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhu Shen
- Department of Dermatology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
6
|
Kaushik A, Chang I, Han X, He Z, Komlosi ZI, Ji X, Cao S, Akdis CA, Boyd S, Pulendran B, Maecker HT, Davis MM, Chinthrajah RS, DeKruyff RH, Nadeau KC. Single cell multi-omic analysis identifies key genes differentially expressed in innate lymphoid cells from COVID-19 patients. Front Immunol 2024; 15:1374828. [PMID: 39026668 PMCID: PMC11255397 DOI: 10.3389/fimmu.2024.1374828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 06/07/2024] [Indexed: 07/20/2024] Open
Abstract
Introduction Innate lymphoid cells (ILCs) are enriched at mucosal surfaces where they respond rapidly to environmental stimuli and contribute to both tissue inflammation and healing. Methods To gain insight into the role of ILCs in the pathology and recovery from COVID-19 infection, we employed a multi-omics approach consisting of Abseq and targeted mRNA sequencing to respectively probe the surface marker expression, transcriptional profile and heterogeneity of ILCs in peripheral blood of patients with COVID-19 compared with healthy controls. Results We found that the frequency of ILC1 and ILC2 cells was significantly increased in COVID-19 patients. Moreover, all ILC subsets displayed a significantly higher frequency of CD69-expressing cells, indicating a heightened state of activation. ILC2s from COVID-19 patients had the highest number of significantly differentially expressed (DE) genes. The most notable genes DE in COVID-19 vs healthy participants included a) genes associated with responses to virus infections and b) genes that support ILC self-proliferation, activation and homeostasis. In addition, differential gene regulatory network analysis revealed ILC-specific regulons and their interactions driving the differential gene expression in each ILC. Discussion Overall, this study provides mechanistic insights into the characteristics of ILC subsets activated during COVID-19 infection.
Collapse
Affiliation(s)
- Abhinav Kaushik
- Sean N. Parker Center for Allergy and Asthma Research, Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Iris Chang
- Sean N. Parker Center for Allergy and Asthma Research, Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Xiaorui Han
- Sean N. Parker Center for Allergy and Asthma Research, Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Ziyuan He
- Sean N. Parker Center for Allergy and Asthma Research, Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Zsolt I. Komlosi
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- Swiss Institute of Allergy and Asthma (SIAF), University of Zurich, Davos, Switzerland
| | - Xuhuai Ji
- Human Immune Monitoring Center, Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, United States
| | - Shu Cao
- Sean N. Parker Center for Allergy and Asthma Research, Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma (SIAF), University of Zurich, Davos, Switzerland
- Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | - Scott Boyd
- Sean N. Parker Center for Allergy and Asthma Research, Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
- Department of Pathology, Stanford University, Stanford, CA, United States
| | - Bali Pulendran
- Department of Pathology, Stanford University, Stanford, CA, United States
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, United States
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Holden T. Maecker
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, United States
| | - Mark M. Davis
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, United States
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, United States
| | - R. Sharon Chinthrajah
- Sean N. Parker Center for Allergy and Asthma Research, Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Rosemarie H. DeKruyff
- Sean N. Parker Center for Allergy and Asthma Research, Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Kari C. Nadeau
- Sean N. Parker Center for Allergy and Asthma Research, Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
7
|
de Oliveira CGN, Perez EC, Alvares-Saraiva AM, Lallo MA. CD8 T lymphocytes from B-1 cell-deficient mice down-regulates fungicidal activity of macrophages challenged with E. Cuniculi. Immunobiology 2024; 229:152827. [PMID: 38878483 DOI: 10.1016/j.imbio.2024.152827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/03/2024] [Accepted: 06/11/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Encephalitozoon cuniculi is an opportunistic intracellular pathogen that establishes a balanced relationship with immunocompetent individuals depending on the activity of their CD8+ T cells lymphocytes. However, lower resistance to experimental infection with E. cuniculi was found in B-1 deficient mice (Xid), besides increased the number of CD8 T lymphocytes. Here, we evaluated the profile of CD8+ T lymphocytes from Balb/c wild-type (WT) or Balb/c Xid mice (with B-1 cell deficiency) on the microbicidal activity of macrophages challenged with E. cuniculi. METHODS Naïve CD8 T lymphocytes from WT or Xid mice uninfected and primed CD8 T lymphocytes from WT or Xid mice infected with E cuniculi were co-cultured with macrophages previously challenged with E. cuniculi. We evaluated macrophages viability and microbicidal activity, and CD8 T lymphocytes viability and presence of activating molecules (CD62L, CD69, and CD107a). RESULTS Macrophages co-cultured with naïve CD8 T lymphocytes from WT demonstrated high microbicidal activity. Naïve CD8 T lymphocytes obtained from WT mice had a higher expression of CD69 and LAMP-1-activating molecules compared to Xid CD8+ T lymphocytes. Primed CD8 T lymphocytes from Xid mice proliferated more than those from WT mice, however, when the expression of the activating molecule CD69 associated with the expression of CD62L was kept low. In conclusion, naïve CD8+ T lymphocytes from Xid mice, deficient in B-1 cells, they had reduced expression of activation molecules and cytotoxic activity.
Collapse
Affiliation(s)
| | - Elizabeth Cristina Perez
- Programa de Pós-Graduação em Patologia Ambiental e Experimental, Universidade Paulista - Unip, Rua Dr Bacelar 1212, CEP 04026002 São Paulo, SP, Brazil
| | - Anuska Marcelino Alvares-Saraiva
- Programa de Pós-Graduação em Patologia Ambiental e Experimental, Universidade Paulista - Unip, Rua Dr Bacelar 1212, CEP 04026002 São Paulo, SP, Brazil
| | - Maria Anete Lallo
- Programa de Pós-Graduação em Patologia Ambiental e Experimental, Universidade Paulista - Unip, Rua Dr Bacelar 1212, CEP 04026002 São Paulo, SP, Brazil.
| |
Collapse
|
8
|
Pekayvaz K, Losert C, Knottenberg V, Gold C, van Blokland IV, Oelen R, Groot HE, Benjamins JW, Brambs S, Kaiser R, Gottschlich A, Hoffmann GV, Eivers L, Martinez-Navarro A, Bruns N, Stiller S, Akgöl S, Yue K, Polewka V, Escaig R, Joppich M, Janjic A, Popp O, Kobold S, Petzold T, Zimmer R, Enard W, Saar K, Mertins P, Huebner N, van der Harst P, Franke LH, van der Wijst MGP, Massberg S, Heinig M, Nicolai L, Stark K. Multiomic analyses uncover immunological signatures in acute and chronic coronary syndromes. Nat Med 2024; 30:1696-1710. [PMID: 38773340 PMCID: PMC11186793 DOI: 10.1038/s41591-024-02953-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 03/26/2024] [Indexed: 05/23/2024]
Abstract
Acute and chronic coronary syndromes (ACS and CCS) are leading causes of mortality. Inflammation is considered a key pathogenic driver of these diseases, but the underlying immune states and their clinical implications remain poorly understood. Multiomic factor analysis (MOFA) allows unsupervised data exploration across multiple data types, identifying major axes of variation and associating these with underlying molecular processes. We hypothesized that applying MOFA to multiomic data obtained from blood might uncover hidden sources of variance and provide pathophysiological insights linked to clinical needs. Here we compile a longitudinal multiomic dataset of the systemic immune landscape in both ACS and CCS (n = 62 patients in total, n = 15 women and n = 47 men) and validate this in an external cohort (n = 55 patients in total, n = 11 women and n = 44 men). MOFA reveals multicellular immune signatures characterized by distinct monocyte, natural killer and T cell substates and immune-communication pathways that explain a large proportion of inter-patient variance. We also identify specific factors that reflect disease state or associate with treatment outcome in ACS as measured using left ventricular ejection fraction. Hence, this study provides proof-of-concept evidence for the ability of MOFA to uncover multicellular immune programs in cardiovascular disease, opening new directions for mechanistic, biomarker and therapeutic studies.
Collapse
Grants
- Deutsche Forschungsgemeinschaft (German Research Foundation)
- Deutsches Zentrum fr Herz-Kreislaufforschung (Deutsches Zentrum fr Herz-Kreislaufforschung e.V.)
- Deutsche Herzstiftung e.V., Frankfurt a.M. Institutional Strategy LMUexcellent of LMU Munich Else-Krner-Fresenius Stiftung DFG Clinician Scientist Programme PRIME DZHK Sule B Antrag DZHK B 21-014 SE
- Was supported by the Helmholtz Association under the joint research school ;Munich School for Data Science MUDS
- DFG GO 3823/1-1, grant number: 510821390 Frderprogramm fr Forschung und Lehre der Medizinischen Fakultt der LMU the Bavarian Cancer Research Center (BZKF) Else Kroner-Fresenius-Stiftung
- Was supported by a grant from the Frderprogramm fur Forschung und Lehre (FFoLe) of the Ludwig Maximilian University (LMU) of Munich.
- DFG SFB 1123, Z02
- DFG EN 1093/2-1
- DFG KO5055-2-1 and KO5055/3-1 the Bavarian Cancer Research Center (BZKF) the international doctoral program i-Target: immunotargeting of cancer the Melanoma Research Alliance (grant number 409510), Marie Sklodowska-Curie Training Network for Optimizing Adoptive T Cell Therapy of Cancer (funded by the Horizon 2020 programme of the European Union; grant 955575), Else Kroner-Fresenius-Stiftung (IOLIN), German Cancer Aid (AvantCAR.de), the Wilhelm-Sander-Stiftung, Ernst Jung Stiftung, Institutional Strategy LMUexcellent of LMU Munich (within the framework of the German Excellence Initiative), the Go-Bio-Initiative, the m4-Award of the Bavarian Ministry for Economical Affairs, Bundesministerium fur Bildung und Forschung, European Research Council (Starting Grant 756017 and PoC Grant 101100460, by the SFB-TRR 338/1 2021452881907, Fritz-Bender Foundation, Deutsche Jose#x0301; Carreras Leuk#x00E4;mie Stiftung, Hector Foundation, the Bavarian Research Foundation, the Bruno and Helene J#x00F6;ster Foundation (360#x00B0; CAR)
- T.P. from the DFG (PE 2704/3-1)
- DFG SFB1243, A14 DFG EN 1093/2-1,
- DZHK Säule B Antrag DZHK B 21-014 SE
- DZHK Säule B Antrag DZHK B 21-014 SE DFG SFB-1470-B03 the Chan Zuckerberg Foundation ERC Advanced Grant under the European Union Horizon 2020 Research and Innovation Program (AdG788970)
- Deutsche Forschungsgemeinschaft (DFG) SFB 914, B02 and Z01 DFG SFB 1123, B06 DFG SFB1321, P10 DFG FOR 2033 ERC-2018-ADG German Centre for Cardiovascular Research (DZHK) MHA 1.4VD
- DZHK project 81Z0600106 Supported by the Chan Zuckerberg Foundation
- DZHK S#x00E4;ule B Antrag DZHK B 21-014 SE Deutsche Herzstiftung e.V., Frankfurt a.M. DFG SFB 1123, B06 DFG NI 2219/2-1 Corona Foundation German Centre for Cardiovascular Research (DZHK) Clinician Scientist Programme the Ernst und Berta Grimmke Stiftung the GTH Junior research grant
- DZHK partner site project Deutsche Forschungsgemeinschaft (DFG) SFB 914, B02 DFG SFB 1123, A07 DFG SFB 359, A03 ERC grant 947611
Collapse
Affiliation(s)
- Kami Pekayvaz
- Medizinische Klinik und Poliklinik I, LMU University Hospital, Munich, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
| | - Corinna Losert
- Institute of Computational Biology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany
- Department of Computer Science, TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
| | | | - Christoph Gold
- Medizinische Klinik und Poliklinik I, LMU University Hospital, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Irene V van Blokland
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Roy Oelen
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Hilde E Groot
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan Walter Benjamins
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sophia Brambs
- Medizinische Klinik und Poliklinik I, LMU University Hospital, Munich, Germany
| | - Rainer Kaiser
- Medizinische Klinik und Poliklinik I, LMU University Hospital, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Adrian Gottschlich
- Department of Medicine III, LMU University Hospital, Munich, Germany
- Division of Clinical Pharmacology, LMU University Hospital, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Gordon Victor Hoffmann
- Division of Clinical Pharmacology, LMU University Hospital, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Luke Eivers
- Medizinische Klinik und Poliklinik I, LMU University Hospital, Munich, Germany
| | | | - Nils Bruns
- Medizinische Klinik und Poliklinik I, LMU University Hospital, Munich, Germany
| | - Susanne Stiller
- Medizinische Klinik und Poliklinik I, LMU University Hospital, Munich, Germany
| | - Sezer Akgöl
- Medizinische Klinik und Poliklinik I, LMU University Hospital, Munich, Germany
| | - Keyang Yue
- Medizinische Klinik und Poliklinik I, LMU University Hospital, Munich, Germany
| | - Vivien Polewka
- Medizinische Klinik und Poliklinik I, LMU University Hospital, Munich, Germany
| | - Raphael Escaig
- Medizinische Klinik und Poliklinik I, LMU University Hospital, Munich, Germany
| | - Markus Joppich
- Department of Informatics, Ludwig-Maximilian University, Munich, Germany
| | - Aleksandar Janjic
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilian University, Munich, Germany
| | - Oliver Popp
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, LMU University Hospital, Member of the German Center for Lung Research (DZL), Munich, Germany
- German Cancer Consortium (DKTK), a partnership between DKFZ and LMU University Hospital, Partner Site Munich, Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Tobias Petzold
- Medizinische Klinik und Poliklinik I, LMU University Hospital, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Charite-Universitätsmedizin Berlin, Berlin, Germany
| | - Ralf Zimmer
- Department of Informatics, Ludwig-Maximilian University, Munich, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilian University, Munich, Germany
| | - Kathrin Saar
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Philipp Mertins
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Norbert Huebner
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Charite-Universitätsmedizin Berlin, Berlin, Germany
| | - Pim van der Harst
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lude H Franke
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Monique G P van der Wijst
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, LMU University Hospital, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Matthias Heinig
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
- Institute of Computational Biology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, Germany.
- Department of Computer Science, TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany.
| | - Leo Nicolai
- Medizinische Klinik und Poliklinik I, LMU University Hospital, Munich, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
| | - Konstantin Stark
- Medizinische Klinik und Poliklinik I, LMU University Hospital, Munich, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
9
|
Ramarapu R, Wulcan JM, Chang H, Moore PF, Vernau W, Keller SM. Single cell RNA-sequencing of feline peripheral immune cells with V(D)J repertoire and cross species analysis of T lymphocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595010. [PMID: 38826195 PMCID: PMC11142102 DOI: 10.1101/2024.05.21.595010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Introduction The domestic cat (Felis catus) is a valued companion animal and a model for virally induced cancers and immunodeficiencies. However, species-specific limitations such as a scarcity of immune cell markers constrain our ability to resolve immune cell subsets at sufficient detail. The goal of this study was to characterize circulating feline T cells and other leukocytes based on their transcriptomic landscape and T-cell receptor repertoire using single cell RNA-sequencing. Methods Peripheral blood from 4 healthy cats was enriched for T cells by flow cytometry cell sorting using a mouse anti-feline CD5 monoclonal antibody. Libraries for whole transcriptome, alpha/beta T cell receptor transcripts and gamma/delta T cell receptor transcripts were constructed using the 10x Genomics Chromium Next GEM Single Cell 5' reagent kit and the Chromium Single Cell V(D)J Enrichment Kit with custom reverse primers for the feline orthologs. Results Unsupervised clustering of whole transcriptome data revealed 7 major cell populations - T cells, neutrophils, monocytic cells, B cells, plasmacytoid dendritic cells, mast cells and platelets. Sub cluster analysis of T cells resolved naive (CD4+ and CD8+), CD4+ effector T cells, CD8+ cytotoxic T cells and gamma/delta T cells. Cross species analysis revealed a high conservation of T cell subsets along an effector gradient with equitable representation of veterinary species (horse, dog, pig) and humans with the cat. Our V(D)J repertoire analysis demonstrated a skewed T-cell receptor alpha gene usage and a restricted T-cell receptor gamma junctional length in CD8+ cytotoxic T cells compared to other alpha/beta T cell subsets. Among myeloid cells, we resolved three clusters of classical monocytes with polarization into pro- and anti-inflammatory phenotypes in addition to a cluster of conventional dendritic cells. Lastly, our neutrophil sub clustering revealed a larger mature neutrophil cluster and a smaller exhausted/activated cluster. Discussion Our study is the first to characterize subsets of circulating T cells utilizing an integrative approach of single cell RNA-sequencing, V(D)J repertoire analysis and cross species analysis. In addition, we characterize the transcriptome of several myeloid cell subsets and demonstrate immune cell relatedness across different species.
Collapse
Affiliation(s)
- Raneesh Ramarapu
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Judit M Wulcan
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, CA, United States
| | - Haiyang Chang
- Department of Mathematics and Statistics, University of Guelph, Guelph, ON, Canada
| | - Peter F Moore
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, CA, United States
| | - William Vernau
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, CA, United States
| | - Stefan M Keller
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, CA, United States
| |
Collapse
|
10
|
Negash M, Chanyalew M, Girma T, Alemu F, Alcantara D, Towler B, Davey G, Boyton RJ, Altmann DM, Howe R, Newport MJ. Evidence for immune activation in pathogenesis of the HLA class II associated disease, podoconiosis. Nat Commun 2024; 15:2020. [PMID: 38448477 PMCID: PMC10917762 DOI: 10.1038/s41467-024-46347-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/23/2024] [Indexed: 03/08/2024] Open
Abstract
Available evidences suggest that podoconiosis is triggered by long term exposure of bare feet to volcanic red clay soil particles. Previous genome-wide studies in Ethiopia showed association between the HLA class II region and disease susceptibility. However, functional relationships between the soil trigger, immunogenetic risk factors and the immunological basis of the disease are uncharted. Therefore, we aimed to characterise the immune profile and gene expression of podoconiosis patients relative to endemic healthy controls. Peripheral blood immunophenotyping of T cells indicated podoconiosis patients had significantly higher CD4 and CD8 T cell surface HLA-DR expression compared to healthy controls while CD62L expression was significantly lower. The levels of the activation markers CD40 and CD86 were significantly higher on monocytes and dendritic cell subsets in patients compared to the controls. RNA sequencing gene expression data indicated higher transcript levels for activation, scavenger receptors, and apoptosis markers while levels were lower for histones, T cell receptors, variable, and constant immunoglobulin chain in podoconiosis patients compared to healthy controls. Our finding provides evidence that podoconiosis is associated with high levels of immune activation and inflammation with over-expression of genes within the pro-inflammatory axis. This offers further support to a working hypothesis of podoconiosis as soil particle-driven, HLA-associated disease of immunopathogenic aetiology.
Collapse
Affiliation(s)
- Mikias Negash
- Brighton and Sussex Centre for Global Health Research, Department of Global Health and Infection, Brighton and Sussex Medical School, Brighton, UK.
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia.
- Department of Medical Laboratory Science, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia.
| | | | - Tigist Girma
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Fekadu Alemu
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Diana Alcantara
- Brighton and Sussex Centre for Global Health Research, Department of Global Health and Infection, Brighton and Sussex Medical School, Brighton, UK
| | - Ben Towler
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, UK
| | - Gail Davey
- Brighton and Sussex Centre for Global Health Research, Department of Global Health and Infection, Brighton and Sussex Medical School, Brighton, UK
- School of Public Health, Addis Ababa University, Addis Ababa, Ethiopia
| | - Rosemary J Boyton
- Department of Infectious Disease, Imperial College London, London, UK
| | - Daniel M Altmann
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Rawleigh Howe
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Melanie J Newport
- Brighton and Sussex Centre for Global Health Research, Department of Global Health and Infection, Brighton and Sussex Medical School, Brighton, UK
| |
Collapse
|
11
|
Pokharel J, Shryki I, Zwijnenburg AJ, Sandu I, Krumm L, Bekiari C, Avramov V, Heinbäck R, Lysell J, Eidsmo L, Harris HE, Gerlach C. The cellular microenvironment regulates CX3CR1 expression on CD8 + T cells and the maintenance of CX3CR1 + CD8 + T cells. Eur J Immunol 2024; 54:e2350658. [PMID: 37816219 DOI: 10.1002/eji.202350658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 10/12/2023]
Abstract
Expression levels of the chemokine receptor CX3CR1 serve as high-resolution marker delineating functionally distinct antigen-experienced T-cell states. The factors that influence CX3CR1 expression in T cells are, however, incompletely understood. Here, we show that in vitro priming of naïve CD8+ T cells failed to robustly induce CX3CR1, which highlights the shortcomings of in vitro priming settings in recapitulating in vivo T-cell differentiation. Nevertheless, in vivo generated memory CD8+ T cells maintained CX3CR1 expression during culture. This allowed us to investigate whether T-cell receptor ligation, cell death, and CX3CL1 binding influence CX3CR1 expression. T-cell receptor stimulation led to downregulation of CX3CR1. Without stimulation, CX3CR1+ CD8+ T cells had a selective survival disadvantage, which was enhanced by factors released from necrotic but not apoptotic cells. Exposure to CX3CL1 did not rescue their survival and resulted in a dose-dependent loss of CX3CR1 surface expression. At physiological concentrations of CX3CL1, CX3CR1 surface expression was only minimally reduced, which did not hamper the interpretability of T-cell differentiation states delineated by CX3CR1. Our data further support the broad utility of CX3CR1 surface levels as T-cell differentiation marker and identify factors that influence CX3CR1 expression and the maintenance of CX3CR1 expressing CD8+ T cells.
Collapse
Affiliation(s)
- Jyoti Pokharel
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Iman Shryki
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Anthonie J Zwijnenburg
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Ioana Sandu
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Laura Krumm
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Christina Bekiari
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Victor Avramov
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Rebecka Heinbäck
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Josefin Lysell
- Dermatology and Venereology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Liv Eidsmo
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
- Leo Foundation Skin Immunology Center, University of Copenhagen, Kobenhavn, Denmark
| | - Helena E Harris
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Carmen Gerlach
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| |
Collapse
|
12
|
Alsalloum A, Alrhmoun S, Shevchenko J, Fisher M, Philippova J, Perik-Zavodskii R, Perik-Zavodskaia O, Lopatnikova J, Kurilin V, Volynets M, Akahori Y, Shiku H, Silkov A, Sennikov S. TCR-Engineered Lymphocytes Targeting NY-ESO-1: In Vitro Assessment of Cytotoxicity against Tumors. Biomedicines 2023; 11:2805. [PMID: 37893178 PMCID: PMC10604587 DOI: 10.3390/biomedicines11102805] [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: 09/29/2023] [Revised: 10/13/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
Adoptive T-cell therapies tailored for the treatment of solid tumors encounter intricate challenges, necessitating the meticulous selection of specific target antigens and the engineering of highly specific T-cell receptors (TCRs). This study delves into the cytotoxicity and functional characteristics of in vitro-cultured T-lymphocytes, equipped with a TCR designed to precisely target the cancer-testis antigen NY-ESO-1. Flow cytometry analysis unveiled a notable increase in the population of cells expressing activation markers upon encountering the NY-ESO-1-positive tumor cell line, SK-Mel-37. Employing the NanoString platform, immune transcriptome profiling revealed the upregulation of genes enriched in Gene Ontology Biological Processes associated with the IFN-γ signaling pathway, regulation of T-cell activation, and proliferation. Furthermore, the modified T cells exhibited robust cytotoxicity in an antigen-dependent manner, as confirmed by the LDH assay results. Multiplex immunoassays, including LEGENDplex™, additionally demonstrated the elevated production of cytotoxicity-associated cytokines driven by granzymes and soluble Fas ligand (sFasL). Our findings underscore the specific targeting potential of engineered TCR T cells against NY-ESO-1-positive tumors. Further comprehensive in vivo investigations are essential to thoroughly validate these results and effectively harness the intrinsic potential of genetically engineered T cells for combating cancer.
Collapse
Affiliation(s)
- Alaa Alsalloum
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
- Faculty of Natural Sciences, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Saleh Alrhmoun
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
- Faculty of Natural Sciences, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Julia Shevchenko
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
| | - Marina Fisher
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
| | - Julia Philippova
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
| | - Roman Perik-Zavodskii
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
| | - Olga Perik-Zavodskaia
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
| | - Julia Lopatnikova
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
| | - Vasily Kurilin
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
| | - Marina Volynets
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
- Faculty of Natural Sciences, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Yasushi Akahori
- Department of Personalized Cancer Immunotherapy, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan;
| | - Hiroshi Shiku
- Department of Personalized Cancer Immunotherapy, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan;
| | - Alexander Silkov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
| | - Sergey Sennikov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
- Department of Immunology, V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, Novosibirsk 630090, Russia
| |
Collapse
|
13
|
Hausmann F, Ergen C, Khatri R, Marouf M, Hänzelmann S, Gagliani N, Huber S, Machart P, Bonn S. DISCERN: deep single-cell expression reconstruction for improved cell clustering and cell subtype and state detection. Genome Biol 2023; 24:212. [PMID: 37730638 PMCID: PMC10510283 DOI: 10.1186/s13059-023-03049-x] [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: 11/15/2022] [Accepted: 08/23/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND Single-cell sequencing provides detailed insights into biological processes including cell differentiation and identity. While providing deep cell-specific information, the method suffers from technical constraints, most notably a limited number of expressed genes per cell, which leads to suboptimal clustering and cell type identification. RESULTS Here, we present DISCERN, a novel deep generative network that precisely reconstructs missing single-cell gene expression using a reference dataset. DISCERN outperforms competing algorithms in expression inference resulting in greatly improved cell clustering, cell type and activity detection, and insights into the cellular regulation of disease. We show that DISCERN is robust against differences between batches and is able to keep biological differences between batches, which is a common problem for imputation and batch correction algorithms. We use DISCERN to detect two unseen COVID-19-associated T cell types, cytotoxic CD4+ and CD8+ Tc2 T helper cells, with a potential role in adverse disease outcome. We utilize T cell fraction information of patient blood to classify mild or severe COVID-19 with an AUROC of 80% that can serve as a biomarker of disease stage. DISCERN can be easily integrated into existing single-cell sequencing workflow. CONCLUSIONS Thus, DISCERN is a flexible tool for reconstructing missing single-cell gene expression using a reference dataset and can easily be applied to a variety of data sets yielding novel insights, e.g., into disease mechanisms.
Collapse
Affiliation(s)
- Fabian Hausmann
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Can Ergen
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Robin Khatri
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Mohamed Marouf
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Sonja Hänzelmann
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Nicola Gagliani
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Samuel Huber
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Pierre Machart
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Stefan Bonn
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
- Hamburg Center for Translational Immunology (HCTI), I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
14
|
Ettel P, Sehgal ANA, Harrison N, Pickl WF, Grabmeier-Pfistershammer K. Glycopeptide Antibiotics Impair Neutrophil Effector Functions. Int Arch Allergy Immunol 2023; 184:932-948. [PMID: 37321197 DOI: 10.1159/000530865] [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: 02/10/2023] [Accepted: 04/21/2023] [Indexed: 06/17/2023] Open
Abstract
INTRODUCTION Neutrophilic granulocytes represent the first line of defense against microorganisms. Granulocytes phagocytose microorganisms and specifically synthesize oxygen radicals against them, which eventually kills the invaders. METHODS Neutrophilic granulocytes were isolated from peripheral blood of healthy volunteer donors. Putative interference of new-generation antibiotics with neutrophil function was tested using a collection of granulocyte-stimulating agents and Amplex™ Red-based plate assay and flow cytometry-based respiratory burst assays. In addition, phagocytosis of E. coli, IL-8 production, bactericidal activity, and CD62L expression of granulocytes were evaluated. RESULTS Of note, we found that the two glycopeptide antibiotics dalbavancin and teicoplanin inhibited ROS production upon granulocyte activation via different signaling pathways in a dose-dependent manner. Dalbavancin also blocked the PMA-induced shedding of CD62L. In contrast, the oxazolidinone antibiotics tedizolid and linezolid had no effect on neutrophil function, while the combination of ceftazidime/avibactam dose dependently inhibited the fMLP/Cytochalasin B-induced granulocyte burst in a dose-dependent manner. Additionally, we showed that dalbavancin and teicoplanin as well as sulfametrole/trimethoprim and ceftazidime/avibactam inhibited baseline and PMA-induced IL-8 production by neutrophilic granulocytes. Moreover, dalbavancin impaired the bactericidal activity of neutrophilic granulocytes. CONCLUSION We here identified hitherto unknown inhibitory effects of several classes of antibiotics on the effector functions of neutrophilic granulocytes.
Collapse
Affiliation(s)
- Paul Ettel
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Vienna, Austria
| | - Al Nasar Ahmed Sehgal
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Vienna, Austria
| | - Nicole Harrison
- Medical University of Vienna, Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Vienna, Austria
| | - Winfried F Pickl
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Vienna, Austria
- Karl Landsteiner University of Health Sciences, Krems, Austria
| | | |
Collapse
|
15
|
Jiang H, Shin DH, Yi Y, Fan X, Gumin J, He J, Gillard AG, Lang FF, Gomez-Manzano C, Fueyo J. Adjuvant Therapy with Oncolytic Adenovirus Delta-24-RGDOX After Intratumoral Adoptive T-cell Therapy Promotes Antigen Spread to Sustain Systemic Antitumor Immunity. CANCER RESEARCH COMMUNICATIONS 2023; 3:1118-1131. [PMID: 37379361 PMCID: PMC10295804 DOI: 10.1158/2767-9764.crc-23-0054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/17/2023] [Accepted: 03/28/2023] [Indexed: 06/30/2023]
Abstract
Cancer cell heterogeneity and immunosuppressive tumor microenvironment (TME) pose a challenge in treating solid tumors with adoptive cell therapies targeting limited tumor-associated antigens (TAA), such as chimeric antigen receptor T-cell therapy. We hypothesize that oncolytic adenovirus Delta-24-RGDOX activates the TME and promote antigen spread to potentiate the abscopal effect of adoptive TAA-targeting T cells in localized intratumoral treatment. Herein, we used C57BL/6 mouse models with disseminated tumors derived from B16 melanoma cell lines to assess therapeutic effects and antitumor immunity. gp100-specific pmel-1 or ovalbumin (OVA)-specific OT-I T cells were injected into the first subcutaneous tumor, followed by three injections of Delta-24-RGDOX. We found TAA-targeting T cells injected into one subcutaneous tumor showed tumor tropism. Delta-24-RGDOX sustained the systemic tumor regression mediated by the T cells, leading to improved survival rate. Further analysis revealed that, in mice with disseminated B16-OVA tumors, Delta-24-RGDOX increased CD8+ leukocyte density within treated and untreated tumors. Importantly, Delta-24-RGDOX significantly reduced the immunosuppression of endogenous OVA-specific CTLs while increasing that of CD8+ leukocytes and, to a lesser extent, adoptive pmel-1 T cells. Consequently, Delta-24-RGDOX drastically increased the density of the OVA-specific CTLs in both tumors, and the combination synergistically enhanced the effect. Consistently, the splenocytes from the combination group showed a significantly stronger response against other TAAs (OVA and TRP2) than gp100, resulted in higher activity against tumor cells. Therefore, our data demonstrate that, as an adjuvant therapy followed TAA-targeting T cells in localized treatment, Delta-24-RGDOX activates TME and promotes antigen spread, leading to efficacious systemic antitumor immunity to overcome tumor relapse. Significance Adjuvant therapy with oncolytic viruses promotes antigen spread to potentiate localized intratumoral adoptive T-cell therapy with limited TAA targets, leading to sustainable systemic antitumor immunity to overcome tumor relapse.
Collapse
Affiliation(s)
- Hong Jiang
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dong Ho Shin
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yanhua Yi
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xuejun Fan
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joy Gumin
- Department of Neuro-Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jiasen He
- Pediatric division, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Andrew G. Gillard
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Frederick F. Lang
- Department of Neuro-Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Juan Fueyo
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
16
|
LeMaster C, Pierce SH, Geanes ES, Khanal S, Elliott SS, Scott AB, Louiselle DA, McLennan R, Maulik D, Lewis T, Pastinen T, Bradley T. The cellular and immunological dynamics of early and transitional human milk. Commun Biol 2023; 6:539. [PMID: 37202439 DOI: 10.1038/s42003-023-04910-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 05/03/2023] [Indexed: 05/20/2023] Open
Abstract
Human milk is essential for infant nutrition and immunity, providing protection against infections and other immune-mediated diseases during the lactation period and beyond in later childhood. Milk contains a broad range of bioactive factors such as nutrients, hormones, enzymes, immunoglobulins, growth factors, cytokines, and antimicrobial factors, as well as heterogeneous populations of maternal cells. The soluble and cellular components of milk are dynamic over time to meet the needs of the growing infant. In this study, we utilize systems-approaches to define and characterize 62 analytes of the soluble component, including immunoglobulin isotypes, as well as the cellular component of human milk during the first two weeks postpartum from 36 mothers. We identify soluble immune and growth factors that are dynamic over time and could be utilized to classify milk into different phenotypic groups. We identify 24 distinct populations of both epithelial and immune cells by single-cell transcriptome analysis of 128,016 human milk cells. We found that macrophage populations have shifting inflammatory profiles during the first two weeks of lactation. This analysis provides key insights into the soluble and cellular components of human milk and serves as a substantial resource for future studies of human milk.
Collapse
Affiliation(s)
- Cas LeMaster
- Genomic Medicine Center, Children's Mercy Research Institute, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Stephen H Pierce
- Genomic Medicine Center, Children's Mercy Research Institute, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Eric S Geanes
- Genomic Medicine Center, Children's Mercy Research Institute, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Santosh Khanal
- Genomic Medicine Center, Children's Mercy Research Institute, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Staci S Elliott
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Allison B Scott
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Daniel A Louiselle
- Genomic Medicine Center, Children's Mercy Research Institute, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Rebecca McLennan
- Genomic Medicine Center, Children's Mercy Research Institute, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Devika Maulik
- Fetal Health Center, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - Tamorah Lewis
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- Department of Pediatrics, UMKC School of Medicine, Kansas City, MO, 64108, USA
| | - Tomi Pastinen
- Genomic Medicine Center, Children's Mercy Research Institute, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
- Department of Pediatrics, UMKC School of Medicine, Kansas City, MO, 64108, USA
| | - Todd Bradley
- Genomic Medicine Center, Children's Mercy Research Institute, Children's Mercy Kansas City, Kansas City, MO, 64108, USA.
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
- Department of Pediatrics, UMKC School of Medicine, Kansas City, MO, 64108, USA.
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
17
|
Shahjin F, Patel M, Machhi J, Cohen JD, Nayan MU, Yeapuri P, Zhang C, Waight E, Hasan M, Abdelmoaty MM, Dash PK, Zhou Y, Andreu I, Gendelman HE, Kevadiya BD. Multipolymer microsphere delivery of SARS-CoV-2 antigens. Acta Biomater 2023; 158:493-509. [PMID: 36581007 PMCID: PMC9791794 DOI: 10.1016/j.actbio.2022.12.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/08/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022]
Abstract
Effective antigen delivery facilitates antiviral vaccine success defined by effective immune protective responses against viral exposures. To improve severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) antigen delivery, a controlled biodegradable, stable, biocompatible, and nontoxic polymeric microsphere system was developed for chemically inactivated viral proteins. SARS-CoV-2 proteins encapsulated in polymeric microspheres induced robust antiviral immunity. The viral antigen-loaded microsphere system can preclude the need for repeat administrations, highlighting its potential as an effective vaccine. STATEMENT OF SIGNIFICANCE: Successful SARS-CoV-2 vaccines were developed and quickly approved by the US Food and Drug Administration (FDA). However, each of the vaccines requires boosting as new variants arise. We posit that injectable biodegradable polymers represent a means for the sustained release of emerging viral antigens. The approach offers a means to reduce immunization frequency by predicting viral genomic variability. This strategy could lead to longer-lasting antiviral protective immunity. The current proof-of-concept multipolymer study for SARS-CoV-2 achieve these metrics.
Collapse
Affiliation(s)
- Farah Shahjin
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Milankumar Patel
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Jacob D Cohen
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Mohammad Ullah Nayan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Pravin Yeapuri
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Chen Zhang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Emiko Waight
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Mahmudul Hasan
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mai Mohamed Abdelmoaty
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Prasanta K Dash
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - You Zhou
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Irene Andreu
- RI Consortium of Nanoscience and Nanotechnology and Department of Chemical Engineering University of Rhode Island, RI, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA.
| | - Bhavesh D Kevadiya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| |
Collapse
|
18
|
Babl N, Hofbauer J, Matos C, Voll F, Menevse AN, Rechenmacher M, Mair R, Beckhove P, Herr W, Siska PJ, Renner K, Kreutz M, Schnell A. Low-density lipoprotein balances T cell metabolism and enhances response to anti-PD-1 blockade in a HCT116 spheroid model. Front Oncol 2023; 13:1107484. [PMID: 36776340 PMCID: PMC9911890 DOI: 10.3389/fonc.2023.1107484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023] Open
Abstract
Introduction The discovery of immune checkpoints and the development of their specific inhibitors was acclaimed as a major breakthrough in cancer therapy. However, only a limited patient cohort shows sufficient response to therapy. Hence, there is a need for identifying new checkpoints and predictive biomarkers with the objective of overcoming immune escape and resistance to treatment. Having been associated with both, treatment response and failure, LDL seems to be a double-edged sword in anti-PD1 immunotherapy. Being embedded into complex metabolic conditions, the impact of LDL on distinct immune cells has not been sufficiently addressed. Revealing the effects of LDL on T cell performance in tumor immunity may enable individual treatment adjustments in order to enhance the response to routinely administered immunotherapies in different patient populations. The object of this work was to investigate the effect of LDL on T cell activation and tumor immunity in-vitro. Methods Experiments were performed with different LDL dosages (LDLlow = 50 μg/ml and LDLhigh = 200 μg/ml) referring to medium control. T cell phenotype, cytokines and metabolism were analyzed. The functional relevance of our findings was studied in a HCT116 spheroid model in the context of anti-PD-1 blockade. Results The key points of our findings showed that LDLhigh skewed the CD4+ T cell subset into a central memory-like phenotype, enhanced the expression of the co-stimulatory marker CD154 (CD40L) and significantly reduced secretion of IL-10. The exhaustion markers PD-1 and LAG-3 were downregulated on both T cell subsets and phenotypical changes were associated with a balanced T cell metabolism, in particular with a significant decrease of reactive oxygen species (ROS). T cell transfer into a HCT116 spheroid model resulted in a significant reduction of the spheroid viability in presence of an anti-PD-1 antibody combined with LDLhigh. Discussion Further research needs to be conducted to fully understand the impact of LDL on T cells in tumor immunity and moreover, to also unravel LDL effects on other lymphocytes and myeloid cells for improving anti-PD-1 immunotherapy. The reason for improved response might be a resilient, less exhausted phenotype with balanced ROS levels.
Collapse
Affiliation(s)
- Nathalie Babl
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Joshua Hofbauer
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Carina Matos
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Florian Voll
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany,Division of Interventional Immunology, Leibniz Institute for Immunotherapy (LIT), Regensburg, Germany
| | - Ayse Nur Menevse
- Division of Interventional Immunology, Leibniz Institute for Immunotherapy (LIT), Regensburg, Germany
| | - Michael Rechenmacher
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Ruth Mair
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Philipp Beckhove
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany,Division of Interventional Immunology, Leibniz Institute for Immunotherapy (LIT), Regensburg, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Peter J. Siska
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Kathrin Renner
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Marina Kreutz
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany,Division of Interventional Immunology, Leibniz Institute for Immunotherapy (LIT), Regensburg, Germany
| | - Annette Schnell
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany,*Correspondence: Annette Schnell,
| |
Collapse
|
19
|
Ten FW, Yuan D, Jabareen N, Phua YJ, Eils R, Lukassen S, Conrad C. resVAE ensemble: Unsupervised identification of gene sets in multi-modal single-cell sequencing data using deep ensembles. Front Cell Dev Biol 2023; 11:1091047. [PMID: 36875765 PMCID: PMC9975353 DOI: 10.3389/fcell.2023.1091047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/24/2023] [Indexed: 02/17/2023] Open
Abstract
Feature identification and manual inspection is currently still an integral part of biological data analysis in single-cell sequencing. Features such as expressed genes and open chromatin status are selectively studied in specific contexts, cell states or experimental conditions. While conventional analysis methods construct a relatively static view on gene candidates, artificial neural networks have been used to model their interactions after hierarchical gene regulatory networks. However, it is challenging to identify consistent features in this modeling process due to the inherently stochastic nature of these methods. Therefore, we propose using ensembles of autoencoders and subsequent rank aggregation to extract consensus features in a less biased manner. Here, we performed sequencing data analyses of different modalities either independently or simultaneously as well as with other analysis tools. Our resVAE ensemble method can successfully complement and find additional unbiased biological insights with minimal data processing or feature selection steps while giving a measurement of confidence, especially for models using stochastic or approximation algorithms. In addition, our method can also work with overlapping clustering identity assignment suitable for transitionary cell types or cell fates in comparison to most conventional tools.
Collapse
Affiliation(s)
- Foo Wei Ten
- Center for Digital Health, Berlin Institute of Health (BIH) at Charité-Universitatsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dongsheng Yuan
- Center for Digital Health, Berlin Institute of Health (BIH) at Charité-Universitatsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Neurology with Experimental Neurology, Berlin, Germany
| | - Nabil Jabareen
- Center for Digital Health, Berlin Institute of Health (BIH) at Charité-Universitatsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Yin Jun Phua
- Department of Computer Science, Tokyo Institute of Technology, Tokyo, Japan
| | - Roland Eils
- Center for Digital Health, Berlin Institute of Health (BIH) at Charité-Universitatsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Health Data Science Unit, Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
| | - Sören Lukassen
- Center for Digital Health, Berlin Institute of Health (BIH) at Charité-Universitatsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christian Conrad
- Center for Digital Health, Berlin Institute of Health (BIH) at Charité-Universitatsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
20
|
Attreed SE, Silva C, Abbott S, Ramirez-Medina E, Espinoza N, Borca MV, Gladue DP, Diaz-San Segundo F. A Highly Effective African Swine Fever Virus Vaccine Elicits a Memory T Cell Response in Vaccinated Swine. Pathogens 2022; 11:pathogens11121438. [PMID: 36558773 PMCID: PMC9783822 DOI: 10.3390/pathogens11121438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/17/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
African Swine Fever Virus (ASFV) is the causative agent of a highly contagious and lethal vector-borne disease in suids. Recently, a live attenuated virus strain, developed using the currently circulating, virulent Georgia strain (ASFV-G) with a single gene deletion (ASFV-G-ΔI177L), resulted in an effective vaccine. Nevertheless, protective immune response mechanisms induced by this candidate are poorly understood. In this study, Yorkshire crossbred swine intramuscularly vaccinated with 106 50% hemadsorption dose (HAD50) of ASFV-G-ΔI177L or a vehicle control were challenged at 28 days post-inoculation (dpi) with 102 HAD50 of ASFV-G. Analysis of purified peripheral blood mononuclear cells following inoculation and challenge revealed that CD4+, CD8+ and CD4+CD8+ central memory T cells (CD44+CD25-CD27-CD62L+CCR7+, Tcm) decreased significantly by 28 dpi in ASFV-G-ΔI177L-vaccinated swine compared to baseline and time-matched controls. Conversely, CD4+, CD8+ and CD4+CD8+ effector memory T cells (CD44+CD25-CD27-CD62-CCR7-, Tem) increased significantly among ASFV-G-ΔI177L-vaccined swine by 28 dpi compared to baseline and time-matched controls. Additionally, the percentage of natural killer (NK), CD4+ and CD4+CD8+ Tem and CD8+ Tcm and Tem positive for IFNγ increased significantly following inoculation, surpassing that of controls by 28 dpi or earlier. These results suggest that NK and memory T cells play a role in protective immunity and suggest that studying these cell populations may be a surrogate immunity marker in ASF vaccination.
Collapse
Affiliation(s)
- Sarah E. Attreed
- U.S. Department of Agriculture, Agricultural Research Service, Plum Island Animal Disease Center, Greenport, New York, NY 11944, USA
- PIADC Research Participation Program, Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN 37830, USA
| | - Christina Silva
- U.S. Department of Agriculture, Agricultural Research Service, Plum Island Animal Disease Center, Greenport, New York, NY 11944, USA
| | - Sophia Abbott
- U.S. Department of Agriculture, Agricultural Research Service, Plum Island Animal Disease Center, Greenport, New York, NY 11944, USA
| | - Elizabeth Ramirez-Medina
- U.S. Department of Agriculture, Agricultural Research Service, Plum Island Animal Disease Center, Greenport, New York, NY 11944, USA
| | - Nallely Espinoza
- U.S. Department of Agriculture, Agricultural Research Service, Plum Island Animal Disease Center, Greenport, New York, NY 11944, USA
| | - Manuel V. Borca
- U.S. Department of Agriculture, Agricultural Research Service, Plum Island Animal Disease Center, Greenport, New York, NY 11944, USA
- Correspondence: (M.V.B.); (D.P.G.); (F.D.-S.S.); Tel.: +1-(631)-323-3131 (M.V.B.); +1-(631)-323-3035 (D.P.G.); +1-(631)-323-3012 (F.D.-S.S.)
| | - Douglas P. Gladue
- U.S. Department of Agriculture, Agricultural Research Service, Plum Island Animal Disease Center, Greenport, New York, NY 11944, USA
- Correspondence: (M.V.B.); (D.P.G.); (F.D.-S.S.); Tel.: +1-(631)-323-3131 (M.V.B.); +1-(631)-323-3035 (D.P.G.); +1-(631)-323-3012 (F.D.-S.S.)
| | - Fayna Diaz-San Segundo
- U.S. Department of Agriculture, Agricultural Research Service, Plum Island Animal Disease Center, Greenport, New York, NY 11944, USA
- Correspondence: (M.V.B.); (D.P.G.); (F.D.-S.S.); Tel.: +1-(631)-323-3131 (M.V.B.); +1-(631)-323-3035 (D.P.G.); +1-(631)-323-3012 (F.D.-S.S.)
| |
Collapse
|
21
|
Cartagena García C, Balandraud N, Roudier J, Lafforgue P, Lambert N, Busnel JM. Leveraging whole blood based functional flow cytometry assays to open new perspectives for rheumatoid arthritis translational research. Sci Rep 2022; 12:12166. [PMID: 35842449 PMCID: PMC9288473 DOI: 10.1038/s41598-022-16622-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 07/12/2022] [Indexed: 11/09/2022] Open
Abstract
Despite introduction of biological disease modifying anti-rheumatic drugs (DMARDs) for Rheumatoid arthritis (RA) treatment, therapeutic strategies do not always lead to disease control and remission. Hence, a more efficient patient stratification and monitoring biomarkers and tools are needed to enable a more personalized medicine. We used a whole blood based functional flow cytometry assay to characterize immune cells from RA patients (treated or not), healthy donors and psoriatic arthritis (PsA) patients according to their responses to LPS and/or anti-TNFα (infliximab, IFX). Activation marker expression was measured using a 10-color flow cytometry panel following a no-wash protocol. Naïve-to-treatment RA patients had a stronger inflammatory profile in comparison to healthy donors at basal level. Higher expression of activation markers (CD69 and/or CD11b) on NK, B cells and granulocytes and lower expression of the adhesion molecule CD62L were measured on monocytes, granulocytes and B cells. After LPS, naïve RA patients' cells were less capable of regulating CD69, CD11b, CD16 or CD62L showing impaired activation capabilities. Upon LPS and IFX co-incubation, hierarchical clustering analysis showed different profiles between cohorts. We believe that this whole blood-based approach should further be assessed for RA patient characterization as it provides new perspectives for stratification and/or monitoring.
Collapse
Affiliation(s)
- Celia Cartagena García
- Research Department, Beckman Coulter Life Sciences, Marseille, France.,INSERM UMRs 1097, Aix Marseille University, Marseille, France
| | - Nathalie Balandraud
- INSERM UMRs 1097, Aix Marseille University, Marseille, France.,AP-HM, Rheumatology, Sainte Marguerite Hospital, 13014, Marseille, France
| | - Jean Roudier
- INSERM UMRs 1097, Aix Marseille University, Marseille, France.,AP-HM, Rheumatology, Sainte Marguerite Hospital, 13014, Marseille, France
| | - Pierre Lafforgue
- AP-HM, Rheumatology, Sainte Marguerite Hospital, 13014, Marseille, France
| | | | - Jean-Marc Busnel
- Research Department, Beckman Coulter Life Sciences, Marseille, France.
| |
Collapse
|
22
|
Cristofoletti C, Bresin A, Fioretti M, Russo G, Narducci MG. Combined High-Throughput Approaches Reveal the Signals Driven by Skin and Blood Environments and Define the Tumor Heterogeneity in Sézary Syndrome. Cancers (Basel) 2022; 14:cancers14122847. [PMID: 35740513 PMCID: PMC9221051 DOI: 10.3390/cancers14122847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Sézary syndrome (SS) is a leukemic and incurable variant of cutaneous T-cell lymphoma characterized by the accumulation of neoplastic CD4+ lymphocytes in the blood, lymph nodes, and skin. With the exception of allogenic transplantation, no curative chance is available to treat SS, and it is a priority to find new therapies that target SS cells within all disease compartments. This review aims to summarize the more recent analyses conducted on skin- and blood-derived SS cells concurrently obtained from the same SS patients. The results highlighted that skin-SS cells were more active/proliferating with respect to matched blood SS cells that instead appeared quiescent. These data shed the light on the possibility to treat blood and skin SS cells with different compounds, respectively. Moreover, this review recaps the more recent findings on the heterogeneity of circulating SS cells that presented a series of novel markers that could improve diagnosis, prognosis and therapy of this lymphoma. Abstract Sézary syndrome (SS) is an aggressive variant of cutaneous t-cell lymphoma characterized by the accumulation of neoplastic CD4+ lymphocytes—the SS cells—mainly in blood, lymph nodes, and skin. The tumor spread pattern of SS makes this lymphoma a unique model of disease that allows a concurrent blood and skin sampling for analysis. This review summarizes the recent studies highlighting the transcriptional programs triggered by the crosstalk between SS cells and blood–skin microenvironments. Emerging data proved that skin-derived SS cells show consistently higher activation/proliferation rates, mainly driven by T-cell receptor signaling with respect to matched blood SS cells that instead appear quiescent. Biochemical analyses also demonstrated an hyperactivation of PI3K/AKT/mTOR, a targetable pathway by multiple inhibitors currently in clinical trials, in skin SS cells compared with a paired blood counterpart. These results indicated that active and quiescent SS cells coexist in this lymphoma, and that they could be respectively treated with different therapeutics. Finally, this review underlines the more recent discoveries into the heterogeneity of circulating SS cells, highlighting a series of novel markers that could improve the diagnosis and that represent novel therapeutic targets (GPR15, PTPN13, KLRB1, and ITGB1) as well as new genetic markers (PD-1 and CD39) able to stratify SS patients for disease aggressiveness.
Collapse
|
23
|
Kokkinou E, Pandey RV, Mazzurana L, Gutierrez-Perez I, Tibbitt CA, Weigel W, Soini T, Carrasco A, Rao A, Nagasawa M, Bal SM, Jangard M, Friberg D, Lindforss U, Nordenvall C, Ljunggren M, Haapaniemi S, Keita ÅV, Söderholm J, Hedin C, Spits H, Bryceson YT, Mjösberg J. CD45RA +CD62L - ILCs in human tissues represent a quiescent local reservoir for the generation of differentiated ILCs. Sci Immunol 2022; 7:eabj8301. [PMID: 35427178 DOI: 10.1126/sciimmunol.abj8301] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Innate lymphoid cells (ILCs) are highly plastic and predominantly mucosal tissue-resident cells that contribute to both homeostasis and inflammation depending on the microenvironment. The discovery of naïve-like ILCs suggests an ILC differentiation process that is akin to naïve T cell differentiation. Delineating the mechanisms that underlie ILC differentiation in tissues is crucial for understanding ILC biology in health and disease. Here, we showed that tonsillar ILCs expressing CD45RA lacked proliferative activity, indicative of cellular quiescence. CD62L distinguished two subsets of CD45RA+ ILCs. CD45RA+CD62L+ ILCs (CD62L+ ILCs) resembled circulating naïve ILCs because they lacked the transcriptional, metabolic, epigenetic, and cytokine production signatures of differentiated ILCs. CD45RA+CD62L- ILCs (CD62L- ILCs) were epigenetically similar to CD62L+ ILCs but showed a transcriptional, metabolic, and cytokine production signature that was more akin to differentiated ILCs. CD62L+ and CD62L- ILCs contained uni- and multipotent precursors of ILC1s/NK cells and ILC3s. Differentiation of CD62L+ and CD62L- ILCs led to metabolic reprogramming including up-regulation of genes associated with glycolysis, which was needed for their effector functions after differentiation. CD62L- ILCs with preferential differentiation capacity toward IL-22-producing ILC3s accumulated in the inflamed mucosa of patients with inflammatory bowel disease. These data suggested distinct differentiation potential of CD62L+ and CD62L- ILCs between tissue microenvironments and identified that manipulation of these cells is a possible approach to restore tissue-immune homeostasis.
Collapse
Affiliation(s)
- Efthymia Kokkinou
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Ram Vinay Pandey
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Luca Mazzurana
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Irene Gutierrez-Perez
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Christopher Andrew Tibbitt
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Whitney Weigel
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Tea Soini
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Anna Carrasco
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Anna Rao
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Maho Nagasawa
- Department of Experimental Immunology, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Suzanne M Bal
- Department of Experimental Immunology, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Mattias Jangard
- ENT Unit, Sophiahemmet University Research Laboratory and Sophiahemmet Hospital, Stockholm, Sweden
| | - Danielle Friberg
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Ulrik Lindforss
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Department of Pelvic Cancer, GI Oncology and Colorectal Surgery Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Caroline Nordenvall
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Department of Pelvic Cancer, GI Oncology and Colorectal Surgery Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Malin Ljunggren
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Department of Pelvic Cancer, GI Oncology and Colorectal Surgery Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Staffan Haapaniemi
- Department of Surgery, Vrinnevi Hospital, Norrköping, Sweden.,Department of Surgery, Linköping University, Linköping, Sweden
| | - Åsa V Keita
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johan Söderholm
- Department of Surgery, Linköping University, Linköping, Sweden.,Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Charlotte Hedin
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Gastroenterology Unit, Department of Gastroenterology, Dermatovenereology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Hergen Spits
- Department of Experimental Immunology, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Yenan T Bryceson
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Jenny Mjösberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
24
|
Kwon H, Schafer JM, Song NJ, Kaneko S, Li A, Xiao T, Ma A, Allen C, Das K, Zhou L, Riesenberg B, Chang Y, Weltge P, Velegraki M, Oh DY, Fong L, Ma Q, Sundi D, Chung D, Li X, Li Z. Androgen conspires with the CD8 + T cell exhaustion program and contributes to sex bias in cancer. Sci Immunol 2022; 7:eabq2630. [PMID: 35420889 DOI: 10.1126/sciimmunol.abq2630] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Sex bias exists in the development and progression of non-reproductive organ cancers, but the underlying mechanisms are enigmatic. Studies so far have focused largely on sexual dimorphisms in cancer biology and socioeconomic factors. Here, we establish a role for CD8+ T cell-dependent anti-tumor immunity in mediating sex differences in tumor aggressiveness, which is driven by the gonadal androgen but not sex chromosomes. A male bias exists in the frequency of intratumoral antigen-experienced Tcf7/TCF1+ progenitor exhausted CD8+ T cells that are devoid of effector activity as a consequence of intrinsic androgen receptor (AR) function. Mechanistically, we identify a novel sex-specific regulon in progenitor exhausted CD8+ T cells and a pertinent contribution from AR as a direct transcriptional trans-activator of Tcf7/TCF1. The T cell intrinsic function of AR in promoting CD8+ T cell exhaustion in vivo was established using multiple approaches including loss-of-function studies with CD8-specific Ar knockout mice. Moreover, ablation of the androgen-AR axis rewires the tumor microenvironment to favor effector T cell differentiation and potentiates the efficacy of anti-PD-1 immune checkpoint blockade. Collectively, our findings highlight androgen-mediated promotion of CD8+ T cell dysfunction in cancer and imply broader opportunities for therapeutic development from understanding sex disparities in health and disease.
Collapse
Affiliation(s)
- Hyunwoo Kwon
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center - The James, Columbus, OH 43210, USA.,Medical Scientist Training Program, College of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Johanna M Schafer
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center - The James, Columbus, OH 43210, USA
| | - No-Joon Song
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center - The James, Columbus, OH 43210, USA
| | - Satoshi Kaneko
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anqi Li
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center - The James, Columbus, OH 43210, USA
| | - Tong Xiao
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center - The James, Columbus, OH 43210, USA
| | - Anjun Ma
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center - The James, Columbus, OH 43210, USA.,Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - Carter Allen
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center - The James, Columbus, OH 43210, USA.,Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - Komal Das
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center - The James, Columbus, OH 43210, USA
| | - Lei Zhou
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center - The James, Columbus, OH 43210, USA
| | - Brian Riesenberg
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center - The James, Columbus, OH 43210, USA
| | - Yuzhou Chang
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center - The James, Columbus, OH 43210, USA.,Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - Payton Weltge
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center - The James, Columbus, OH 43210, USA
| | - Maria Velegraki
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center - The James, Columbus, OH 43210, USA
| | - David Y Oh
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lawrence Fong
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.,Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Qin Ma
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - Debasish Sundi
- Department of Urology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Dongjun Chung
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center - The James, Columbus, OH 43210, USA.,Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA
| | - Xue Li
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Samuel Oschin Comprehensive Cancer Institute, Departments of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Zihai Li
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center - The James, Columbus, OH 43210, USA
| |
Collapse
|
25
|
Rao R, Shah S, Bhattacharya D, Toukam DK, Cáceres R, Pomeranz Krummel DA, Sengupta S. Ligand-Gated Ion Channels as Targets for Treatment and Management of Cancers. Front Physiol 2022; 13:839437. [PMID: 35350689 PMCID: PMC8957973 DOI: 10.3389/fphys.2022.839437] [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: 12/20/2021] [Accepted: 02/07/2022] [Indexed: 12/24/2022] Open
Abstract
Ligand-gated ion channels are an ionotropic receptor subtype characterized by the binding of an extracellular ligand, followed by the transient passage of ions through a transmembrane pore. Ligand-gated ion channels are commonly subcategorized into three superfamilies: purinoreceptors, glutamate receptors, and Cys-loop receptors. This classification is based on the differing topographical morphology of the receptors, which in turn confers functional differences. Ligand-gated ion channels have a diverse spatial and temporal expression which implicate them in key cellular processes. Given that the transcellular electrochemical gradient is finely tuned in eukaryotic cells, any disruption in this homeostasis can contribute to aberrancies, including altering the activity of pro-tumorigenic molecular pathways, such as the MAPK/ERK, RAS, and mTOR pathways. Ligand-gated ion channels therefore serve as a potential targetable system for cancer therapeutics. In this review, we analyze the role that each of the three ligand-gated ion channel superfamilies has concerning tumor proliferation and as a target for the treatment of cancer symptomatology.
Collapse
Affiliation(s)
| | | | | | | | | | - Daniel A. Pomeranz Krummel
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Soma Sengupta
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
26
|
Meldgaard TS, Blengio F, Maffione D, Sammicheli C, Tavarini S, Nuti S, Kratzer R, Medini D, Siena E, Bertholet S. Single-Cell Analysis of Antigen-Specific CD8+ T-Cell Transcripts Reveals Profiles Specific to mRNA or Adjuvanted Protein Vaccines. Front Immunol 2021; 12:757151. [PMID: 34777370 PMCID: PMC8586650 DOI: 10.3389/fimmu.2021.757151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/05/2021] [Indexed: 12/29/2022] Open
Abstract
CD8+ T cells play a key role in mediating protective immunity after immune challenges such as infection or vaccination. Several subsets of differentiated CD8+ T cells have been identified, however, a deeper understanding of the molecular mechanism that underlies T-cell differentiation is lacking. Conventional approaches to the study of immune responses are typically limited to the analysis of bulk groups of cells that mask the cells' heterogeneity (RNA-seq, microarray) and to the assessment of a relatively limited number of biomarkers that can be evaluated simultaneously at the population level (flow and mass cytometry). Single-cell analysis, on the other hand, represents a possible alternative that enables a deeper characterization of the underlying cellular heterogeneity. In this study, a murine model was used to characterize immunodominant hemagglutinin (HA533-541)-specific CD8+ T-cell responses to nucleic- and protein-based influenza vaccine candidates, using single-cell sorting followed by transcriptomic analysis. Investigation of single-cell gene expression profiles enabled the discovery of unique subsets of CD8+ T cells that co-expressed cytotoxic genes after vaccination. Moreover, this method enabled the characterization of antigen specific CD8+ T cells that were previously undetected. Single-cell transcriptome profiling has the potential to allow for qualitative discrimination of cells, which could lead to novel insights on biological pathways involved in cellular responses. This approach could be further validated and allow for more informed decision making in preclinical and clinical settings.
Collapse
Affiliation(s)
- Trine Sundebo Meldgaard
- Research & Development, GSK, Siena, Italy
- Biochemistry & Molecular Biology, University of Siena, Siena, Italy
| | - Fabiola Blengio
- Chemical & Biological Sciences, University of Torino, Torino, Italy
| | - Denise Maffione
- Chemical & Biological Sciences, University of Torino, Torino, Italy
| | | | | | - Sandra Nuti
- Research & Development, GSK, Siena, Italy
- Research & Development, GSK, Rockville, MD, United States
| | | | | | | | - Sylvie Bertholet
- Research & Development, GSK, Siena, Italy
- Research & Development, GSK, Rockville, MD, United States
| |
Collapse
|
27
|
Herrera A, Cheng A, Mimitou EP, Seffens A, George D, Bar-Natan M, Heguy A, Ruggles KV, Scher JU, Hymes K, Latkowski JA, Ødum N, Kadin ME, Ouyang Z, Geskin LJ, Smibert P, Buus TB, Koralov SB. Multimodal single-cell analysis of cutaneous T-cell lymphoma reveals distinct subclonal tissue-dependent signatures. Blood 2021; 138:1456-1464. [PMID: 34232982 PMCID: PMC8532199 DOI: 10.1182/blood.2020009346] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 04/03/2021] [Indexed: 11/20/2022] Open
Abstract
Cutaneous T-cell lymphoma (CTCL) is a heterogeneous group of mature T-cell neoplasms characterized by the accumulation of clonal malignant CD4+ T cells in the skin. The most common variant of CTCL, mycosis fungoides (MF ), is confined to the skin in early stages but can be accompanied by extracutaneous dissemination of malignant T cells to the blood and lymph nodes in advanced stages of disease. Sézary syndrome (SS), a leukemic form of disease, is characterized by significant blood involvement. Little is known about the transcriptional and genomic relationship between skin- and blood-residing malignant T cells in CTCL. To identify and interrogate malignant clones in matched skin and blood from patients with leukemic MF and SS, we combine T-cell receptor clonotyping with quantification of gene expression and cell surface markers at the single cell level. Our data reveal clonal evolution at a transcriptional and genetic level within the malignant populations of individual patients. We highlight highly consistent transcriptional signatures delineating skin- and blood-derived malignant T cells. Analysis of these 2 populations suggests that environmental cues, along with genetic aberrations, contribute to transcriptional profiles of malignant T cells. Our findings indicate that the skin microenvironment in CTCL promotes a transcriptional response supporting rapid malignant expansion, as opposed to the quiescent state observed in the blood, potentially influencing efficacy of therapies. These results provide insight into tissue-specific characteristics of cancerous cells and underscore the need to address the patients' individual malignant profiles at the time of therapy to eliminate all subclones.
Collapse
Affiliation(s)
- Alberto Herrera
- Department of Pathology, New York University School of Medicine, New York, NY
| | - Anthony Cheng
- Department of Genetic and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA
| | - Eleni P Mimitou
- Technology Innovation Laboratory, New York Genome Center, New York, NY
| | - Angelina Seffens
- Department of Pathology, New York University School of Medicine, New York, NY
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Dean George
- Department of Dermatology, Boston University and Roger Williams Medical Center, Brown University, Providence, RI
| | - Michal Bar-Natan
- Department of Pathology, New York University School of Medicine, New York, NY
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Adriana Heguy
- Department of Pathology, New York University School of Medicine, New York, NY
- Genome Technology Center, New York University School of Medicine, New York, NY
| | | | - Jose U Scher
- Division of Rheumatology, Department of Medicine
| | | | - Jo-Ann Latkowski
- Department of Dermatology, New York University School of Medicine, New York, NY
| | - Niels Ødum
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Marshall E Kadin
- Department of Dermatology, Boston University and Roger Williams Medical Center, Brown University, Providence, RI
| | - Zhengqing Ouyang
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA
| | - Larisa J Geskin
- Department of Dermatology, Columbia University, New York, NY
| | - Peter Smibert
- Technology Innovation Laboratory, New York Genome Center, New York, NY
| | - Terkild B Buus
- Department of Pathology, New York University School of Medicine, New York, NY
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Sergei B Koralov
- Department of Pathology, New York University School of Medicine, New York, NY
| |
Collapse
|
28
|
Segura J, He B, Ireland J, Zou Z, Shen T, Roth G, Sun PD. The Role of L-Selectin in HIV Infection. Front Microbiol 2021; 12:725741. [PMID: 34659153 PMCID: PMC8511817 DOI: 10.3389/fmicb.2021.725741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/27/2021] [Indexed: 11/20/2022] Open
Abstract
HIV envelope glycoprotein is the most heavily glycosylated viral protein complex identified with over 20 glycans on its surface. This glycan canopy is thought to primarily shield the virus from host immune recognition as glycans are poor immunogens in general, however rare HIV neutralizing antibodies nevertheless potently recognize the glycan epitopes. While CD4 and chemokine receptors have been known as viral entry receptor and coreceptor, for many years the role of viral glycans in HIV entry was controversial. Recently, we showed that HIV envelope glycan binds to L-selectin in solution and on CD4 T lymphocytes. The viral glycan and L-selectin interaction functions to facilitate the viral adhesion and entry. Upon entry, infected CD4 T lymphocytes are stimulated to progressively shed L-selectin and suppressing this lectin receptor shedding greatly reduced HIV viral release and caused aggregation of diminutive virus-like particles within experimental infections and from infected primary T lymphocytes derived from both viremic and aviremic individuals. As shedding of L-selectin is mediated by ADAM metalloproteinases downstream of host-cell stimulation, these findings showed a novel mechanism for HIV viral release and offer a potential new class of anti-HIV compounds.
Collapse
Affiliation(s)
- Jason Segura
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Biao He
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Joanna Ireland
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Zhongcheng Zou
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Thomas Shen
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Gwynne Roth
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Peter D Sun
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
29
|
Kremsreiter SM, Kroell ASH, Weinberger K, Boehm H. Glycan-Lectin Interactions in Cancer and Viral Infections and How to Disrupt Them. Int J Mol Sci 2021; 22:10577. [PMID: 34638920 PMCID: PMC8508825 DOI: 10.3390/ijms221910577] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023] Open
Abstract
Glycan-lectin interactions play an essential role in different cellular processes. One of their main functions is involvement in the immune response to pathogens or inflammation. However, cancer cells and viruses have adapted to avail themselves of these interactions. By displaying specific glycosylation structures, they are able to bind to lectins, thus promoting pathogenesis. While glycan-lectin interactions promote tumor progression, metastasis, and/or chemoresistance in cancer, in viral infections they are important for viral entry, release, and/or immune escape. For several years now, a growing number of investigations have been devoted to clarifying the role of glycan-lectin interactions in cancer and viral infections. Various overviews have already summarized and highlighted their findings. In this review, we consider the interactions of the lectins MGL, DC-SIGN, selectins, and galectins in both cancer and viral infections together. A possible transfer of ways to target and disrupt them might lead to new therapeutic approaches in different pathological backgrounds.
Collapse
Affiliation(s)
- Stefanie Maria Kremsreiter
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Ruprecht Karls University Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany; (S.M.K.); (A.-S.H.K.); (K.W.)
| | - Ann-Sophie Helene Kroell
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Ruprecht Karls University Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany; (S.M.K.); (A.-S.H.K.); (K.W.)
| | - Katharina Weinberger
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Ruprecht Karls University Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany; (S.M.K.); (A.-S.H.K.); (K.W.)
| | - Heike Boehm
- Max-Planck-Institute for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
| |
Collapse
|
30
|
MacFarlane AW, Yeung HM, Alpaugh RK, Dulaimi E, Engstrom PF, Dasari A, Campbell KS, Vijayvergia N. Impacts of pembrolizumab therapy on immune phenotype in patients with high-grade neuroendocrine neoplasms. Cancer Immunol Immunother 2021; 70:1893-1906. [PMID: 33398390 PMCID: PMC8195815 DOI: 10.1007/s00262-020-02811-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023]
Abstract
High grade neuroendocrine neoplasms (G3 NENs) are rare aggressive tumors with limited treatment options. Twenty-one previously treated patients with metastatic extra-pulmonary G3 NENs were treated with pembrolizumab. Baseline tumor samples were assessed for PD-L1 and tumor infiltrating lymphocytes (TIL). Peripheral blood samples drawn pre-treatment, prior to cycle three, and at disease progression were analyzed by flow cytometry. One patient achieved partial response, two had stable disease, and 18 exhibited progressive disease. The partially responding patient did not progress after 392 days, and the median progression-free survival (PFS) was 59 days. Longer PFS correlated independently with higher pre-treatment peripheral blood T-cell counts and lower pre-treatment activation state (CD69 expression) of naïve T cells and NK cells. Peripheral T-cell viability was reduced in patients with greater TILs. Post-treatment, T cells had reduced numbers of CD4+ cells, reduced PD-1 expression, increased activation of effector (CD62L-) cells, and increased expression of TIGIT. Baseline TIGIT expression on peripheral T cells also correlated positively with Ki67 in tumor. Patients with higher baseline T-cell expression of TIM-3 had shorter PFS. Despite limited activity of pembrolizumab, this study highlights the immune phenotype in this rare tumor type before and after treatment. High baseline peripheral T-cell count and reduced activation of T and NK cell subsets were associated with improved outcomes. Furthermore, increased post-treatment TIGIT and elevated baseline TIM-3 expression suggest that these may limit the efficacy of pembrolizumab, providing a rationale for combination immunotherapy (PD-1 with TIGIT and/or TIM-3 antibodies) to treat extra-pulmonary G3 NENs.
Collapse
Affiliation(s)
- Alexander W MacFarlane
- Blood Cell Development and Function Program, Institute for Cancer Research, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Ho-Man Yeung
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - R Katherine Alpaugh
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Essel Dulaimi
- Department of Pathology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Paul F Engstrom
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Arvind Dasari
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kerry S Campbell
- Blood Cell Development and Function Program, Institute for Cancer Research, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA.
| | - Namrata Vijayvergia
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA.
| |
Collapse
|
31
|
Liao HQ, Han MT, Cheng W, Zhang C, Li H, Li MQ, Zhu R. Decidual-derived RANKL facilitates macrophages accumulation and residence at the maternal-fetal interface in human early pregnancy. Am J Reprod Immunol 2021; 86:e13406. [PMID: 33629434 DOI: 10.1111/aji.13406] [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] [Received: 12/11/2020] [Revised: 01/13/2021] [Accepted: 02/22/2021] [Indexed: 01/18/2023] Open
Abstract
PROBLEM During the first trimester, the accumulation of macrophages, which is the second largest decidual leukocyte population (~20%) at the maternal-fetal interface, is quite vital for a successful pregnancy, including embryo implantation, trophoblast invasion, and vascular remodeling. The mechanism of the enrichment and redistribution of macrophages in the uterine decidua of early pregnancy is largely unclear. METHOD OF STUDY A total of 37 women with normal early pregnancies were included. Primary decidual macrophages (dMφs) (n = 37) and primary decidual stromal cells (DSCs) (n = 37) were isolated, and the adhesion molecules were analyzed by flow cytometry (FCM). Adhesive experiment was carried out to evaluate the adhesion capacity by counting cell numbers of dMφs adhered to DSCs in a co-culture system. RESULTS We found that RANK+ dMφs was the dominating subtype at the maternal-fetal interface. The expression of adhesion molecules (eg, CD29, CD31, CD54, and CD62L) on the surface of RANK+ dMφs was higher than that of RANK- dMφs. After co-culture with DSCs, the expression of adhesion molecules on dMφs was up-regulated in a RANKL-dependent manner. Meanwhile, dMφs promoted the releasing of RANKL on DSCs after co-culture. Consistently, dMφs exhibited the lessoned capacity of adhesion to DSCs when blocking the crosstalk of RANKL-RANK between the DSCs and dMφs in vitro. CONCLUSION These results suggest that the interaction of RANKL-RANK up-regulates the expression of adhesion molecules on the surface of dMφs, contributing to the accumulation and residence of dMφs in human early pregnancy.
Collapse
Affiliation(s)
- Hai-Qiong Liao
- Center for Human Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Mu-Tian Han
- Center for Human Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Wei Cheng
- Center for Human Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Ce Zhang
- Center for Human Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Hong Li
- Center for Human Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Rui Zhu
- Center for Human Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
32
|
Rückert M, Deloch L, Frey B, Schlücker E, Fietkau R, Gaipl US. Combinations of Radiotherapy with Vaccination and Immune Checkpoint Inhibition Differently Affect Primary and Abscopal Tumor Growth and the Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13040714. [PMID: 33572437 PMCID: PMC7916259 DOI: 10.3390/cancers13040714] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 12/11/2022] Open
Abstract
Radiotherapy (RT) is known to have immune-modulatory properties. We hypothesized that RT and inactivated whole tumor cell vaccines generated with high hydrostatic pressure (HHP) synergize to retard the tumor growth which can be additionally improved with anti-PD-1 treatment. In abscopal tumor models, we injected mice with B16-F10 melanoma or TS/A mammary tumors. To evaluate the efficiency of RT in combination with HHP vaccines, we locally irradiated only one tumor with 2 × 8 Gy or 3 × 8 Gy. HHP vaccines further retarded the growth of locally irradiated (2 × 8 Gy) tumors. However, HHP vaccination combined with RT failed to induce abscopal anti-tumor immune responses, namely those to non-irradiated tumors, and even partly abrogated those which were induced with RT plus anti-PD-1. In the latter group, the abscopal effects were accompanied by an elevated infiltration of CD8+ T cells, monocytes/macrophages, and dendritic cells. 3 × 8 Gy failed to induce abscopal effects in association with increased expression of immunosuppressive checkpoint molecules compared to 2 × 8 Gy. We conclude that HHP vaccines induce anti-tumor effects, but only if the tumor microenvironment was previously modulated by hypofractionated RT with not too many fractions, but failed to improve RT plus anti-PD-induced abscopal responses that are characterized by distinct immune alterations.
Collapse
Affiliation(s)
- Michael Rückert
- Department of Radiation Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.R.); (L.D.); (B.F.); (R.F.)
| | - Lisa Deloch
- Department of Radiation Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.R.); (L.D.); (B.F.); (R.F.)
| | - Benjamin Frey
- Department of Radiation Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.R.); (L.D.); (B.F.); (R.F.)
| | - Eberhard Schlücker
- Institute of Process Machinery and Systems Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany;
| | - Rainer Fietkau
- Department of Radiation Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.R.); (L.D.); (B.F.); (R.F.)
| | - Udo S. Gaipl
- Department of Radiation Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (M.R.); (L.D.); (B.F.); (R.F.)
- Correspondence: ; Tel.: +49-(0)9131-85-44258
| |
Collapse
|
33
|
L-Selectin expression is associated with inflammatory microenvironment and favourable prognosis in breast cancer. 3 Biotech 2021; 11:38. [PMID: 33479593 DOI: 10.1007/s13205-020-02549-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 11/06/2020] [Indexed: 12/24/2022] Open
Abstract
L-selectin is a cell adhesion molecule that plays an important role in modulating immune cell trafficking. The expression of L-selectin has been found to be upregulated in several human cancers. However, the association of L-selectin expression with the immune profile and its prognostic value in breast cancer has not been explored in detail. We utilized TCGA and Oncomine datasets to compare SELL (L-selectin gene) expression between tumor and normal breast tissues. The association of SELL expression with its promoter DNA methylation and infiltrating immune cells was evaluated by using Wanderer, TIMER, and CIBERSORT tools. Single cell RNA sequencing data was utilised to determine the cell specific expression of L-selectin in breast cancer. Furthermore, the relationship between SELL expression and patient survival was evaluated using the Kaplan-Meier plotter. Gene set enrichment analysis was performed to determine functional associations of SELL expression. We found that SELL expression was significantly higher in breast tumors and regulated by DNA methylation. L-selectin exhibited a strong positive correlation with markers of the inflammatory microenvironment, including M1 macrophages. Interestingly, single cell sequencing data analysis revealed that B-cells and T-cells exhibited comparable expression levels of SELL, suggesting both B-cells and T cells contribute to SELL expression in breast cancer. Higher expression of SELL was associated with better survival outcome in basal, Her2 + and luminal B subtypes of breast cancer. GSEA revealed association of SELL expression with several immunological features in breast cancer. SELL expression increases in breast tumor tissues with reduced DNA methylation and associated inflammatory microenvironment. Also, high SELL expression is associated with favorable survival outcomes in breast cancer.
Collapse
|
34
|
Ebelt ND, Zuniga E, Marzagalli M, Zamloot V, Blazar BR, Salgia R, Manuel ER. Salmonella-Based Therapy Targeting Indoleamine 2,3-Dioxygenase Restructures the Immune Contexture to Improve Checkpoint Blockade Efficacy. Biomedicines 2020; 8:E617. [PMID: 33339195 PMCID: PMC7765568 DOI: 10.3390/biomedicines8120617] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/09/2020] [Accepted: 12/13/2020] [Indexed: 12/15/2022] Open
Abstract
Therapeutic options for non-small cell lung cancer (NSCLC) treatment have changed dramatically in recent years with the advent of novel immunotherapeutic approaches. Among these, immune checkpoint blockade (ICB) using monoclonal antibodies has shown tremendous promise in approximately 20% of patients. In order to better predict patients that will respond to ICB treatment, biomarkers such as tumor-associated CD8+ T cell frequency, tumor checkpoint protein status and mutational burden have been utilized, however, with mixed success. In this study, we hypothesized that significantly altering the suppressive tumor immune landscape in NSCLC could potentially improve ICB efficacy. Using sub-therapeutic doses of our Salmonella typhimurium-based therapy targeting the suppressive molecule indoleamine 2,3-dioxygenase (shIDO-ST) in tumor-bearing mice, we observed dramatic changes in immune subset phenotypes that included increases in antigen presentation markers, decreased regulatory T cell frequency and overall reduced checkpoint protein expression. Combination shIDO-ST treatment with anti-PD-1/CTLA-4 antibodies enhanced tumor growth control, compared to either treatment alone, which was associated with significant intratumoral infiltration by CD8+ and CD4+ T cells. Ultimately, we show that increases in antigen presentation markers and infiltration by T cells is correlated with significantly increased survival in NSCLC patients. These results suggest that the success of ICB therapy may be more accurately predicted by taking into account multiple factors such as potential for antigen presentation and immune subset repertoire in addition to markers already being considered. Alternatively, combination treatment with agents such as shIDO-ST could be used to create a more conducive tumor microenvironment for improving responses to ICB.
Collapse
Affiliation(s)
- Nancy D. Ebelt
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (N.D.E.); (E.Z.); (M.M.); (V.Z.)
| | - Edith Zuniga
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (N.D.E.); (E.Z.); (M.M.); (V.Z.)
| | - Monica Marzagalli
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (N.D.E.); (E.Z.); (M.M.); (V.Z.)
| | - Vic Zamloot
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (N.D.E.); (E.Z.); (M.M.); (V.Z.)
| | - Bruce R. Blazar
- Department of Pediatrics, Division of Blood and Bone Marrow Transplantation, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA;
| | - Edwin R. Manuel
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (N.D.E.); (E.Z.); (M.M.); (V.Z.)
| |
Collapse
|
35
|
Todorova D, Zhang Y, Chen Q, Liu J, He J, Fu X, Xu Y. hESC-derived immune suppressive dendritic cells induce immune tolerance of parental hESC-derived allografts. EBioMedicine 2020; 62:103120. [PMID: 33242828 PMCID: PMC7695963 DOI: 10.1016/j.ebiom.2020.103120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 09/28/2020] [Accepted: 10/26/2020] [Indexed: 12/21/2022] Open
Abstract
Background With their inherent capability of unlimited self-renewal and unique potential to differentiate into functional cells of the three germ layers, human embryonic stem cells (hESCs) hold great potential in regenerative medicine. A major challenge in the application of hESC-based cell therapy is the allogeneic immune rejection of hESC-derived allografts. Methods We derived dendritic cell-like cells (DCLs) from wild type and CTLA4-Ig/PD-L1 knock-in hESCs, denoted WT DCLs and CP DCLs. The expression of DC-related genes and surface molecules was evaluated, as well as their DCL capacity to stimulate allogeneic T cells and induce regulatory T (Treg) cells in vitro. Using an immune system humanized mouse model, we investigated whether the adoptive transfer of CP DCLs can induce long-term immune tolerance of parental hESC-derived smooth muscle and cardiomyocyte allografts. Findings CP DCLs can maintain immune suppressive properties after robust inflammatory stimulation and induce Treg cells. While CP DCLs survive transiently in vivo, they induce long-term immune tolerance of parental hESC-derived allografts. Interpretation This strategy does not cause systemic immune suppression but induces immune tolerance specific for DCL-specific HLAs, and thus it presents a safe and effective approach to induce immune tolerance of allografts derived from any clinically approved hESC line. Funding NSFC, leading talents of Guangdong Province Program (No. 00201516), Key R&D Program of Guangdong Province (2019B020235003), Science and Technology Innovation Committee of Shenzhen Municipality (JCYJ20180504170301309), National High-tech R&D Program (863 Program No. 2015AA020310), Shenzhen “Sanming” Project of Medicine (SZSM201602102), Development and Reform Commission of Shenzhen Municipality (S2016004730009), CIRM (DISC2–10559).
Collapse
Affiliation(s)
- Dilyana Todorova
- Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Yue Zhang
- Guangzhou University of Chinese Medicine, Second Clinical Medical College, 232 Waihuan Road E, Guangzhou, Guangdong 510006, China
| | - Qu Chen
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Jingfeng Liu
- Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Jingjin He
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Xuemei Fu
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China; Shenzhen Children's Hospital, Shenzhen 518026, China..
| | - Yang Xu
- Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Guangzhou University of Chinese Medicine, Second Clinical Medical College, 232 Waihuan Road E, Guangzhou, Guangdong 510006, China; The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China.
| |
Collapse
|
36
|
Ramadhani AH, Nafisah W, Isnanto H, Sholeha TK, Jatmiko YD, Tsuboi H, Rifa'i M. Immunomodulatory Effects of Cyperus rotundus Extract on 7,12 Dimethylbenz[a]anthracene (DMBA) Exposed BALB/c Mice. PHARMACEUTICAL SCIENCES 2020. [DOI: 10.34172/ps.2020.61] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Background: The carcinogenic substance 7,12-Dimethylbenz[a]anthracene (DMBA) was commonly used to induce tumor formation in rodents. The development of tumor may trigger higher expression of pro-inflammatory cytokines, which in turn supports tumor progression. In this study, we examined the efficacy of Cyperus rotundus extract (CRE) that was reported to have anti-inflammatory properties. We focused on investigating the levels of activated T lymphocytes and the pro-inflammatory cytokines expressed by macrophages. Methods: Female BALB/c were injected with DMBA subcutaneously. The DMBA exposed mice were given CRE orally in three different doses; 63.33, 158.4, and 316.8 mg/kg. After 14 days, the levels of activated T lymphocytes and pro-inflammatory cytokines were analyzed using flow cytometry. Graphical analysis was done with FlowJo v10 and followed by statistical analysis. Results: The treatment of CRE reduced the population of CD4 and CD8 T cells. The number of activated CD4 and CD8 T cells were also significantly suppressed. The population of macrophages marked by CD11b cells was significantly reduced. Finally, the CRE treatment suppressed the levels of TNF-α, IFN-γ, IL-1β, and IL-6 expressed by macrophages. Conclusion: Our findings suggest that CRE could be a potential agent useful in therapeutic approaches for curing the disease caused by aberrant cells.
Collapse
Affiliation(s)
- Abu Hanifah Ramadhani
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Jl. Veteran Malang 65145, East Java, Indonesia
| | - Wirdatun Nafisah
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Jl. Veteran Malang 65145, East Java, Indonesia
| | - Hary Isnanto
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Jl. Veteran Malang 65145, East Java, Indonesia
| | - Tri Kurniawati Sholeha
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Jl. Veteran Malang 65145, East Java, Indonesia
| | - Yoga Dwi Jatmiko
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Jl. Veteran Malang 65145, East Java, Indonesia
| | - Hideo Tsuboi
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Muhaimin Rifa'i
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Jl. Veteran Malang 65145, East Java, Indonesia
- Pusat Studi Biosistem, Brawijaya University, Jl. Veteran Malang 65145, East Java, Indonesia
| |
Collapse
|
37
|
Prakash S, Roy S, Srivastava R, Coulon PG, Dhanushkodi NR, Vahed H, Jankeel A, Geertsema R, Amezquita C, Nguyen L, Messaoudi I, Burkhardt AM, BenMohamed L. Unique molecular signatures of antiviral memory CD8 + T cells associated with asymptomatic recurrent ocular herpes. Sci Rep 2020; 10:13843. [PMID: 32796943 PMCID: PMC7427992 DOI: 10.1038/s41598-020-70673-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 07/28/2020] [Indexed: 12/14/2022] Open
Abstract
The nature of antiviral CD8+ T cells associated with protective and pathogenic herpes simplex virus type 1 (HSV-1) infections remains unclear. We compared the transcriptome, phenotype, and function of memory CD8+ T cells, sharing the same HSV-1 epitope-specificities, from infected HLA-A*0201 positive symptomatic (SYMP) vs. asymptomatic (ASYMP) individuals and HLA-A*0201 transgenic rabbits. Compared to higher frequencies of multifunctional effector memory CD8+ TEM cells in ASYMP individuals, the SYMP individuals presented dysfunctional CD8+ TEM cells, expressing major exhaustion pathways. Compared to protected ASYMP HLA transgenic rabbits, the trigeminal ganglia of non-protected SYMP HLA transgenic rabbits had higher frequencies of dysfunctional tissue-resident CD8+ TRM cells. Moreover, blockade of T cell exhaustion pathways restored the function of CD8+ T cells, reduced virus reactivation, and diminished recurrent disease in HLA transgenic rabbits. These findings reveal unique molecular signatures of protective CD8+ T cells and pave the way for T-cell-based immunotherapy to combat recurrent ocular herpes.
Collapse
Affiliation(s)
- Swayam Prakash
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Hewitt Hall, Room 2032; 843 Health Sciences Rd, Irvine, CA, 92697, USA
| | - Soumyabrata Roy
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Hewitt Hall, Room 2032; 843 Health Sciences Rd, Irvine, CA, 92697, USA
| | - Ruchi Srivastava
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Hewitt Hall, Room 2032; 843 Health Sciences Rd, Irvine, CA, 92697, USA
| | - Pierre-Gregoire Coulon
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Hewitt Hall, Room 2032; 843 Health Sciences Rd, Irvine, CA, 92697, USA
| | - Nisha R Dhanushkodi
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Hewitt Hall, Room 2032; 843 Health Sciences Rd, Irvine, CA, 92697, USA
| | - Hawa Vahed
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Hewitt Hall, Room 2032; 843 Health Sciences Rd, Irvine, CA, 92697, USA
| | - Allen Jankeel
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, Irvine, CA, 92697, USA
| | - Roger Geertsema
- University Laboratory Animal Resources, University of California Irvine, Irvine, CA, 92697, USA
| | - Cassandra Amezquita
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Hewitt Hall, Room 2032; 843 Health Sciences Rd, Irvine, CA, 92697, USA
| | - Lan Nguyen
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Hewitt Hall, Room 2032; 843 Health Sciences Rd, Irvine, CA, 92697, USA
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, Irvine, CA, 92697, USA
| | - Amanda M Burkhardt
- Vaccine Research and Development Center, Department of Physiology & Biophysics, University of California, Irvine, CA, 92617, USA
- Institute for Immunology, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - Lbachir BenMohamed
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Hewitt Hall, Room 2032; 843 Health Sciences Rd, Irvine, CA, 92697, USA.
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, Irvine, CA, 92697, USA.
- Vaccine Research and Development Center, Department of Physiology & Biophysics, University of California, Irvine, CA, 92617, USA.
- Institute for Immunology, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
38
|
Piconese S, Campello S, Natalini A. Recirculation and Residency of T Cells and Tregs: Lessons Learnt in Anacapri. Front Immunol 2020; 11:682. [PMID: 32431695 PMCID: PMC7214633 DOI: 10.3389/fimmu.2020.00682] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/26/2020] [Indexed: 02/01/2023] Open
Abstract
"Location, location, and location": according to this mantra, the place where living beings settle has a key impact on the success of their activities; in turn, the living beings can, in many ways, modify their environment. This idea has now become more and more true for T cells. The ability of T cells to recirculate throughout blood or lymph, or to stably reside in certain tissues, turned out to determine immunity to pathogens, and tumors. If location matters also for human beings, the inspiring environment of Capri Island has contributed to the success of the EFIS-EJI Ruggero Ceppellini Advanced School of Immunology focused on "T cell memory," held in Anacapri from October 12, 2018 to October 15, 2018. In this minireview, we would like to highlight some novel concepts about T cell migration and residency and discuss their implications in relation to recent advances in the field, including the mechanisms regulating compartmentalization and cell cycle entry of T cells during activation, the role of mitochondrial metabolism in T cell movement, and the residency of regulatory T cells.
Collapse
Affiliation(s)
- Silvia Piconese
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Silvia Campello
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Ambra Natalini
- Institute of Molecular Biology and Pathology, National Research Council (CNR), Rome, Italy.,Dipartimento di Medicina Molecolare (DMM), Sapienza Università di Roma, Rome, Italy
| |
Collapse
|
39
|
Drescher HK, Schippers A, Rosenhain S, Gremse F, Bongiovanni L, de Bruin A, Eswaran S, Gallage SU, Pfister D, Szydlowska M, Heikenwalder M, Weiskirchen S, Wagner N, Trautwein C, Weiskirchen R, Kroy DC. L-Selectin/CD62L is a Key Driver of Non-Alcoholic Steatohepatitis in Mice and Men. Cells 2020; 9:cells9051106. [PMID: 32365632 PMCID: PMC7290433 DOI: 10.3390/cells9051106] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
CD62L (L-Selectin) dependent lymphocyte infiltration is known to induce inflammatory bowel disease (IBD), while its function in the liver, especially in non-alcoholic steatohepatitis (NASH), remains unclear. We here investigated the functional role of CD62L in NASH in humans as well as in two mouse models of steatohepatitis. Hepatic expression of a soluble form of CD62L (sCD62L) was measured in patients with steatosis and NASH. Furthermore, CD62L−/− mice were fed with a methionine and choline deficient (MCD) diet for 4 weeks or with a high fat diet (HFD) for 24 weeks. Patients with NASH displayed increased serum levels of sCD62L. Hepatic CD62L expression was higher in patients with steatosis and increased dramatically in NASH patients. Interestingly, compared to wild type (WT) mice, MCD and HFD-treated CD62L−/− mice were protected from diet-induced steatohepatitis. This was reflected by less fat accumulation in hepatocytes and a dampened manifestation of the metabolic syndrome with an improved insulin resistance and decreased cholesterol and triglyceride levels. Consistent with ameliorated disease, CD62L−/− animals exhibited an enhanced hepatic infiltration of Treg cells and a strong activation of an anti-oxidative stress response. Those changes finally resulted in less fibrosis in CD62L−/− mice. Additionally, this effect could be reproduced in a therapeutic setting by administrating an anti-CD62L blocking antibody. CD62L expression in humans and mice correlates with disease activity of steatohepatitis. CD62L knockout and anti-CD62L-treated mice are protected from diet-induced steatohepatitis suggesting that CD62L is a promising target for therapeutic interventions in NASH.
Collapse
Affiliation(s)
- Hannah K. Drescher
- Department of Internal Medicine III, University Hospital, RWTH Aachen, 52074 Aachen, Germany; (C.T.); (D.C.K.)
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Correspondence:
| | - Angela Schippers
- Department of Pediatrics, University Hospital, RWTH Aachen, 52074 Aachen, Germany; (A.S.); (S.E.); (N.W.)
| | - Stefanie Rosenhain
- Institute for Experimental Molecular Imaging, University Hospital, RWTH Aachen University, 52074 Aachen, Germany; (S.R.); (F.G.)
| | - Felix Gremse
- Institute for Experimental Molecular Imaging, University Hospital, RWTH Aachen University, 52074 Aachen, Germany; (S.R.); (F.G.)
| | - Laura Bongiovanni
- Dutch Molecular Pathology Centre, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, 3508 Utrecht, The Netherlands; (L.B.); (A.d.B.)
| | - Alain de Bruin
- Dutch Molecular Pathology Centre, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, 3508 Utrecht, The Netherlands; (L.B.); (A.d.B.)
| | - Sreepradha Eswaran
- Department of Pediatrics, University Hospital, RWTH Aachen, 52074 Aachen, Germany; (A.S.); (S.E.); (N.W.)
| | - Suchira U. Gallage
- Division of Chronic Inflammation and Cancer, German Cancer Research Center Heidelberg (DKFZ), 69120 Heidelberg, Germany; (S.U.G.); (D.P.); (M.S.); (M.H.)
| | - Dominik Pfister
- Division of Chronic Inflammation and Cancer, German Cancer Research Center Heidelberg (DKFZ), 69120 Heidelberg, Germany; (S.U.G.); (D.P.); (M.S.); (M.H.)
| | - Marta Szydlowska
- Division of Chronic Inflammation and Cancer, German Cancer Research Center Heidelberg (DKFZ), 69120 Heidelberg, Germany; (S.U.G.); (D.P.); (M.S.); (M.H.)
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center Heidelberg (DKFZ), 69120 Heidelberg, Germany; (S.U.G.); (D.P.); (M.S.); (M.H.)
| | - Sabine Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), University Hospital, RWTH Aachen University, 52074 Aachen, Germany; (S.W.); (R.W.)
| | - Norbert Wagner
- Department of Pediatrics, University Hospital, RWTH Aachen, 52074 Aachen, Germany; (A.S.); (S.E.); (N.W.)
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital, RWTH Aachen, 52074 Aachen, Germany; (C.T.); (D.C.K.)
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), University Hospital, RWTH Aachen University, 52074 Aachen, Germany; (S.W.); (R.W.)
| | - Daniela C. Kroy
- Department of Internal Medicine III, University Hospital, RWTH Aachen, 52074 Aachen, Germany; (C.T.); (D.C.K.)
| |
Collapse
|
40
|
Phenotypic and functional characterization of natural killer cells in rheumatoid arthritis-regulation with interleukin-15. Sci Rep 2020; 10:5858. [PMID: 32246007 PMCID: PMC7125139 DOI: 10.1038/s41598-020-62654-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 03/13/2020] [Indexed: 11/23/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovial inflammation and joint destruction. Previous studies have shown that natural killer (NK) cells may play an important role in the pathogenesis of RA. Interleukin (IL)-15, a pro-inflammatory cytokine which induces proliferation and differentiation of NK cells, is overexpressed in RA. In this present study, we examine various NKRs and adhesion molecule expression on NK cells from RA patients and their response to IL-15 stimulation. We also sought to study cytokine-induced memory-like (CIML) NK cells in RA patients. We established that 1. RA patients had higher NK cell percentages in peripheral blood and their serum IL-15 levels were higher compared to healthy volunteers; 2. NK cells from RA patients showed lower NKp46 expression and an impaired CD69 response to IL-15; 3. NK cells from RA patients showed higher CD158b and CD158e expression but lower CD62L expression; 4. exogenous IL-15 up-regulated CD69, CD158b, CD158e but down-regulated NKp46 and CD62L expression in RA; 5. As to CIML NK cells, restimulation - induced NK cytotoxicity and IFN-γ production was impaired in RA patients, 6. Reduced NKp46, perforin, and granzyme B expression on NK cells was found in RA patients with bone deformity and erosion, 7. RA disease activity (DAS28) showed inverse correlation with the percentages of CD56+CD3− NK cells, and NKp46 and perforin expression on NK cells, respectively. Taken together, our study demonstrated differential expression of various NK receptors in RA patients. NKp46, CD158e, and perforin expression on NK cells may serve as markers of RA severity.
Collapse
|
41
|
Bonte S, De Munter S, Goetgeluk G, Ingels J, Pille M, Billiet L, Taghon T, Leclercq G, Vandekerckhove B, Kerre T. T-cells with a single tumor antigen-specific T-cell receptor can be generated in vitro from clinically relevant stem cell sources. Oncoimmunology 2020; 9:1727078. [PMID: 32117593 PMCID: PMC7028335 DOI: 10.1080/2162402x.2020.1727078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/05/2019] [Accepted: 12/19/2019] [Indexed: 11/08/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cells have shown great promise in the treatment of B-cell malignancies. For acute myeloid leukemia (AML), however, the optimal target surface antigen has yet to be discovered. Alternatively, T-cell receptor (TCR)-redirected T-cells target intracellular antigens, marking a broader territory of available target antigens. Currently, adoptive TCR T-cell therapy uses peripheral blood lymphocytes for the introduction of a transgenic TCR. However, this can cause graft-versus-host disease, due to mispairing of introduced and endogenous TCR chains. Therefore, we started from hematopoietic stem and progenitor cells (HSPC), that do not express a TCR yet, isolated from healthy donors, patients in remission after chemotherapy and AML patients at diagnosis. Using the OP9-DL1 in vitro co-culture system and agonist selection, TCR-transduced HSPC develop into mature tumor antigen-specific T-cells with only one TCR. We show here that this approach is feasible with adult HSPC from clinically relevant sources, albeit with slower maturation and lower cell yield compared to cord blood HSPC. Moreover, cryopreservation of HSPC does not have an effect on cell numbers or functionality of the generated T-cells. In conclusion, we show here that it is feasible to generate TA-specific T-cells from HSPC from adult healthy donors and patients and we believe these T-cells could be of use as a very valuable form of patient-tailored T-cell immunotherapy.
Collapse
Affiliation(s)
- Sarah Bonte
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Stijn De Munter
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Glenn Goetgeluk
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Joline Ingels
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Melissa Pille
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Lore Billiet
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Tom Taghon
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Georges Leclercq
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Bart Vandekerckhove
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Tessa Kerre
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium.,Department of Hematology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
42
|
Bing T, Zhang N, Shangguan D. Cell-SELEX, an Effective Way to the Discovery of Biomarkers and Unexpected Molecular Events. ACTA ACUST UNITED AC 2019; 3:e1900193. [PMID: 32648677 DOI: 10.1002/adbi.201900193] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/26/2019] [Indexed: 12/15/2022]
Abstract
Cell-SELEX can not only generate aptamers for specific cell isolation/detection, diagnosis, and therapy, but also lead to the discovery of biomarkers and unexpected molecular events. However, most cell-SELEX research is concentrated on aptamer generation and applications. In this progress report, recent research progress with cell-SELEX in terms of the discovery of biomarkers and unexpected molecular events is highlighted. In particular, the key technical challenges for cell-SELEX-based biomarker discovery, namely, the methods for identification and validation of target proteins of aptamers, are discussed in detail. Finally, the prospects of the applications of cell-SELEX in this field now and in the near future are described. It is expected that this report will attract attention to the benefit of cell-SELEX and provide a practical reference for biomedical researchers.
Collapse
Affiliation(s)
- Tao Bing
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Nan Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dihua Shangguan
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
43
|
Islam SMS, Kim HA, Choi B, Jung JY, Lee SM, Suh CH, Sohn S. Differences in Expression of Human Leukocyte Antigen Class II Subtypes and T Cell Subsets in Behçet's Disease with Arthritis. Int J Mol Sci 2019; 20:ijms20205044. [PMID: 31614573 PMCID: PMC6829274 DOI: 10.3390/ijms20205044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/30/2019] [Accepted: 10/08/2019] [Indexed: 12/15/2022] Open
Abstract
It has been reported Human Leukocyte Antigen (HLA) gene polymorphism is a risk factor for the development of Behçet’s disease (BD). In this study, the association of HLA class II subtypes HLA-DP, DQ, DR, and T cell subsets in BD patients with arthritis was evaluated. Frequencies of HLA-DP, DQ, DR positive cells, and T cell subsets in peripheral blood leukocytes (PBL) were measured by flow cytometric analysis in BD, and compared to rheumatoid arthritis as disease controls and healthy controls. Frequencies of HLA-DQ were significantly decreased in whole PBL and granulocytes of BD active patients as compared to healthy controls. In monocytes populations, proportions of HLA-DR positive cells were significantly increased in BD active patients as compared to healthy controls. Proportions of CD4+CCR7+ and CD8+CCR7+ cells were significantly higher in BD active patients than in BD inactive in whole PBL. Frequencies of CD4+CD62L- and CD8+CD62L- cells in lymphocytes were significantly decreased in active BD than those in inactive BD. There were also correlations between disease activity markers and T cell subsets. Our results revealed HLA-DP, DQ, and DR expressing cell frequencies and several T cell subsets were significantly correlated with BD arthritis symptoms.
Collapse
Affiliation(s)
- S M Shamsul Islam
- Department of Biomedical Science, Ajou University School of Medicine, Suwon 16499, Korea.
| | - Hyoun-Ah Kim
- Department of Rheumatology, Ajou University School of Medicine, Suwon 16499, Korea.
| | - Bunsoon Choi
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Korea.
| | - Ju-Yang Jung
- Department of Rheumatology, Ajou University School of Medicine, Suwon 16499, Korea.
| | - Sung-Min Lee
- Department of Rheumatology, Ajou University School of Medicine, Suwon 16499, Korea.
| | - Chang-Hee Suh
- Department of Rheumatology, Ajou University School of Medicine, Suwon 16499, Korea.
| | - Seonghyang Sohn
- Department of Biomedical Science, Ajou University School of Medicine, Suwon 16499, Korea.
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Korea.
| |
Collapse
|
44
|
Perfilyeva YV, Ostapchuk YO, Abdolla N, Tleulieva R, Krasnoshtanov VC, Belyaev NN. Exogenous Melatonin Up-Regulates Expression of CD62L by Lymphocytes in Aged Mice under Inflammatory and Non-Inflammatory Conditions. Immunol Invest 2019; 48:632-643. [PMID: 30887869 DOI: 10.1080/08820139.2019.1586918] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
It is well documented that age-related impaired functioning of immunocompetent cells is associated with an increase in the rates of chronic inflammatory diseases. Recently, an ability of melatonin to modulate inflammatory processes by regulating leucocyte recruitment has been demonstrated. However, to date, no studies have attempted to determine the impact of melatonin on the expression of CD62L by lymphocytes. CD62L, also known as L-selectin, is required for the entry of lymphocytes into secondary lymphoid organs, sites of tumor growth and chronic inflammation through high endothelial venules. Here, we investigated the effect of melatonin at physiological concentrations on the expression of CD62L by T and NK cells in vivo and in vitro. We demonstrated that NK and CD3+ T cells obtained from the spleen of aged mice were characterized by decreased expression of CD62L compared to young mice. Melatonin administration up-regulated the levels of surface CD62L on NK and T cell populations in aged mice under non-inflammatory conditions and on CD8+ T cells in aged mice with chronic inflammation. Pre-incubation with melatonin prevented the reduction in CD62L expression by CD8+ T cells induced by the co-cultivation of peripheral blood mononuclear cells with human pancreatic adenocarcinoma cell line (MiaPaCa-2). The obtained results suggest that melatonin can modulate lymphocyte homing into lymph nodes and sites of chronic inflammation and, therefore, can stimulate immune responses in chronic inflammatory conditions associated with aging.
Collapse
Affiliation(s)
- Yuliya V Perfilyeva
- a Laboratory of Molecular Immunology and Immunobiotechnology , M.A.Aitkhozhin's Institute of Molecular Biology and Biochemistry , Almaty , Kazakhstan
| | - Yekaterina O Ostapchuk
- a Laboratory of Molecular Immunology and Immunobiotechnology , M.A.Aitkhozhin's Institute of Molecular Biology and Biochemistry , Almaty , Kazakhstan
| | - Nurshat Abdolla
- a Laboratory of Molecular Immunology and Immunobiotechnology , M.A.Aitkhozhin's Institute of Molecular Biology and Biochemistry , Almaty , Kazakhstan
- b Department of Biophysics and Biomedicine , Al-Farabi Kazakh National University , Almaty , Kazakhstan
| | - Raikhan Tleulieva
- a Laboratory of Molecular Immunology and Immunobiotechnology , M.A.Aitkhozhin's Institute of Molecular Biology and Biochemistry , Almaty , Kazakhstan
| | | | - Nikolai N Belyaev
- d Department of New Technologies , Saint-Petersburg Pasteur Institute , Saint-Petersburg , Russia
| |
Collapse
|
45
|
Ivetic A, Hoskins Green HL, Hart SJ. L-selectin: A Major Regulator of Leukocyte Adhesion, Migration and Signaling. Front Immunol 2019; 10:1068. [PMID: 31139190 PMCID: PMC6527602 DOI: 10.3389/fimmu.2019.01068] [Citation(s) in RCA: 289] [Impact Index Per Article: 48.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/26/2019] [Indexed: 12/12/2022] Open
Abstract
L-selectin (CD62L) is a type-I transmembrane glycoprotein and cell adhesion molecule that is expressed on most circulating leukocytes. Since its identification in 1983, L-selectin has been extensively characterized as a tethering/rolling receptor. There is now mounting evidence in the literature to suggest that L-selectin plays a role in regulating monocyte protrusion during transendothelial migration (TEM). The N-terminal calcium-dependent (C-type) lectin domain of L-selectin interacts with numerous glycans, including sialyl Lewis X (sLex) for tethering/rolling and proteoglycans for TEM. Although the signals downstream of L-selectin-dependent adhesion are poorly understood, they will invariably involve the short 17 amino acid cytoplasmic tail. In this review we will detail the expression of L-selectin in different immune cell subsets, and its influence on cell behavior. We will list some of the diverse glycans known to support L-selectin-dependent adhesion, within luminal and abluminal regions of the vessel wall. We will describe how each domain within L-selectin contributes to adhesion, migration and signal transduction. A significant focus on the L-selectin cytoplasmic tail and its proposed contribution to signaling via the ezrin-radixin-moesin (ERM) family of proteins will be outlined. Finally, we will discuss how ectodomain shedding of L-selectin during monocyte TEM is essential for the establishment of front-back cell polarity, bestowing emigrated cells the capacity to chemotax toward sites of damage.
Collapse
Affiliation(s)
- Aleksandar Ivetic
- King's College London, School of Cardiovascular Medicine and Sciences, BHF Center of Research Excellence, London, United Kingdom
| | - Hannah Louise Hoskins Green
- King's College London, School of Cardiovascular Medicine and Sciences, BHF Center of Research Excellence, London, United Kingdom
| | - Samuel James Hart
- King's College London, School of Cardiovascular Medicine and Sciences, BHF Center of Research Excellence, London, United Kingdom
| |
Collapse
|
46
|
Borsig L. Selectins in cancer immunity. Glycobiology 2018; 28:648-655. [PMID: 29272415 DOI: 10.1093/glycob/cwx105] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/18/2017] [Indexed: 12/12/2022] Open
Abstract
Selectins are vascular adhesion molecules that mediate physiological responses such as inflammation, immunity and hemostasis. During cancer progression, selectins promote various steps enabling the interactions between tumor cells and the blood constituents, including platelets, endothelial cells and leukocytes. Selectins are carbohydrate-binding molecules that bind to sialylated, fucosylated glycan structures. The increased selectin ligand expression on tumor cells correlates with enhanced metastasis and poor prognosis for cancer patients. While, many studies focused on the role of selectin as a mediator of tumor cell adhesion and extravasation during metastasis, there is evidence for selectins to activate signaling cascade that regulates immune responses within a tumor microenvironment. L-Selectin binding induces activation of leukocytes, which can be further modulated by selectin-mediated interactions with platelets and endothelial cells. Selectin ligand on leukocytes, PSGL-1, triggers intracellular signaling in leukocytes that are induced through platelet's P-selectin or endothelial E-selectin binding. In this review, I summarize the evidence for selectin-induced immune modulation in cancer progression that represents a possible target for controlling tumor immunity.
Collapse
Affiliation(s)
- Lubor Borsig
- Institute of Physiology, University of Zurich and Zurich Center for Integrative Human Physiology, Winterthurerstrasse 190, Zurich, Switzerland
| |
Collapse
|
47
|
Kornej J, Büttner P, Hammer E, Engelmann B, Dinov B, Sommer P, Husser D, Hindricks G, Völker U, Bollmann A. Circulating proteomic patterns in AF related left atrial remodeling indicate involvement of coagulation and complement cascade. PLoS One 2018; 13:e0198461. [PMID: 30496173 PMCID: PMC6264811 DOI: 10.1371/journal.pone.0198461] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 11/12/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Left atrial (LA) electro-anatomical remodeling and diameter increase in atrial fibrillation (AF) indicates disease progression and is associated with poor therapeutic success. Furthermore, AF leads to a hypercoagulable state, which in turn promotes the development of a substrate for AF and disease progression in the experimental setting. The aim of this study was to identify pathways associated with LA remodeling in AF patients using untargeted proteomics approach. METHODS Peripheral blood samples of 48 patients (62±10 years, 63% males, 59% persistent AF) undergoing AF catheter ablation were collected before ablation. 23 patients with left atrial low voltage areas (LVA), defined as <0.5 mV, and 25 patients without LVA were matched for age, gender and CHA2DS2-VASc score. Untargeted proteome analysis was performed using LC-ESI-Tandem mass spectrometry in a label free intensity based workflow. Significantly different abundant proteins were identified and used for pathway analysis and protein-protein interaction analysis. RESULTS Analysis covered 280 non-redundant circulating plasma proteins. The presence of LVA correlated with 30 differentially abundant proteins of coagulation and complement cascade (q<0.05). CONCLUSIONS This pilot proteomic study identified plasma protein candidates associated with electro-anatomical remodeling in AF and pointed towards an imbalance in coagulation and complement pathway, tissue remodeling and inflammation.
Collapse
Affiliation(s)
- Jelena Kornej
- Department of Electrophysiology, Heart Center Leipzig, University of Leipzig, Leipzig, Germany
- * E-mail:
| | - Petra Büttner
- Department of Electrophysiology, Heart Center Leipzig, University of Leipzig, Leipzig, Germany
| | - Elke Hammer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Beatrice Engelmann
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Borislav Dinov
- Department of Electrophysiology, Heart Center Leipzig, University of Leipzig, Leipzig, Germany
| | - Philipp Sommer
- Department of Electrophysiology, Heart Center Leipzig, University of Leipzig, Leipzig, Germany
| | - Daniela Husser
- Department of Electrophysiology, Heart Center Leipzig, University of Leipzig, Leipzig, Germany
| | - Gerhard Hindricks
- Department of Electrophysiology, Heart Center Leipzig, University of Leipzig, Leipzig, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Andreas Bollmann
- Department of Electrophysiology, Heart Center Leipzig, University of Leipzig, Leipzig, Germany
| |
Collapse
|
48
|
Campbell JD, Fraser AR. Flow cytometric assays for identity, safety and potency of cellular therapies. CYTOMETRY PART B-CLINICAL CYTOMETRY 2018; 94:569-579. [DOI: 10.1002/cyto.b.21735] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 06/18/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022]
|
49
|
Safya H, Mellouk A, Legrand J, Le Gall SM, Benbijja M, Kanellopoulos-Langevin C, Kanellopoulos JM, Bobé P. Variations in Cellular Responses of Mouse T Cells to Adenosine-5'-Triphosphate Stimulation Do Not Depend on P2X7 Receptor Expression Levels but on Their Activation and Differentiation Stage. Front Immunol 2018. [PMID: 29535730 PMCID: PMC5835135 DOI: 10.3389/fimmu.2018.00360] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A previous report has shown that regulatory T cells (Treg) were markedly more sensitive to adenosine-5′-triphosphate (ATP) than conventional T cells (Tconv). Another one has shown that Tregs and CD45RBlow Tconvs, but not CD45RBhigh Tconvs, displayed similar high sensitivity to ATP. We have previously reported that CD45RBlow Tconvs expressing B220/CD45RABC molecules in a pre-apoptotic stage are resistant to ATP stimulation due to the loss of P2X7 receptor (P2X7R) membrane expression. To gain a clearer picture on T-cell sensitivity to ATP, we have quantified four different cellular activities triggered by ATP in mouse T cells at different stages of activation/differentiation, in correlation with levels of P2X7R membrane expression. P2X7R expression significantly increases on Tconvs during differentiation from naive CD45RBhighCD44low to effector/memory CD45RBlowCD44high stage. Maximum levels of upregulation are reached on recently activated CD69+ naive and memory Tconvs. Ectonucleotidases CD39 and CD73 expression levels increase in parallel with those of P2X7R. Recently activated CD69+ CD45RBhighCD44low Tconvs, although expressing high levels of P2X7R, fail to cleave homing receptor CD62L after ATP treatment, but efficiently form pores and externalize phosphatidylserine (PS). In contrast, naive CD45RBhighCD44low Tconvs cleave CD62L with high efficiency although they express a lower level of P2X7, thus suggesting that P2X7R levels are not a limiting factor for signaling ATP-induced cellular responses. Contrary to common assumption, P2X7R-mediated cellular activities in mouse Tconvs are not triggered in an all-or-none manner, but depend on their stage of activation/differentiation. Compared to CD45RBlow Tconvs, CD45RBlowFoxp3+ Tregs show significantly higher levels of P2X7R membrane expression and of sensitivity to ATP as evidenced by their high levels of CD62L shedding, pore formation and PS externalization observed after ATP treatment. In summary, the different abilities of ATP-treated Tconvs to form pore or cleave CD62L depending on their activation and differentiation state suggests that P2X7R signaling varies according to the physiological role of T convs during antigen activation in secondary lymphoid organs or trafficking to inflammatory sites.
Collapse
Affiliation(s)
- Hanaa Safya
- UMR1174, INSERM, Université Paris-Sud, Orsay, France
| | - Amine Mellouk
- UMR1174, INSERM, Université Paris-Sud, Orsay, France
| | - Julie Legrand
- Institut André Lwoff, CNRS, Université Paris-Sud, Villejuif, France
| | - Sylvain M Le Gall
- Institut André Lwoff, CNRS, Université Paris-Sud, Villejuif, France.,UMR 970, INSERM, Université Paris Descartes, Paris, France
| | - Mohcine Benbijja
- Institut André Lwoff, CNRS, Université Paris-Sud, Villejuif, France.,UMR 1012, INSERM, Université Paris-Sud, Le Kremlin Bicêtre, France
| | | | | | - Pierre Bobé
- UMR1174, INSERM, Université Paris-Sud, Orsay, France.,Institut André Lwoff, CNRS, Université Paris-Sud, Villejuif, France
| |
Collapse
|
50
|
Shen L, Bing T, Liu X, Wang J, Wang L, Zhang N, Shangguan D. Flow Cytometric Bead Sandwich Assay Based on a Split Aptamer. ACS APPLIED MATERIALS & INTERFACES 2018; 10:2312-2318. [PMID: 29276885 DOI: 10.1021/acsami.7b16192] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
A few aptamers still bind their targets after being split into two moieties. Split aptamers have shown great potential in the development of aptameric sensors. However, only a few split aptamers have been generated because of lack of knowledge on the binding structure of their parent aptamers. Here, we report the design of a new split aptamer and a flow cytometric bead sandwich assay using a split aptamer instead of double antibodies. Through DMS footprinting and mutation assay, we figured out the target-binding moiety and the structure-stabilizing moiety of the l-selectin aptamer, Sgc-3b. By separating the duplex strand in the structure-stabilizing moiety, we obtained a split aptamer that bound l-selectin. After optimization of one part of the split sequence to eliminate the nonspecific binding of the split sequence pair, we developed a split-aptamer-based cytometric bead assay (SACBA) for the detection of soluble l-selectin. SACBA showed good sensitivity and selectivity to l-selectin and was successfully applied for the detection of spiked l-selectin in the human serum. The strategies for generating split aptamers and designing the split-aptamer-based sandwich assay are simple and efficient and show good practicability in aptamer engineering.
Collapse
Affiliation(s)
- Luyao Shen
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Tao Bing
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Xiangjun Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Junyan Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Linlin Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Nan Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Dihua Shangguan
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| |
Collapse
|