1
|
Boulaftali Y, Massberg S, Nicolai L. Platelets in vascular inflammation: fire-fighters or pyromaniacs? Curr Opin Hematol 2025; 32:221-230. [PMID: 40359086 DOI: 10.1097/moh.0000000000000877] [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] [Indexed: 05/15/2025]
Abstract
PURPOSE OF REVIEW In this review, we aim to highlight recent insights into the mechanisms through which platelets contribute to vascular inflammation. We will discuss how platelets interact with other cellular players in the vascular milieu, their role in shaping inflammatory responses, and the potential therapeutic implications of targeting platelet function in inflammatory vascular diseases. RECENT FINDINGS Platelets are essential components in the processes of hemostasis and thrombosis. Their role is now widely acknowledged as far more complex than merely acting as "band-aids" or helping to "clog a pipe". Platelets are now recognized as crucial mediators in inflammatory reactions, particularly in various diseases of the vasculature, where they contribute to the onset and progression of injury. Through their interactions with leukocytes, vascular cells, and by supporting the coagulation cascade, platelets are able to finely regulate the extent and intensity of vascular damage. SUMMARY Recent findings underscore the remarkable diversity and functionality of platelets in vascular diseases. Mechanistic studies in preclinical models reveal promising therapeutic opportunities, which require further validation before being translated into clinical practice.
Collapse
Affiliation(s)
- Yacine Boulaftali
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Leo Nicolai
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
2
|
Knowles EEM, Peralta JM, Rodrigue AL, Mathias SR, Mollon J, Leandro AC, Curran JE, Blangero J, Glahn DC. Differential gene expression study in whole blood identifies candidate genes for psychosis in African American individuals. Schizophr Res 2025; 280:85-94. [PMID: 40267851 DOI: 10.1016/j.schres.2025.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 03/10/2025] [Accepted: 04/13/2025] [Indexed: 04/25/2025]
Abstract
Genome-wide association has identified regions of the genome that mediate risk for psychosis. It is possible that variants in these regions confer risk by altering gene expression. This work has predominantly been conducted in individuals of European descent and has focused narrowly on schizophrenia rather than psychosis as a syndrome. In the present study we investigated alterations in gene expression in African American individuals with a range of psychotic diagnoses to increase understanding of the etiology in an underserved population. We performed RNA-seq in whole bloody to survey the transcriptome in 126 patients with a psychosis-spectrum disorder and 217 healthy controls and applied differential gene expression analyses across the genome while controlling for age, sex, population stratification and batch. We found 18 differentially expressed genes (DEGs), some of the locations of the corresponding genes overlap with previously implicated regions for psychosis, but many of which were novel associations. Enrichment analysis of nominally significant genes (p < 0.05) revealed overrepresentation of biological processes relating to platelet, immune and cellular function, and sensory perception. Weighted gene co-expression network analysis, applied to identify modules of co-expressed genes associated with psychosis, revealed 10 modules, one of which was significantly associated with psychosis. This module was significantly enriched for DEGs, and for platelet function. These results support the potential role of immune function in the etiology of psychosis, identify novel candidate gene expression phenotypes that correspond to both established and new genomic regions, in individuals of African American ancestry.
Collapse
Affiliation(s)
- E E M Knowles
- Department of Psychiatry, Boston Children's Hospital, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| | - J M Peralta
- South Texas Diabetes and Obesity Institute and Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - A L Rodrigue
- Department of Psychiatry, Boston Children's Hospital, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - S R Mathias
- Department of Psychiatry, Boston Children's Hospital, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - J Mollon
- Department of Psychiatry, Boston Children's Hospital, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - A C Leandro
- South Texas Diabetes and Obesity Institute and Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - J E Curran
- South Texas Diabetes and Obesity Institute and Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - J Blangero
- South Texas Diabetes and Obesity Institute and Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - D C Glahn
- Department of Psychiatry, Boston Children's Hospital, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Olin Neuropsychiatric Research Center, Institute of Living, Hartford Hospital, Hartford, CT, USA
| |
Collapse
|
3
|
Mangiola S, Brown R, Zhan C, Berthelet J, Guleria S, Liyanage C, Ostrouska S, Wilcox J, Merdas M, Fuge-Larsen P, Bell C, Schröder J, Mielke LA, Mariadason JM, Tsao SCH, Chen Y, Yadav VK, Vodala S, Anderson RL, Merino D, Behren A, Yeo B, Papenfuss AT, Pal B. Circulating immune cells exhibit distinct traits linked to metastatic burden in breast cancer. Breast Cancer Res 2025; 27:73. [PMID: 40340807 PMCID: PMC12063295 DOI: 10.1186/s13058-025-01982-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 02/14/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Circulating immune cells play a crucial role in the anti-tumour immune response, yet the systemic immune system in metastatic breast cancers is not fully characterised. Investigating the cellular and molecular changes in peripheral blood mononuclear cells (PBMCs) from breast cancer patients could elucidate the role of circulating immune cells in metastasis and aid in identifying biomarkers for disease burden and progression. METHODS In this study, we characterised the systemic immune landscape associated with varying levels of metastatic burden by analysing the single-cell transcriptomes of PBMCs from breast cancer patients and healthy controls. Our research focused on identifying changes in immune cell composition, transcriptional programs, and immune-cell communication networks linked to metastatic burden. Additionally, we compared these PBMC features onto a single-cell atlas of primary breast tumours to study corresponding traits in tumour-infiltrating immune cells. RESULTS In metastatic breast cancer, PBMCs exhibit a significant downregulation of the adaptive immune system and a decreased number and activity of unconventional T cells, such as γδ T cells. Additionally, metastatic burden is associated with impaired cell communication pathways involved in immunomodulatory functions. We also identified a gene signature derived from myeloid cells shared between tumour immune infiltrates and circulating immune cells in breast cancer patients. CONCLUSIONS Our study provides a comprehensive single-cell molecular profile of the peripheral immune system in breast cancer, offering a valuable resource for understanding metastatic disease in terms of tumour burden. By identifying immune traits linked to metastasis, we have unveiled potential new biomarkers of metastatic disease.
Collapse
Affiliation(s)
- S Mangiola
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia.
- South Australian immunoGENomics Cancer Institute, Adelaide, SA, 5005, Australia.
| | - R Brown
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - C Zhan
- South Australian immunoGENomics Cancer Institute, Adelaide, SA, 5005, Australia
| | - J Berthelet
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - S Guleria
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - C Liyanage
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - S Ostrouska
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - J Wilcox
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - M Merdas
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - P Fuge-Larsen
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - C Bell
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
- Austin Health, Heidelberg, VIC, 3084, Australia
| | - J Schröder
- Peter Doherty Institute for Infection and Immunity, Parkville, VIC, 3052, Australia
- The University of Melbourne, Parkville, VIC, 3052, Australia
| | - L A Mielke
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - J M Mariadason
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - S Chang-Hao Tsao
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
- Austin Health, Heidelberg, VIC, 3084, Australia
| | - Y Chen
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - V K Yadav
- Rutgers New Jersey Medical School, Newark, NJ, USA
| | - S Vodala
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, USA
| | - R L Anderson
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - D Merino
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - A Behren
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - B Yeo
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
- Austin Health, Heidelberg, VIC, 3084, Australia
| | - A T Papenfuss
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia.
| | - B Pal
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia.
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.
| |
Collapse
|
4
|
Nisnboym M, Sneiderman CT, Jaswal AP, Xiong Z, Vincze SR, Sever RE, Zou H, Frederico SC, Agnihotri S, Hu B, Drappatz J, Pollack IF, Kohanbash G, Raphael I. Assessment of anti-CD69 antibody therapy alone or in combination with anti-PD-1 in murine GBM. Expert Rev Clin Immunol 2025; 21:239-247. [PMID: 39402706 DOI: 10.1080/1744666x.2024.2412770] [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: 06/30/2024] [Accepted: 09/27/2024] [Indexed: 02/02/2025]
Abstract
BACKGROUND Glioblastoma (GBM) is an aggressive cancer with limited treatment options. Immunotherapy targeting CD69, an early activation marker on T cells, has shown promise in preclinical models of non-CNS malignancies. This study investigates anti-CD69 therapy alone or in combination with anti-PD-1 in a preclinical GBM model. RESEARCH DESIGN AND METHODS CD69 expression in GBM patient tissues was analyzed using the TCGA database. Therapeutic efficacy of anti-CD69 was tested in a murine GBM model with different regimens. Immune cell populations in the tumor microenvironment (TME) were assessed by flow cytometry. RESULTS Increased CD69 expression was observed in GBM patients compared to normal brain tissue and was associated with worse prognosis. Anti-CD69 treatment reduced percentages of CD69+ immune cells but did not improve survival in GBM-bearing mice. Increased PD-1 expression on NK cells was observed following anti-CD69 treatment. Anti-CD69 treatment was not improved by the addition of anti-PD-1 in vivo. CONCLUSIONS This is the first study evaluating anti-CD69 therapy in a preclinical GBM model. Despite promising preclinical data in other cancers, anti-CD69 monotherapy or combination therapy with anti-PD-1 did not improve survival in this GBM model.
Collapse
MESH Headings
- Animals
- Glioblastoma/immunology
- Glioblastoma/drug therapy
- Glioblastoma/therapy
- Antigens, Differentiation, T-Lymphocyte/immunology
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Mice
- Lectins, C-Type/immunology
- Lectins, C-Type/antagonists & inhibitors
- Lectins, C-Type/metabolism
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Humans
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Brain Neoplasms/immunology
- Brain Neoplasms/drug therapy
- Brain Neoplasms/therapy
- Immunotherapy/methods
- Tumor Microenvironment/immunology
- Disease Models, Animal
- Killer Cells, Natural/immunology
- Cell Line, Tumor
- Immune Checkpoint Inhibitors/therapeutic use
- Female
Collapse
Affiliation(s)
- Michal Nisnboym
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, Tel-Aviv Sourasky Medical Center, Tel-Aviv University, Tel-Aviv, Israel
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Chaim T Sneiderman
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ambika P Jaswal
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zujian Xiong
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sarah R Vincze
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - ReidAnn E Sever
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Han Zou
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stephen C Frederico
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sameer Agnihotri
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Baoli Hu
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jan Drappatz
- Departments of Neurology and Medicine, Division of Hematology/Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Ian F Pollack
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Gary Kohanbash
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Itay Raphael
- Department of Neurological Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
5
|
Sugihara J, Iwamura C, Tateishi T, Hosoya T, Shimada S, Hirahara K, Yasuda S, Miyazaki Y. Prolonged high Myl9 levels are associated with the pathogenesis and respiratory symptom of post-acute COVID-19 syndrome: A 6-month follow-up study. Clinics (Sao Paulo) 2025; 80:100584. [PMID: 39879907 PMCID: PMC11814511 DOI: 10.1016/j.clinsp.2025.100584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Post-acute COVID-19 Syndrome (PACS) occurs in some COVID-19 patients long after acute infection and significantly affects patients' health. However, the mechanism by which PACS develops is unknown. Myosin light chain 9 (Myl9), produced by activated platelets, plays a role in immune dysregulation and microthrombi formation during acute COVID-19. However, in the PACS phase, the association between Myl9 and residual symptoms remains unclear, and further investigation is needed. METHODS In this prospective cohort study, serum Myl9 concentrations were measured in 195 COVID-19 patients during hospitalization and at 3- and 6-month follow-up visits. Gaussian mixture modeling was used to identify groups on the basis of Myl9 levels. Relationships between Myl9 levels and residual symptoms were evaluated. Clinical characteristics influencing Myl9 levels were analyzed via logistic regression. RESULTS A total of 304 serum samples from 195 patients were collected. Two distinct groups were identified in the Myl9 distribution with a cutoff of 386 ng/mL by Gaussian mixture modeling in this cohort. The high-Myl9 group presented significant residual respiratory symptoms at 6 months post-infection (p < 0.05). Elevated Myl9 levels at 6 months were correlated with increased neutrophil counts (p < 0.01) and respiratory comorbidities at diagnosis (p < 0.05) according to univariate regression analysis. Multivariate regression analysis confirmed the relationship between the neutrophil count and high Myl9 levels. CONCLUSION Prolonged high Myl9 levels are associated with respiratory symptoms, suggesting the potential involvement of prolonged inflammation or endothelial damage in PACS.
Collapse
Affiliation(s)
- Jun Sugihara
- Department of Respiratory Medicine, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Chiaki Iwamura
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tomoya Tateishi
- Department of Respiratory Medicine, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan.
| | - Tadashi Hosoya
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Sho Shimada
- Department of Respiratory Medicine, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Kiyoshi Hirahara
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shinsuke Yasuda
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Yasunari Miyazaki
- Department of Respiratory Medicine, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
Kozole SL, Beningo KA. Myosin Light Chains in the Progression of Cancer. Cells 2024; 13:2081. [PMID: 39768172 PMCID: PMC11674124 DOI: 10.3390/cells13242081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/06/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
The myosin light chains (MLCs) of non-muscle myosin II are known to regulate cellular architecture and generate cellular forces; they also have an increasingly emerging role in the progression of cancer. The phosphorylation state of the myosin light chains controls the activity of myosins that are implicated in invasion and proliferation. In cancers, when proliferation is greatly increased, cytokinesis relies on phosphorylated light chains to activate the contractile forces used to separate the cells. Likewise, during metastasis, kinase pathways culminate in aligning MLC structures for enhanced cell motility through stress fiber contraction and the accumulation of myosin filaments at the leading edge. This review summarizes the myosin light chain family members known to promote cancer progression and evidence of how their altered activities change the behavior of cells involving the mechanical-based processes of proliferation and cell movements during metastasis. In addition, myosin light chains impact the immune response to cancers and currently serve as biomarkers in staging this disease; a brief summary of these topics is provided at the end of the review.
Collapse
Affiliation(s)
| | - Karen A. Beningo
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA;
| |
Collapse
|
7
|
Noval Rivas M, Kocatürk B, Franklin BS, Arditi M. Platelets in Kawasaki disease: mediators of vascular inflammation. Nat Rev Rheumatol 2024; 20:459-472. [PMID: 38886559 DOI: 10.1038/s41584-024-01119-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 06/20/2024]
Abstract
Kawasaki disease, a systemic vasculitis that affects young children and can result in coronary artery aneurysms, is the leading cause of acquired heart disease among children. A hallmark of Kawasaki disease is increased blood platelet counts and platelet activation, which is associated with an increased risk of developing resistance to intravenous immunoglobulin and coronary artery aneurysms. Platelets and their releasate, including granules, microparticles, microRNAs and transcription factors, can influence innate immunity, enhance inflammation and contribute to vascular remodelling. Growing evidence indicates that platelets also interact with immune and non-immune cells to regulate inflammation. Platelets boost NLRP3 inflammasome activation and IL-1β production by human immune cells by releasing soluble mediators. Activated platelets form aggregates with leukocytes, such as monocytes and neutrophils, enhancing numerous functions of these cells and promoting thrombosis and inflammation. Leukocyte-platelet aggregates are increased in children with Kawasaki disease during the acute phase of the disease and can be used as biomarkers for disease severity. Here we review the role of platelets in Kawasaki disease and discuss progress in understanding the immune-effector role of platelets in amplifying inflammation related to Kawasaki disease vasculitis and therapeutic strategies targeting platelets or platelet-derived molecules.
Collapse
Affiliation(s)
- Magali Noval Rivas
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Begüm Kocatürk
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Bernardo S Franklin
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Moshe Arditi
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA.
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
8
|
Qin L, Li B, Wang S, Tang Y, Fahira A, Kou Y, Li T, Hu Z, Huang Z. Construction of an immune-related prognostic signature and lncRNA-miRNA-mRNA ceRNA network in acute myeloid leukemia. J Leukoc Biol 2024; 116:146-165. [PMID: 38393298 DOI: 10.1093/jleuko/qiae041] [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: 08/17/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
The progression of acute myeloid leukemia (AML) is influenced by the immune microenvironment in the bone marrow and dysregulated intracellular competing endogenous RNA (ceRNA) networks. Our study utilized data from UCSC Xena, The Cancer Genome Atlas Program, the Gene Expression Omnibus, and the Immunology Database and Analysis Portal. Using Cox regression analysis, we identified an immune-related prognostic signature. Genomic analysis of prognostic messenger RNA (mRNA) was conducted through Gene Set Cancer Analysis (GSCA), and a prognostic ceRNA network was constructed using the Encyclopedia of RNA Interactomes. Correlations between signature mRNAs and immune cell infiltration, checkpoints, and drug sensitivity were assessed using R software, gene expression profiling interactive analysis (GEPIA), and CellMiner, respectively. Adhering to the ceRNA hypothesis, we established a potential long noncoding RNA (lncRNA)/microRNA (miRNA)/mRNA regulatory axis. Our findings pinpointed 9 immune-related prognostic mRNAs (KIR2DL1, CSRP1, APOBEC3G, CKLF, PLXNC1, PNOC, ANGPT1, IL1R2, and IL3RA). GSCA analysis revealed the impact of copy number variations and methylation on AML. The ceRNA network comprised 14 prognostic differentially expressed lncRNAs (DE-lncRNAs), 6 prognostic DE-miRNAs, and 3 prognostic immune-related DE-mRNAs. Correlation analyses linked these mRNAs' expression to 22 immune cell types and 6 immune checkpoints, with potential sensitivity to 27 antitumor drugs. Finally, we identified a potential LINC00963/hsa-miR-431-5p/CSRP1 axis. This study offers innovative insights for AML diagnosis and treatment through a novel immune-related signature and ceRNA axis. Identified novel biomarkers, including 2 mRNAs (CKLF, PNOC), 1 miRNA (hsa-miR-323a-3p), and 10 lncRNAs (SNHG25, LINC01857, AL390728.6, AC127024.5, Z83843.1, AP002884.1, AC007038.1, AC112512, AC020659.1, AC005921.3) present promising candidates as potential targets for precision medicine, contributing to the ongoing advancements in the field.
Collapse
Affiliation(s)
- Ling Qin
- Department of Hematology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, No. 24 Jinghua Road, Jianxi District, Luoyang 471003, China
| | - Boya Li
- Department of Hematology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, No. 24 Jinghua Road, Jianxi District, Luoyang 471003, China
| | - Shijie Wang
- Department of Hematology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, No. 24 Jinghua Road, Jianxi District, Luoyang 471003, China
| | - Yulai Tang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer-Aided Drug Design of Dongguan City, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, No. 1 Xincheng Road, Songshan Lake District, Dongguan 523808, Guangdong, China
| | - Aamir Fahira
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer-Aided Drug Design of Dongguan City, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, No. 1 Xincheng Road, Songshan Lake District, Dongguan 523808, Guangdong, China
| | - Yanqi Kou
- Department of Hematology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, No. 24 Jinghua Road, Jianxi District, Luoyang 471003, China
| | - Tong Li
- Department of Hematology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, No. 24 Jinghua Road, Jianxi District, Luoyang 471003, China
| | - Zhigang Hu
- School of Medical Technology and Engineering, Henan University of Science and Technology, No.263 Kaiyuan Avenue, Luolong District, Luoyang 471000, China
| | - Zunnan Huang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer-Aided Drug Design of Dongguan City, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, No. 1 Xincheng Road, Songshan Lake District, Dongguan 523808, Guangdong, China
| |
Collapse
|
9
|
Bhuiyan SA, Xu M, Yang L, Semizoglou E, Bhatia P, Pantaleo KI, Tochitsky I, Jain A, Erdogan B, Blair S, Cat V, Mwirigi JM, Sankaranarayanan I, Tavares-Ferreira D, Green U, McIlvried LA, Copits BA, Bertels Z, Del Rosario JS, Widman AJ, Slivicki RA, Yi J, Sharif-Naeini R, Woolf CJ, Lennerz JK, Whited JL, Price TJ, Gereau RW, Renthal W. Harmonized cross-species cell atlases of trigeminal and dorsal root ganglia. SCIENCE ADVANCES 2024; 10:eadj9173. [PMID: 38905344 PMCID: PMC11804847 DOI: 10.1126/sciadv.adj9173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 05/16/2024] [Indexed: 06/23/2024]
Abstract
Sensory neurons in the dorsal root ganglion (DRG) and trigeminal ganglion (TG) are specialized to detect and transduce diverse environmental stimuli to the central nervous system. Single-cell RNA sequencing has provided insights into the diversity of sensory ganglia cell types in rodents, nonhuman primates, and humans, but it remains difficult to compare cell types across studies and species. We thus constructed harmonized atlases of the DRG and TG that describe and facilitate comparison of 18 neuronal and 11 non-neuronal cell types across six species and 31 datasets. We then performed single-cell/nucleus RNA sequencing of DRG from both human and the highly regenerative axolotl and found that the harmonized atlas also improves cell type annotation, particularly of sparse neuronal subtypes. We observed that the transcriptomes of sensory neuron subtypes are broadly similar across vertebrates, but the expression of functionally important neuropeptides and channels can vary notably. The resources presented here can guide future studies in comparative transcriptomics, simplify cell-type nomenclature differences across studies, and help prioritize targets for future analgesic development.
Collapse
Affiliation(s)
- Shamsuddin A. Bhuiyan
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Mengyi Xu
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Alan Edwards Center for Research on Pain and Department of Physiology, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Lite Yang
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Evangelia Semizoglou
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Parth Bhatia
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Katerina I. Pantaleo
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ivan Tochitsky
- F.M. Kirby Neurobiology Center and Department of Neurobiology, Boston Children’s Hospital and Harvard Medical School, 3 Blackfan Cir., Boston, MA 02115, USA
| | - Aakanksha Jain
- F.M. Kirby Neurobiology Center and Department of Neurobiology, Boston Children’s Hospital and Harvard Medical School, 3 Blackfan Cir., Boston, MA 02115, USA
| | - Burcu Erdogan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Steven Blair
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Victor Cat
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Juliet M. Mwirigi
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Diana Tavares-Ferreira
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Ursula Green
- Department of Pathology, Center for Integrated Diagnostics, Massachussetts General Hospital and Havard Medical School, Boston, MA 02114, USA
| | - Lisa A. McIlvried
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Bryan A. Copits
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Zachariah Bertels
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - John S. Del Rosario
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Allie J. Widman
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Richard A. Slivicki
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Jiwon Yi
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Reza Sharif-Naeini
- Alan Edwards Center for Research on Pain and Department of Physiology, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Clifford J. Woolf
- F.M. Kirby Neurobiology Center and Department of Neurobiology, Boston Children’s Hospital and Harvard Medical School, 3 Blackfan Cir., Boston, MA 02115, USA
| | - Jochen K. Lennerz
- Department of Pathology, Center for Integrated Diagnostics, Massachussetts General Hospital and Havard Medical School, Boston, MA 02114, USA
| | - Jessica L. Whited
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Theodore J. Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Robert W. Gereau
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - William Renthal
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
10
|
Wu S, Xiao X, Zhang Y, Zhang X, Wang G, Peng Q. Novel endotypes of antisynthetase syndrome identified independent of anti-aminoacyl transfer RNA synthetase antibody specificity that improve prognostic stratification. Ann Rheum Dis 2024; 83:775-786. [PMID: 38395605 DOI: 10.1136/ard-2023-225284] [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: 11/14/2023] [Accepted: 02/09/2024] [Indexed: 02/25/2024]
Abstract
OBJECTIVES To systemically analyse the heterogeneity in the clinical manifestations and prognoses of patients with antisynthetase syndrome (ASS) and evaluate the transcriptional signatures related to different clinical phenotypes. METHODS A total of 701 patients with ASS were retrospectively enrolled. The clinical presentation and prognosis were assessed in association with four anti-aminoacyl transfer RNA synthetase (ARS) antibodies: anti-Jo1, anti-PL7, anti-PL12 and anti-EJ. Unsupervised machine learning was performed for patient clustering independent of anti-ARS antibodies. Transcriptome sequencing was conducted in clustered ASS patients and healthy controls. RESULTS Patients with four different anti-ARS antibody subtypes demonstrated no significant differences in the incidence of rapidly progressive interstitial lung disease (RP-ILD) or prognoses. Unsupervised machine learning, independent of anti-ARS specificity, identified three endotypes with distinct clinical features and outcomes. Endotype 1 (RP-ILD cluster, 23.7%) was characterised by a high incidence of RP-ILD and a high mortality rate. Endotype 2 (dermatomyositis (DM)-like cluster, 14.5%) corresponded to patients with DM-like skin and muscle symptoms with an intermediate prognosis. Endotype 3 (arthritis cluster, 61.8%) was characterised by arthritis and mechanic's hands, with a good prognosis. Transcriptome sequencing revealed that the different endotypes had distinct gene signatures and biological processes. CONCLUSIONS Anti-ARS antibodies were not significant in stratifying ASS patients into subgroups with greater homogeneity in RP-ILD and prognoses. Novel ASS endotypes were identified independent of anti-ARS specificity and differed in clinical outcomes and transcriptional signatures, providing new insights into the pathogenesis of ASS.
Collapse
Affiliation(s)
- Shiyu Wu
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, People's Republic of China
- Department of Rheumatology, Key Lab of Myositis, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Xinyue Xiao
- Department of Rheumatology, Key Lab of Myositis, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Yingfang Zhang
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, People's Republic of China
- Department of Rheumatology, Key Lab of Myositis, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Xinxin Zhang
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, People's Republic of China
- Department of Rheumatology, Key Lab of Myositis, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Guochun Wang
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, People's Republic of China
- Department of Rheumatology, Key Lab of Myositis, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Qinglin Peng
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, People's Republic of China
- Department of Rheumatology, Key Lab of Myositis, China-Japan Friendship Hospital, Beijing, People's Republic of China
| |
Collapse
|
11
|
Ahmad I, Omura S, Sato F, Park AM, Khadka S, Gavins FNE, Tanaka H, Kimura MY, Tsunoda I. Exploring the Role of Platelets in Virus-Induced Inflammatory Demyelinating Disease and Myocarditis. Int J Mol Sci 2024; 25:3460. [PMID: 38542433 PMCID: PMC10970283 DOI: 10.3390/ijms25063460] [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: 02/20/2024] [Revised: 03/14/2024] [Accepted: 03/16/2024] [Indexed: 12/26/2024] Open
Abstract
Theiler's murine encephalomyelitis virus (TMEV) infection has been used as a mouse model for two virus-induced organ-specific immune-mediated diseases. TMEV-induced demyelinating disease (TMEV-IDD) in the central nervous system (CNS) is a chronic inflammatory disease with viral persistence and an animal model of multiple sclerosis (MS) in humans. TMEV infection can also cause acute myocarditis with viral replication and immune cell infiltration in the heart, leading to cardiac fibrosis. Since platelets have been reported to modulate immune responses, we aimed to determine the role of platelets in TMEV infection. In transcriptome analyses of platelets, distinct sets of immune-related genes, including major histocompatibility complex (MHC) class I, were up- or downregulated in TMEV-infected mice at different time points. We depleted platelets from TMEV-infected mice by injecting them with platelet-specific antibodies. The platelet-depleted mice had significantly fewer viral antigen-positive cells in the CNS. Platelet depletion reduced the severities of TMEV-IDD and myocarditis, although the pathology scores did not reach statistical significance. Immunologically, the platelet-depleted mice had an increase in interferon (IFN)-γ production with a higher anti-TMEV IgG2a/IgG1 ratio. Thus, platelets may play roles in TMEV infection, such as gene expression, viral clearance, and anti-viral antibody isotype responses.
Collapse
Affiliation(s)
- Ijaz Ahmad
- Department of Microbiology, Faculty of Medicine, Kindai University, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan; (I.A.); (S.O.); (F.S.); (A.-M.P.); (S.K.)
| | - Seiichi Omura
- Department of Microbiology, Faculty of Medicine, Kindai University, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan; (I.A.); (S.O.); (F.S.); (A.-M.P.); (S.K.)
| | - Fumitaka Sato
- Department of Microbiology, Faculty of Medicine, Kindai University, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan; (I.A.); (S.O.); (F.S.); (A.-M.P.); (S.K.)
| | - Ah-Mee Park
- Department of Microbiology, Faculty of Medicine, Kindai University, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan; (I.A.); (S.O.); (F.S.); (A.-M.P.); (S.K.)
- Department of Arts and Sciences, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan
| | - Sundar Khadka
- Department of Microbiology, Faculty of Medicine, Kindai University, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan; (I.A.); (S.O.); (F.S.); (A.-M.P.); (S.K.)
- Department of Immunology, Duke University, Durham, NC 27708, USA
| | - Felicity N. E. Gavins
- Department of Biosciences, Centre for Inflammation Research and Translational Medicine, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK;
| | - Hiroki Tanaka
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa 078-8510, Japan;
| | - Motoko Y. Kimura
- Department of Experimental Immunology, Graduate School of Medicine, Chiba University, Chiba 263-8522, Japan;
| | - Ikuo Tsunoda
- Department of Microbiology, Faculty of Medicine, Kindai University, 377-2 Ohnohigashi, Osakasayama, Osaka 589-8511, Japan; (I.A.); (S.O.); (F.S.); (A.-M.P.); (S.K.)
| |
Collapse
|
12
|
Pérez-Jurado LA, Cáceres A, Balagué-Dobón L, Esko T, López de Heredia M, Quintela I, Cruz R, Lapunzina P, Carracedo Á, González JR. Clonal chromosomal mosaicism and loss of chromosome Y in elderly men increase vulnerability for SARS-CoV-2. Commun Biol 2024; 7:202. [PMID: 38374351 PMCID: PMC10876565 DOI: 10.1038/s42003-024-05805-6] [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: 02/18/2022] [Accepted: 01/11/2024] [Indexed: 02/21/2024] Open
Abstract
The pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2, COVID-19) had an estimated overall case fatality ratio of 1.38% (pre-vaccination), being 53% higher in males and increasing exponentially with age. Among 9578 individuals diagnosed with COVID-19 in the SCOURGE study, we found 133 cases (1.42%) with detectable clonal mosaicism for chromosome alterations (mCA) and 226 males (5.08%) with acquired loss of chromosome Y (LOY). Individuals with clonal mosaic events (mCA and/or LOY) showed a 54% increase in the risk of COVID-19 lethality. LOY is associated with transcriptomic biomarkers of immune dysfunction, pro-coagulation activity and cardiovascular risk. Interferon-induced genes involved in the initial immune response to SARS-CoV-2 are also down-regulated in LOY. Thus, mCA and LOY underlie at least part of the sex-biased severity and mortality of COVID-19 in aging patients. Given its potential therapeutic and prognostic relevance, evaluation of clonal mosaicism should be implemented as biomarker of COVID-19 severity in elderly people.
Collapse
Affiliation(s)
- Luis A Pérez-Jurado
- Genetics Unit, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
- Genetics Service, Hospital del Mar & Hospital del Mar Research Institute (IMIM), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Barcelona, Spain.
| | - Alejandro Cáceres
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
- Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Laura Balagué-Dobón
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
- Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Tonu Esko
- Estonian Genome Science Centre, University of Tartu, Tartu, Estonia
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Miguel López de Heredia
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Barcelona, Spain
| | - Inés Quintela
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Barcelona, Spain
- Centro Nacional de Genotipado (CEGEN), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Raquel Cruz
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Barcelona, Spain
- Centro Nacional de Genotipado (CEGEN), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Pablo Lapunzina
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Barcelona, Spain
- Instituto de Genética Médica y Molecular (INGEMM), Hospital Universitario La Paz-IDIPAZ, Madrid, Spain
- ERN-ITHACA-European Reference Network, Paris, France
| | - Ángel Carracedo
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Barcelona, Spain
- Centro Nacional de Genotipado (CEGEN), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica, Sistema Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Juan R González
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain.
- Department of Mathematics, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| |
Collapse
|
13
|
Li Y, Gu Y, Yang P, Wang Y, Yu X, Li Y, Jin Z, Xu L. CD69 is a Promising Immunotherapy and Prognosis Prediction Target in Cancer. Immunotargets Ther 2024; 13:1-14. [PMID: 38223406 PMCID: PMC10787557 DOI: 10.2147/itt.s439969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 12/22/2023] [Indexed: 01/16/2024] Open
Abstract
Immunotherapy utilizing T cells that attack tumors is a promising strategy for treatment, but immune suppressive T cell subsets, such as regulatory T cell (Treg), and immune checkpoint molecules, including programmed death-1 (PD-1), can suppress the intensity of a T cell immune reaction and thereby impair tumor clearance. Cluster of differentiation 69 (CD69), known as an early leukocyte activation marker, can be used as a measure or early marker of T cell activation. In recent years, the functions of CD69 in the regulation of Treg/Th17 (T helper cell 17) differentiation and in the tissue retention of T cells have attracted considerable interest. These functions are related to the role of CD69 in immune suppression in tumor environments (TME). In this review, we first summarized current perspectives in the biological function of CD69 and demonstrated that CD69 acts as a regulator of T cell activation, differentiation, retention, and exhaustion. Then, we discussed recent advances in understanding of CD69 deficiency and anti-CD69 antibody administration and shed light on the value of targeting on CD69 for cancer immunotherapy and prognosis prediction.
Collapse
Affiliation(s)
- Yuchen Li
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Yinfeng Gu
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Pengyue Yang
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Yan Wang
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Xibao Yu
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Yangqiu Li
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Zhenyi Jin
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, People’s Republic of China
- Department of Pathology, School of Medicine, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Ling Xu
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, People’s Republic of China
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, 510632, People’s Republic of China
| |
Collapse
|
14
|
Deng S, Cheng D, Wang J, Gu J, Xue Y, Jiang Z, Qin L, Mao F, Cao Y, Cai K. MYL9 expressed in cancer-associated fibroblasts regulate the immune microenvironment of colorectal cancer and promotes tumor progression in an autocrine manner. J Exp Clin Cancer Res 2023; 42:294. [PMID: 37926835 PMCID: PMC10626665 DOI: 10.1186/s13046-023-02863-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 10/12/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND The tumor microenvironment (TME) is an important factor that regulates the progression of colorectal cancer (CRC). Cancer-associated fibroblasts (CAFs) are the main mesenchymal cells in the TME and play a vital role in tumor progression; however, the specific underlying mechanisms require further study. METHODS Multiple single-cell and transcriptome data were analyzed and validated. Primary CAFs isolation, CCK8 assay, co-culture assay, western blotting, multiple immunofluorescence, qRT-PCR, ELISA, immunoprecipitation, ChIP, double luciferase, and animal experiments were used to explore the potential mechanism of MYL9 regulation in CRC. RESULTS Our findings revealed that MYL9 was predominantly localized and expressed in CAFs rather than in CRC cells, and bioinformatics analysis revealed that high MYL9 expression was strongly associated with poor overall and disease-free survival in various tumors. In addition, high MYL9 expression is closely associated with M2 macrophage infiltration, which can lead to an immunosuppressive microenvironment in CRC, making it insensitive to immunotherapy. Mechanically, MYL9 can regulate the secretion of CAFs on CCL2 and TGF-β1, thus affecting the immune microenvironment and progression of CRC. In addition, MYL9 bounded with IQGAP1 to regulate CCL2 and TGF-β1 secretion through the ERK 1/2 pathway, and CCL2 and TGF-β1 synergistically promoted CRC cells progression through the PI3K-AKT pathway. Furthermore, MYL9 promotes epithelial-mesenchymal transition (EMT) in CRC. During the upstream regulation of MYL9 in CAFs, we found that the EMT transcription factor ZEB1 could bind to the MYL9 promoter in CAFs, enhancing the activity and function of MYL9. Therefore, MYL9 is predominantly expressed in CAFs and can indirectly influence tumor biology and EMT by affecting CAFs protein expression in CRC. CONCLUSIONS MYL9 regulates the secretion of cytokines and chemokines in CAFs, which can affect the immune microenvironment of CRC and promote CRC progression. The relationship between MYL9 expression and CRC clinical staging and immunotherapy is closer in CAFs than in tumor cells; therefore, studies using CAFs as a model deserve more attention when exploring tumor molecular targets in clinical research.
Collapse
Affiliation(s)
- Shenghe Deng
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Denglong Cheng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jun Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Junnan Gu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yifan Xue
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhenxing Jiang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Le Qin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fuwei Mao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yinghao Cao
- Department of Digestive Surgical Oncology, Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Kailin Cai
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
15
|
Roato I, Pavone L, Pedraza R, Bosso I, Baima G, Erovigni F, Mussano F. Denosumab and Zoledronic Acid Differently Affect Circulating Immune Subsets: A Possible Role in the Onset of MRONJ. Cells 2023; 12:2430. [PMID: 37887274 PMCID: PMC10605172 DOI: 10.3390/cells12202430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/29/2023] [Accepted: 10/08/2023] [Indexed: 10/28/2023] Open
Abstract
This work investigated whether the anti-resorptive drugs (ARDs) zoledronic acid (Zol) and denosumab (Dmab) affect differently the levels of circulating immune cell subsets, possibly predicting the risk of developing medication-related ONJ (MRONJ) during the first 18 months of treatment. Blood samples were collected from 10 bone metastatic breast cancer patients receiving cyclin inhibitors at 0, 6, 12, and 18 months from the beginning of Dmab or Zol treatment. Eight breast cancer patients already diagnosed with MRONJ and treated with cyclin inhibitors and ARDs were in the control group. PBMCs were isolated; the trend of circulating immune subsets during the ARD treatment was monitored, and 12 pro-inflammatory cytokines were analyzed in sera using flow cytometry. In Dmab-treated patients, activated T cells were stable or increased, as were the levels of IL-12, TNF-α, GM-CSF, IL-5, and IL-10, sustaining them. In Zol-treated patients, CD8+T cells decreased, and the level of IFN-γ was undetectable. γδT cells were not altered in Dmab-treated patients, while they dramatically decreased in Zol-treated patients. In the MRONJ control group, Zol-ONJ patients showed a reduction in activated T cells and γδT cells compared to Dmab-ONJ patients. Dmab was less immunosuppressive than Zol, not affecting γδT cells and increasing activated T cells.
Collapse
Affiliation(s)
- Ilaria Roato
- Bone and Dental Bioengineering Laboratory, CIR-Dental School, Department of Surgical Sciences, University of Turin, Via Nizza 230, 10126 Turin, Italy; (L.P.); (R.P.); (G.B.); (F.M.)
| | - Lorenzo Pavone
- Bone and Dental Bioengineering Laboratory, CIR-Dental School, Department of Surgical Sciences, University of Turin, Via Nizza 230, 10126 Turin, Italy; (L.P.); (R.P.); (G.B.); (F.M.)
| | - Riccardo Pedraza
- Bone and Dental Bioengineering Laboratory, CIR-Dental School, Department of Surgical Sciences, University of Turin, Via Nizza 230, 10126 Turin, Italy; (L.P.); (R.P.); (G.B.); (F.M.)
- Institute of Sciences and Technologies for Sustainable Energy and Mobility, National Council of Research, 10135 Turin, Italy
- DIMEAS, Politecnico di Torino, 10129 Turin, Italy
| | - Ilaria Bosso
- CIR-Dental School, Città della Scienza e della Salute, 10126 Turin, Italy; (I.B.); (F.E.)
| | - Giacomo Baima
- Bone and Dental Bioengineering Laboratory, CIR-Dental School, Department of Surgical Sciences, University of Turin, Via Nizza 230, 10126 Turin, Italy; (L.P.); (R.P.); (G.B.); (F.M.)
- DIMEAS, Politecnico di Torino, 10129 Turin, Italy
| | - Francesco Erovigni
- CIR-Dental School, Città della Scienza e della Salute, 10126 Turin, Italy; (I.B.); (F.E.)
| | - Federico Mussano
- Bone and Dental Bioengineering Laboratory, CIR-Dental School, Department of Surgical Sciences, University of Turin, Via Nizza 230, 10126 Turin, Italy; (L.P.); (R.P.); (G.B.); (F.M.)
| |
Collapse
|
16
|
Susukida T, Sasaki SI, Shirayanagi T, Aoki S, Ito K, Hayakawa Y. Drug-induced altered self-presentation increases tumor immunogenicity. Biomed Pharmacother 2023; 165:115241. [PMID: 37523987 DOI: 10.1016/j.biopha.2023.115241] [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: 06/08/2023] [Revised: 07/25/2023] [Accepted: 07/25/2023] [Indexed: 08/02/2023] Open
Abstract
Anti-human immunodeficiency virus (HIV) drug abacavir (ABC) binds to the specific allele of human leukocyte antigen (HLA-B*57:01) and activates CD8+ T cells by presenting altered abnormal peptides. Here, we examined the effect of ABC-induced altered self-presentation by HLA-B*57:01 on immunogenicity of cancer cells and CD8+ T-cell-dependent anti-tumor immunity. We established human-mouse chimeric HLA-B*57:01-expressing tumor cell lines (B16F10 and 3LL) and tested the anti-tumor effect of ABC in vivo. ABC treatment inhibited the growth of HLA-B*57:01-expressing tumors by a CD8+ T-cell-dependent mechanism. ABC treatment induced CXCR3-dependent infiltration of CD8+ T cells into HLA-B*57:01-expressing tumors, and activated those tumor-infiltrating CD8+ T cells to proliferate and secrete IFN-γ. The activation of CD8+ T cells using drug-induced altered self-presentation may be a new strategy to increase tumor immunogenicity and improve the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Takeshi Susukida
- Laboratory of Cancer Biology and Immunology, Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - So-Ichiro Sasaki
- Laboratory of Cancer Biology and Immunology, Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Tomohiro Shirayanagi
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Shigeki Aoki
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Kousei Ito
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Yoshihiro Hayakawa
- Laboratory of Cancer Biology and Immunology, Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan.
| |
Collapse
|
17
|
Bhuiyan SA, Xu M, Yang L, Semizoglou E, Bhatia P, Pantaleo KI, Tochitsky I, Jain A, Erdogan B, Blair S, Cat V, Mwirigi JM, Sankaranarayanan I, Tavares-Ferreira D, Green U, McIlvried LA, Copits BA, Bertels Z, Del Rosario JS, Widman AJ, Slivicki RA, Yi J, Woolf CJ, Lennerz JK, Whited JL, Price TJ, Gereau RW, Renthal W. Harmonized cross-species cell atlases of trigeminal and dorsal root ganglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.04.547740. [PMID: 37461736 PMCID: PMC10350076 DOI: 10.1101/2023.07.04.547740] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Peripheral sensory neurons in the dorsal root ganglion (DRG) and trigeminal ganglion (TG) are specialized to detect and transduce diverse environmental stimuli including touch, temperature, and pain to the central nervous system. Recent advances in single-cell RNA-sequencing (scRNA-seq) have provided new insights into the diversity of sensory ganglia cell types in rodents, non-human primates, and humans, but it remains difficult to compare transcriptomically defined cell types across studies and species. Here, we built cross-species harmonized atlases of DRG and TG cell types that describe 18 neuronal and 11 non-neuronal cell types across 6 species and 19 studies. We then demonstrate the utility of this harmonized reference atlas by using it to annotate newly profiled DRG nuclei/cells from both human and the highly regenerative axolotl. We observe that the transcriptomic profiles of sensory neuron subtypes are broadly similar across vertebrates, but the expression of functionally important neuropeptides and channels can vary notably. The new resources and data presented here can guide future studies in comparative transcriptomics, simplify cell type nomenclature differences across studies, and help prioritize targets for future pain therapy development.
Collapse
Affiliation(s)
- Shamsuddin A Bhuiyan
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Mengyi Xu
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Alan Edwards Center for Research on Pain and Department of Physiology, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Lite Yang
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Program in Neurosciences, Division of Biology and Biomedical Sciences, Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Evangelia Semizoglou
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Parth Bhatia
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Katerina I Pantaleo
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ivan Tochitsky
- F.M. Kirby Neurobiology Center and Department of Neurobiology, Boston Children's Hospital and Harvard Medical School, 3 Blackfan Cir. Boston, MA 02115
| | - Aakanksha Jain
- F.M. Kirby Neurobiology Center and Department of Neurobiology, Boston Children's Hospital and Harvard Medical School, 3 Blackfan Cir. Boston, MA 02115
| | - Burcu Erdogan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, 02138
| | - Steven Blair
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, 02138
| | - Victor Cat
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, 02138
| | - Juliet M Mwirigi
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080
| | - Diana Tavares-Ferreira
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080
| | - Ursula Green
- Department of Pathology, Center for Integrated Diagnostics, Massachussetts General Hospital and Havard Medical School, Boston, MA 02114
| | - Lisa A McIlvried
- Program in Neurosciences, Division of Biology and Biomedical Sciences, Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Bryan A Copits
- Program in Neurosciences, Division of Biology and Biomedical Sciences, Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Zachariah Bertels
- Program in Neurosciences, Division of Biology and Biomedical Sciences, Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - John S Del Rosario
- Program in Neurosciences, Division of Biology and Biomedical Sciences, Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Allie J Widman
- Program in Neurosciences, Division of Biology and Biomedical Sciences, Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Richard A Slivicki
- Program in Neurosciences, Division of Biology and Biomedical Sciences, Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Jiwon Yi
- Program in Neurosciences, Division of Biology and Biomedical Sciences, Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Clifford J Woolf
- F.M. Kirby Neurobiology Center and Department of Neurobiology, Boston Children's Hospital and Harvard Medical School, 3 Blackfan Cir. Boston, MA 02115
| | - Jochen K Lennerz
- Department of Pathology, Center for Integrated Diagnostics, Massachussetts General Hospital and Havard Medical School, Boston, MA 02114
| | - Jessica L Whited
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, 02138
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080
| | - Robert W Gereau
- Program in Neurosciences, Division of Biology and Biomedical Sciences, Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - William Renthal
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
18
|
Multiple Genetic Loci Associated with Pug Dog Thoracolumbar Myelopathy. Genes (Basel) 2023; 14:genes14020385. [PMID: 36833311 PMCID: PMC9957375 DOI: 10.3390/genes14020385] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Pug dogs with thoracolumbar myelopathy (PDM) present with a specific clinical phenotype that includes progressive pelvic limb ataxia and paresis, commonly accompanied by incontinence. Vertebral column malformations and lesions, excessive scar tissue of the meninges, and central nervous system inflammation have been described. PDM has a late onset and affects more male than female dogs. The breed-specific presentation of the disorder suggests that genetic risk factors are involved in the disease development. To perform a genome-wide search for PDM-associated loci, we applied a Bayesian model adapted for mapping complex traits (BayesR) and a cross-population extended haplotype homozygosity test (XP-EHH) in 51 affected and 38 control pugs. Nineteen associated loci (harboring 67 genes in total, including 34 potential candidate genes) and three candidate regions under selection (with four genes within or next to the signal) were identified. The multiple candidate genes identified have implicated functions in bone homeostasis, fibrotic scar tissue, inflammatory responses, or the formation, regulation, and differentiation of cartilage, suggesting the potential relevance of these processes to the pathogenesis of PDM.
Collapse
|
19
|
Kobayashi H, Kimura MY, Hasegawa I, Suganuma E, Ikehara Y, Azuma K, Ito T, Ebata R, Kurashima Y, Kawasaki Y, Shiko Y, Saito N, Iwase H, Lee Y, Noval Rivas M, Arditi M, Zuka M, Hamada H, Nakayama T. Increased Myosin light chain 9 expression during Kawasaki disease vasculitis. Front Immunol 2023; 13:1036672. [PMID: 36685558 PMCID: PMC9853906 DOI: 10.3389/fimmu.2022.1036672] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction Kawasaki disease (KD) is an acute systemic vasculitis that predominantly afflicts children. KD development is known to be associated with an aberrant immune response and abnormal platelet activation, however its etiology is still largely unknown. Myosin light chain 9 (Myl9) is known to regulate cellular contractility of both non-muscle and smooth muscle cells, and can be released from platelets, whereas any relations of Myl9 expression to KD vasculitis have not been examined. Methods Plasma Myl9 concentrations in KD patients and children with febrile illness were measured and associated with KD clinical course and prognosis. Myl9 release from platelets in KD patients was also evaluated in vitro. Myl9 expression was determined in coronary arteries from Lactobacillus casei cell wall extract (LCWE)-injected mice that develop experimental KD vasculitis, as well as in cardiac tissues obtained at autopsy from KD patients. Results and discussion Plasma Myl9 levels were significantly higher in KD patients during the acute phase compared with healthy controls or patients with other febrile illnesses, declined following IVIG therapy in IVIG-responders but not in non-responders. In vitro, platelets from KD patients released Myl9 independently of thrombin stimulation. In the LCWE-injected mice, Myl9 was detected in cardiac tissue at an early stage before inflammatory cell infiltration was observed. In tissues obtained at autopsy from KD patients, the highest Myl9 expression was observed in thrombi during the acute phase and in the intima and adventitia of coronary arteries during the chronic phase. Thus, our studies show that Myl9 expression is significantly increased during KD vasculitis and that Myl9 levels may be a useful biomarker to estimate inflammation and IVIG responsiveness to KD.
Collapse
Affiliation(s)
- Hironobu Kobayashi
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Experimental Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Motoko Y. Kimura
- Department of Experimental Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Chiba University “Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Japan Initiative for World-leading Vaccine Research and Development Centers, Japan Agency for Medical Research and Development (AMED), Chiba, Japan, Chiba, Japan
| | - Ichita Hasegawa
- Department of Experimental Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Eisuke Suganuma
- Division of Infectious Diseases and Immunology, Allergy, Saitama Children’s Medical Center, Saitama, Japan
| | - Yuzuru Ikehara
- Department of Molecular and Tumor Pathology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kazuhiko Azuma
- Department of Molecular and Tumor Pathology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshihiro Ito
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ryota Ebata
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yosuke Kurashima
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yohei Kawasaki
- Clinical Research Center, Chiba University Hospital, Chiba, Japan
| | - Yuki Shiko
- Clinical Research Center, Chiba University Hospital, Chiba, Japan
| | - Naoki Saito
- Department of Legal Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hirotaro Iwase
- Department of Legal Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Youngho Lee
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Moshe Arditi
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Masahiko Zuka
- Department of Forensic Medicine and Pathology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Hiromichi Hamada
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Japan Agency for Medical Research and Development (AMED)-Core Research for Evolutional Science and Technology (CREST), AMED, Chiba, Japan
| |
Collapse
|
20
|
Zhang Y, Li Y, Zuo Z, Li T, An Y, Zhang W. An epithelial-mesenchymal transition-related mRNA signature associated with the prognosis, immune infiltration and therapeutic response of colon adenocarcinoma. Pathol Oncol Res 2023; 29:1611016. [PMID: 36910014 PMCID: PMC9998511 DOI: 10.3389/pore.2023.1611016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/14/2023] [Indexed: 03/14/2023]
Abstract
Background: Epithelial-mesenchymal transition (EMT) is closely associated with cancer cell metastasis. Colon adenocarcinoma (COAD) is one of the most common malignancies in the world, and its metastasis leading to poor prognosis remains a challenge for clinicians. The purpose of this study was to explore the prognostic value of EMT-related genes (EMTRGs) by bioinformatics analysis and to develop a new EMTRGs prognostic signature for COAD. Methods: The TCGA-COAD dataset was downloaded from the TCGA portal as the training cohort, and the GSE17538 and GSE29621 datasets were obtained from the GEO database as the validation cohort. The best EMTRGs prognostic signature was constructed by differential expression analysis, Cox, and LASSO regression analysis. Gene set enrichment analysis (GSEA) is used to reveal pathways that are enriched in high-risk and low-risk groups. Differences in tumor immune cell levels were analyzed using microenvironmental cell population counter and single sample gene set enrichment analysis. Subclass mapping analysis and Genomics of Drug Sensitivity in Cancer were applied for prediction of immunotherapy response and chemotherapy response, respectively. Results: A total of 77 differentially expressed EMTRGs were identified in the TCGA-COAD cohort, and they were significantly associated with functions and pathways related to cancer cell metastasis, proliferation, and apoptosis. We constructed EMTRGs prognostic signature with COMP, MYL9, PCOLCE2, SCG2, and TIMP1 as new COAD prognostic biomarkers. The high-risk group had a poorer prognosis with enhanced immune cell infiltration. The GSEA demonstrated that the high-risk group was involved in "ECM Receptor Interaction," "WNT Signaling Pathway" and "Colorectal Cancer." Furthermore, patients with high risk scores may respond to anti-CTLA4 therapy and may be more resistant to targeted therapy agents BI 2536 and ABT-888. Conclusion: Together, we developed a new EMTRGs prognostic signature that can be an independent prognostic factor for COAD. This study has guiding implications for individualized counseling and treatment of COAD patients.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China.,Yunnan Digestive Endoscopy Clinical Medical Center, Kunming, China
| | - Yan Li
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China.,Yunnan Digestive Endoscopy Clinical Medical Center, Kunming, China
| | - Zan Zuo
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China.,Yunnan Digestive Endoscopy Clinical Medical Center, Kunming, China
| | - Ting Li
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China.,Yunnan Digestive Endoscopy Clinical Medical Center, Kunming, China
| | - Ying An
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China.,Yunnan Digestive Endoscopy Clinical Medical Center, Kunming, China
| | - Wenjing Zhang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.,Department of Medical Oncology, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
21
|
Edwards KJ, Chang B, Babazada H, Lohith K, Park DH, Farwell MD, Sellmyer MA. Using CD69 PET Imaging to Monitor Immunotherapy-Induced Immune Activation. Cancer Immunol Res 2022; 10:1084-1094. [PMID: 35862229 PMCID: PMC10026840 DOI: 10.1158/2326-6066.cir-21-0874] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 04/13/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022]
Abstract
ABSTRACT Immune checkpoint inhibitors (ICI) have been effective in treating a subset of refractory solid tumors, but only a small percentage of treated patients benefit from these therapies. Thus, there is a clinical need for reliable tools that allow for the early assessment of response to ICIs, as well as a preclinical need for imaging tools that aid in the future development and understanding of immunotherapies. Here we demonstrate that CD69, a canonical early-activation marker expressed on a variety of activated immune cells, including cytotoxic T cells and natural killer (NK) cells, is a promising biomarker for the early assessment of response to immunotherapies. We have developed a PET probe by radiolabeling a highly specific CD69 mAb, H1.2F3, with Zirconium-89 (89Zr), [89Zr]-deferoxamine (DFO)-H1.2F3. [89Zr]-DFO-H1.2F3 detected changes in CD69 expression on primary mouse T cells in vitro and detected activated immune cells in a syngeneic tumor immunotherapy model. In vitro uptake studies with [89Zr]-DFO-H1.2F3 showed a 15-fold increase in CD69 expression for activated primary mouse T cells, relative to untreated resting T cells. In vivo PET imaging showed that tumors of ICI-responsive mice had greater uptake than the tumors of nonresponsive and untreated mice. Ex vivo biodistribution, autoradiography, and IHC analyses supported the PET imaging findings. These data suggest that the CD69 PET imaging approach detects CD69 expression with sufficient sensitivity to quantify immune cell activation in a syngeneic mouse immunotherapy model and could allow for the prediction of therapeutic immune responses to novel immunotherapies.
Collapse
Affiliation(s)
- Kimberly J Edwards
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bryan Chang
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hasan Babazada
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Katheryn Lohith
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniel H Park
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael D Farwell
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mark A Sellmyer
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia
| |
Collapse
|
22
|
Wei H, Xie A, Li J, Fang C, Liu L, Xing J, Shi F, Mo F, Chen D, Xie H, Yang Q, Pan X, Tang X, Huang J. PD-1+ CD4 T cell immune response is mediated by HIF-1α/NFATc1 pathway after P. yoelii infection. Front Immunol 2022; 13:942862. [PMID: 36091043 PMCID: PMC9449323 DOI: 10.3389/fimmu.2022.942862] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/05/2022] [Indexed: 11/16/2022] Open
Abstract
The morbidity and mortality of malaria are still high. Programmed cell death-1(PD-1) is an important co-inhibitory factor and CD8 T cells with PD-1 were reported to be exhausted cells. It remains unknown what the role of CD4 T cells expressing PD-1 is and what the upstream regulating molecules of PD-1 in CD4 T cells are. The C57BL/6 mice were injected with Plasmodium yoelii (P. yoelii) in this study. Expressions of PD-1, activation markers, and cytokines were tested. The differentially expressed genes between PD-1+/- CD4 T cells were detected by microarray sequencing. Western blot, chromatin immunoprecipitation (ChIP), siRNA, hypoxia inducible factor-1α (HIF-1α) inducer and inhibitor were used to explore PD-1’s upstream molecules, respectively. The proportions of PD-1+ CD4 T cells increased post P. yoelii infection. PD-1+ CD4 T cells expressed more activated surface markers and could produce more cytokines. Nuclear factor of activated T cells 1 (NFATc1) was found to be a key transcription factor to induce PD-1 expression after infection. Both the inducer and the inhibitor of HIF-1α could change the expressions of NFATc1 and PD-1 in vivo and in vitro, respectively. Taken together, P. yoelii infection induced NFATc1 expression by HIF-1α. The highly expressed NFATc1 entered the nucleus and initiated PD-1 expression. PD-1+ CD4 T cells appeared to be more activated and could secrete more cytokines to regulate the host’s immune responses against malaria.
Collapse
Affiliation(s)
- Haixia Wei
- Department of Infectious Diseases, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Basic Medical Science, China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Anqi Xie
- Department of Basic Medical Science, China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Jiajie Li
- Department of Basic Medical Science, China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Chao Fang
- Department of Basic Medical Science, China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Lin Liu
- Department of Basic Medical Science, China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Junmin Xing
- Department of Basic Medical Science, China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Feihu Shi
- Department of Infectious Diseases, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Feng Mo
- Department of Infectious Diseases, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dianhui Chen
- Department of Infectious Diseases, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Basic Medical Science, China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Hongyan Xie
- Department of Basic Medical Science, China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Quan Yang
- Department of Basic Medical Science, China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Xingfei Pan
- Department of Infectious Diseases, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Xingfei Pan, ; Xiaoping Tang, ; Jun Huang,
| | - Xiaoping Tang
- Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Xingfei Pan, ; Xiaoping Tang, ; Jun Huang,
| | - Jun Huang
- Department of Basic Medical Science, China Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Xingfei Pan, ; Xiaoping Tang, ; Jun Huang,
| |
Collapse
|
23
|
Hu ZW, Sun W, Wen YH, Ma RQ, Chen L, Chen WQ, Lei WB, Wen WP. CD69 and SBK1 as potential predictors of responses to PD-1/PD-L1 blockade cancer immunotherapy in lung cancer and melanoma. Front Immunol 2022; 13:952059. [PMID: 36045683 PMCID: PMC9421049 DOI: 10.3389/fimmu.2022.952059] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundPD-1/PD-L1 blockade is a promising immunotherapeutic strategy with the potential to improve the outcomes of various cancers. However, there is a critically unmet need for effective biomarkers of response to PD-1/PD-L1 blockade.Materials and methodsPotential biomarkers of response to PD-1/PD-L1 blockade were obtained from the Cancer Treatment Response gene signature Database (CTR-DB). A comprehensive pan-cancer analysis was done on The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) datasets. Correlations between gene expression and infiltration by immune cells were assessed using TIMER, EPIC, MCPcounter, xCell, CIBERSORT, and quanTIseq. Immunophenoscore (IPS) was used to assess the potential application of the biomarkers to all TCGA tumors.ResultsAnalysis of CTR-DB data identified CD69 and SBK1 as potential biomarkers of response to PD-1/PD-L1 blockade. Correlation analysis revealed that in various TCGA cancer datasets, CD69 expression level correlated positively with most immune checkpoints and tumor-infiltrating immune cells, while SBK1 expression level correlated negatively with infiltrating immune cells. IPS analysis demonstrated the ability of CD69 and SBK1 to predict PD-1/PD-L1 blockade responses in various cancers.ConclusionCD69 and SBK1 are potential predictors of response to cancer immunotherapy using PD-1/PD-L1 blockade. These biomarkers may guide treatment decisions, leading to precise treatment and minimizing the waste of medical resources.
Collapse
Affiliation(s)
- Zhang-Wei Hu
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
| | - Wei Sun
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
| | - Yi-Hui Wen
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
| | - Ren-Qiang Ma
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
| | - Lin Chen
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
| | - Wen-Qing Chen
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
| | - Wen-Bin Lei
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Wei-Ping Wen, ; Wen-Bin Lei,
| | - Wei-Ping Wen
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
- Department of Otolaryngology, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Wei-Ping Wen, ; Wen-Bin Lei,
| |
Collapse
|
24
|
Clinical and Histological Effects of Partial Blood Flow Impairment in Vascularized Lymph Node Transfer. J Clin Med 2022; 11:jcm11144052. [PMID: 35887816 PMCID: PMC9322400 DOI: 10.3390/jcm11144052] [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: 06/14/2022] [Revised: 06/28/2022] [Accepted: 07/10/2022] [Indexed: 11/28/2022] Open
Abstract
Regarding vascularized lymph node transfer (VLNT) for lymphedema, partial blood flow impairment in transferred lymph node (LN) flaps may adversely affect the therapeutic results. We investigated the clinical and histological effects of partial blood flow impairment in LN flaps. In upper extremity lymphedema cases, based on ultrasonographic examination at 2 weeks after VLNT, we compared the treatment results depending on whether the postoperative blood flow in transferred LNs was good (Group G) or poor (Group P). Novel partial ischemia and congestion of LN flap mouse models were developed to determine their histological features. In 42 cases, significant differences were observed between Group G (n = 37) and Group P (n = 5) based on the amount of volume reduction (136.7 ± 91.7 mL and 55.4 ± 60.4 mL, respectively; p = 0.04) and lymph flow recanalization rate in indocyanine green fluorescent lymphography (67.6% and 0%, respectively; p = 0.0007). In mouse models, thrombi formation in the marginal sinus and numerous Myl9/12-positive immunocompetent cells in follicles were observed in congested LNs. Blood flow maintenance in the transferred LNs is an essential factor influencing the therapeutic effect of VLNT. Postoperatively, surgeons should closely monitor blood flow in the transferred LNs, particularly in cases of congestion.
Collapse
|
25
|
Koyaman-Nasu R, Wang Y, Hasegawa I, Endo Y, Nakayama T, Kimura MY. The cellular and molecular basis of CD69 function in anti-tumor immunity. Int Immunol 2022; 34:555-561. [PMID: 35689672 DOI: 10.1093/intimm/dxac024] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/09/2022] [Indexed: 12/16/2022] Open
Abstract
Cancer immunotherapy utilizes our immune system to attack cancer cells and is an extremely promising strategy for cancer treatment. Although immune-checkpoint blockade, such as anti-PD-1 antibody (Ab), has demonstrated significant enhancement of anti-tumor immunity and has induced notable clinical outcomes, its response rates remain low, and adverse effects are always a matter of concern; therefore, new targets for cancer immunotherapy are always desired. In this situation, new concepts are needed to fuel the investigation of new target molecules for cancer immunotherapy. We propose that CD69 is one such target molecule. CD69 is known to be an activation marker of leukocytes and is also considered a crucial regulator of various immune responses through its interacting proteins. CD69 promotes T cell retention in lymphoid tissues via sphingosine-1-phosphate receptor 1 (S1P1) internalization and also plays roles in the pathogenesis of inflammatory disorders through interacting with its functional ligands Myl9/12 (myosin light chains 9, 12a and 12b). In anti-tumor immunity, CD69 is known to be expressed on T cells in the tumor microenvironment (TME) and tumor-draining lymph nodes (TDLNs). We revealed that CD69 negatively regulates the effector function of intratumoral T cells and importantly controls the 'exhaustion' of CD8 T cells. In addition, we and others showed that either CD69 deficiency or the administration of anti-CD69 monoclonal antibody enhances anti-tumor immunity. Thus, CD69 is an attractive target for cancer immunotherapy.
Collapse
Affiliation(s)
- Ryo Koyaman-Nasu
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Yangsong Wang
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Ichita Hasegawa
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Yukihiro Endo
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan.,AMED-CREST, AMED, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Motoko Y Kimura
- Department of Experimental Immunology, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| |
Collapse
|
26
|
Extracellular Vesicles—A New Potential Player in the Immunology of Renal Cell Carcinoma. J Pers Med 2022; 12:jpm12050772. [PMID: 35629194 PMCID: PMC9144962 DOI: 10.3390/jpm12050772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/20/2022] [Accepted: 04/28/2022] [Indexed: 02/08/2023] Open
Abstract
The incidence of renal cell carcinoma (RCC) has doubled in the developed world within the last fifty years, and now it is responsible for 2–3% of diagnosed cancers. The delay in diagnosis and the not fully understood pathogenesis are the main challenges that have to be overcome. It seems that extracellular vesicles (EVs) are one of the key players in tumor development since they ensure a proper microenvironment for the tumor cells. The stimulation of angiogenesis and immunosuppression is mediated by molecules contained in EVs. It was shown that EVs derived from cancer cells can inhibit T cell proliferation, natural killer lymphocyte activation, and dendritic cell maturation by this mechanism. Moreover, EVs may be a biomarker for the response to anti-cancer treatment. In this review, we sum up the knowledge about the role of EVs in RCC pathogenesis and show their future perspectives in this field.
Collapse
|
27
|
Gu K, Wang T, Peng L, Zhao Y. FIP-fve Stimulates Cell Proliferation and Enhances IL-2 Release by Activating MAP2K3/p38α (MAPK14) Signaling Pathway in Jurkat E6-1 Cells. Front Nutr 2022; 9:881924. [PMID: 35614983 PMCID: PMC9125247 DOI: 10.3389/fnut.2022.881924] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/28/2022] [Indexed: 11/30/2022] Open
Abstract
FIP-fve, a fungal fruiting body protein from Flammulina velutipes, has potential immunomodulatory properties. Here, we investigated the immunomodulation mechanism of FIP-fve in Jurkat E6-1 cells by conducting a cell viability assay and IL-2 release assay. Kinase inhibitors experiment and proteomics analysis were also involved in the mechanism study. It was found that FIP-fve stimulated cell proliferation and enhanced IL-2 secretion in a dose-dependent manner in Jurkat E6-1 cells. Unbiased high-throughput proteomics analysis showed that 4 T cell immune activation markers, including ZAP-70, CD69, CD82, and KIF23, were upregulated in response to FIP-fve treatment. Further pathway analysis indicated that MAP2K3/p38 pathway-related proteins, including MAP2K, p38, ELK, AATF, FOS, and JUN-B, were unregulated. In addition, losmapimod (p38 inhibitor) and gossypetin (MAP2K3 inhibitor) inhibited FIP-fve enhanced cell proliferation and IL-2 release in Jurkat E6-1 cells. Our results demonstrate that FIP-fve stimulates cell proliferation and enhances IL-2 secretion through MAP2K3/p38α activation.
Collapse
Affiliation(s)
- Kefei Gu
- Institute for Agri-Food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, Shanghai, China
| | - Tan Wang
- Institute for Agri-Food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, Shanghai, China
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
- Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture, Shanghai, China
| | - Liying Peng
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Science, Shanghai, China
| | - Yueliang Zhao
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
- Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture, Shanghai, China
- *Correspondence: Yueliang Zhao, ;
| |
Collapse
|
28
|
Zhang W, Zhang Y, Li X, Cao Z, Mo Q, Sheng R, Ling C, Chi J, Yao Q, Chen J, Wang H. Multifunctional polyphenol-based silk hydrogel alleviates oxidative stress and enhances endogenous regeneration of osteochondral defects. Mater Today Bio 2022; 14:100251. [PMID: 35469254 PMCID: PMC9034395 DOI: 10.1016/j.mtbio.2022.100251] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/08/2022] [Accepted: 03/31/2022] [Indexed: 01/25/2023] Open
Abstract
In osteochondral defects, oxidative stress caused by elevated levels of reactive oxygen species (ROS) can disrupt the normal endogenous repair process. In this study, a multifunctional hydrogel composed of silk fibroin (SF) and tannic acid (TA), the FDA-approved ingredients, was developed to alleviate oxidative stress and enhance osteochondral regeneration. In this proposed hydrogel, SF first interacts with TA to form a hydrogen-bonded supramolecular structure, which is subsequently enzymatically crosslinked to form a stable hydrogel. Furthermore, TA had multiple phenolic hydroxyl groups that formed interactions with the therapeutic molecule E7 peptide for controlled drug delivery. In vitro investigations showed that SF-TA and SF-TA-E7 hydrogels exhibited a multitude of biological effects including scavenging of ROS, maintaining cell viability, and promoting the proliferation of bone marrow mesenchymal stem cells (BMSCs) against oxidative stress. The proteomic analysis indicated that SF-TA and SF-TA-E7 hydrogels suppressed oxidative stress, which in turn improved cell proliferation in multiple proliferation and apoptosis-related pathways. In rabbit osteochondral defect model, SF-TA and SF-TA-E7 hydrogels promoted enhanced regeneration of both cartilage and subchondral bone as compared to hydrogel without TA incorporation. These findings indicated that the multifunctional SF-TA hydrogel provided a microenvironment suitable for the endogenous regeneration of osteochondral defects.
Collapse
Affiliation(s)
- Wei Zhang
- School of Medicine, Southeast University, 210009, Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
| | - Yanan Zhang
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Xiaolong Li
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Zhicheng Cao
- School of Medicine, Southeast University, 210009, Nanjing, China
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
| | - Qingyun Mo
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Renwang Sheng
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Chen Ling
- School of Medicine, Southeast University, 210009, Nanjing, China
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
| | - Jiayu Chi
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
| | - Jialin Chen
- School of Medicine, Southeast University, 210009, Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
| | - Hongmei Wang
- School of Medicine, Southeast University, 210009, Nanjing, China
- Department of Pharmaceutical Sciences, Binzhou Medical University, 264003, Yantai, Shandong, China
| |
Collapse
|
29
|
Lv M, Luo L, Chen X. The landscape of prognostic and immunological role of myosin light chain 9 (MYL9) in human tumors. IMMUNITY INFLAMMATION AND DISEASE 2021; 10:241-254. [PMID: 34729929 PMCID: PMC8767521 DOI: 10.1002/iid3.557] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/09/2021] [Accepted: 10/20/2021] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Recent studies have shown that myosin light chain 9 (MYL9) plays a vital role in immune infiltration, tumor invasion, and metastasis; however, the prognostic and immunological role of MYL9 has not been reported. The purpose of this study was to explore the potential prognostic and immunological roles of MYL9 in human cancers by public datasets mainly including the cancer genome atlas (TCGA) and Gene expression omnibus. METHODS The expression pattern and prognostic value of MYL9 were analyzed across multiple public datasets in different cancer. The correlations between MYL9 expression and immune infiltration among multiple cancers were analyzed by using the TIMER2.0. The MYL9-related gene enrichment analysis was implemented by mainly using KEGG and GO datasets. RESULTS MYL9 was lowly expressed in most cancers, such as breast cancer, lung adenocarcinoma and squamous cell carcinoma, and stomach adenocarcinoma; but it was highly expressed in several cancers, such as cholangiocarcinoma, head and neck squamous cell carcinoma, and liver hepatocellular carcinoma. Furthermore, MYL9 expression was distinctively associated with prognosis in adrenocortical carcinoma, colon adenocarcinoma, brain glioma, lung cancer, ovarian cancer, gastric cancer, breast cancer, blood cancer, and prostate cancer patients. The expressions of MYL9 were significantly associated with the infiltration of cancer-associated fibroblasts, B cell, CD8+ T cell, CD4+ T cell, macrophage, neutrophil, dendritic cell in different tumors as well as immune markers. In addition, we found that the functional mechanisms of MYL9 involved muscle contraction and focal adhesion. CONCLUSION MYL9 can serve as a prognostic signature in pan-cancer and is associated with immune infiltration. This pan-cancer study is the first to show a relatively comprehensive understanding of the prognostic and immunological roles of MYL9 across different cancers.
Collapse
Affiliation(s)
- Minghe Lv
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lumeng Luo
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xue Chen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Gao P, Ren G. Identification of potential target genes of non-small cell lung cancer in response to resveratrol treatment by bioinformatics analysis. Aging (Albany NY) 2021; 13:23245-23261. [PMID: 34633989 PMCID: PMC8544309 DOI: 10.18632/aging.203616] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/28/2021] [Indexed: 12/24/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the most common type in lung cancer in the world, and it severely threatens the life of patients. Resveratrol has been reported to inhibit cancer. However, mechanisms of resveratrol inhibiting NSCLC were unclear. The aim of this study was to identify differentially expressed genes (DEGs) of NSCLC treated with resveratrol and reveal the potential targets of resveratrol in NSCLC. We obtained mRNA expression profiles of two datasets from the National Center for Biotechnology Information Gene Expression Omnibus (NCBI-GEO) and 271 DEGs were selected for further analysis. Data from STRING shown that 177 nodes and 342 edges were in the protein-protein interaction (PPI) network, and 10 hub genes (ANPEP, CD69, ITGAL, PECAM1, PTPRC, CD34, ITGA1, CCL2, SOX2, and EGFR) were identified by Cytoscape plus-in cytoHubba. Survival analysis revealed that NSCLC patients showing low expression of PECAM1, ANPEP, CD69, ITGAL, and PTPRC were associated with worse overall survival (OS) (P < 0.05), and high expression of SOX2 and EGFR was associated with worse OS for NSCLC patients (P < 0.05). Overall, we identified ANPEP, CD69, ITGAL, and PTPRC as potential candidate genes which were main effects of resveratrol on the treatment of NSCLC. ANPEP, ITGAL, CD69, and PTPRC are all clusters of differentiation (CD) antigens, might be the targets of resveratrol. The bioinformatic results suggested that the inhibitory effect of resveratrol on lung cancer may be related to the immune signaling pathway. Further studies are needed to validate these findings and to explore their functional mechanisms.
Collapse
Affiliation(s)
- Peng Gao
- Institute of Microvascular Medicine, Medical Research Center, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Guanghui Ren
- Shandong Provincial Key Laboratory of Animal Resistant, School of Life Sciences, Shandong Normal University, Jinan, Shandong 250014, China
| |
Collapse
|
31
|
Xie S, Wei H, Peng A, Xie A, Li J, Fang C, Shi F, Yang Q, Huang H, Xie H, Pan X, Tian X, Huang J. Ikzf2 Regulates the Development of ICOS + Th Cells to Mediate Immune Response in the Spleen of S. japonicum-Infected C57BL/6 Mice. Front Immunol 2021; 12:687919. [PMID: 34475870 PMCID: PMC8406689 DOI: 10.3389/fimmu.2021.687919] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/26/2021] [Indexed: 11/29/2022] Open
Abstract
Background Th cells (helper T cells) have multiple functions in Schistosoma japonicum (S. japonicum) infection. Inducible co-stimulator (ICOS) is induced and expressed in activated T lymphocytes, which enhances the development of B cells and antibody production through the ICOS/ICOSL pathway. It remains unclear about the role and possible regulating mechanism of ICOS+ Th cells in the spleen of S. japonicum-infected C57BL/6 mice. Methods C57BL/6 mice were infected with cercariae of S. japonicum through the abdomen. The expression of ICOS, activation markers, and the cytokine production on CD4+ ICOS+ Th cells were detected by flow cytometry (FCM) and quantitative real-time PCR (qRT-PCR). Moreover, the differentially expressed gene data of ICOS+ and ICOS- Th cells from the spleen of infected mice were obtained by mRNA sequencing. Besides, Western blot and chromatin immunoprecipitation (ChIP) were used to explore the role of Ikzf2 on ICOS expression. Results After S. japonicum infection, the expression of ICOS molecules gradually increased in splenic lymphocytes, especially in Th cells (P < 0.01). Compared with ICOS- Th cells, more ICOS+ Th cells expressed CD69, CD25, CXCR5, and CD40L (P < 0.05), while less of them expressed CD62L (P < 0.05). Also, ICOS+ Th cells expressed more cytokines, such as IFN-γ, IL-4, IL-10, IL-2, and IL-21 (P < 0.05). RNA sequencing results showed that many transcription factors were increased significantly in ICOS+ Th cells, especially Ikzf2 (P < 0.05). And then, the expression of Ikzf2 was verified to be significantly increased and mainly located in the nuclear of ICOS+ Th cells. Finally, ChIP experiments and dual-luciferase reporter assay confirmed that Ikzf2 could directly bind to the ICOS promoter in Th cells. Conclusion In this study, ICOS+ Th cells were found to play an important role in S. japonicum infection to induce immune response in the spleen of C57BL/6 mice. Additionally, Ikzf2 was found to be one important transcription factor that could regulate the expression of ICOS in the spleen of S. japonicum-infected C57BL/6 mice.
Collapse
Affiliation(s)
- Shihao Xie
- Department of Infectious Diseases, Key Laboratory for Major Obsteric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Haixia Wei
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Anping Peng
- Biological Resource Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Anqi Xie
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiajie Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chao Fang
- Department of Infectious Diseases, Key Laboratory for Major Obsteric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Feihu Shi
- Department of Infectious Diseases, Key Laboratory for Major Obsteric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Quan Yang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - He Huang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hongyan Xie
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xingfei Pan
- Department of Infectious Diseases, Key Laboratory for Major Obsteric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xu Tian
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- College of Pharmacy, Guangzhou Medical University, Guangzhou, China
| | - Jun Huang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
32
|
Astaxanthin Protects Dendritic Cells from Lipopolysaccharide-Induced Immune Dysfunction. Mar Drugs 2021; 19:md19060346. [PMID: 34204220 PMCID: PMC8235365 DOI: 10.3390/md19060346] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/13/2021] [Accepted: 06/15/2021] [Indexed: 12/29/2022] Open
Abstract
Astaxanthin, originating from seafood, is a naturally occurring red carotenoid pigment. Previous studies have focused on its antioxidant properties; however, whether astaxanthin possesses a desired anti-inflammatory characteristic to regulate the dendritic cells (DCs) for sepsis therapy remains unknown. Here, we explored the effects of astaxanthin on the immune functions of murine DCs. Our results showed that astaxanthin reduced the expressions of LPS-induced inflammatory cytokines (TNF-α, IL-6, and IL-10) and phenotypic markers (MHCII, CD40, CD80, and CD86) by DCs. Moreover, astaxanthin promoted the endocytosis levels in LPS-treated DCs, and hindered the LPS-induced migration of DCs via downregulating CCR7 expression, and then abrogated allogeneic T cell proliferation. Furthermore, we found that astaxanthin inhibited the immune dysfunction of DCs induced by LPS via the activation of the HO-1/Nrf2 axis. Finally, astaxanthin with oral administration remarkably enhanced the survival rate of LPS-challenged mice. These data showed a new approach of astaxanthin for potential sepsis treatment through avoiding the immune dysfunction of DCs.
Collapse
|
33
|
Wang J, Xiang Y, Jiang S, Li H, Caviezel F, Katawatin S, Duangjinda M. Involvement of the VEGF signaling pathway in immunosuppression and hypoxia stress: analysis of mRNA expression in lymphocytes mediating panting in Jersey cattle under heat stress. BMC Vet Res 2021; 17:209. [PMID: 34098948 PMCID: PMC8186226 DOI: 10.1186/s12917-021-02912-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 05/20/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Extreme panting under heat stress threatens dairy cattle milk production. Previous research has revealed that the gas exchange-mediated respiratory drive in critically ill dairy cattle with low O2 saturation induces panting. Vascular endothelial growth factor (VEGF) signaling may play important roles in immunosuppression and oxidative stress during severe respiratory stress responses in heat-stressed cattle. The objectives of this study were to transcriptomically analyze mRNA expression mediating heat-induced respiratory stress-associated panting, evaluate gas exchange, screen hub genes, and verify the expression of proteins encoded by differentially expressed genes in lymphocyte pathways. RESULTS Jersey cattle were naturally heat-exposed. Physiological data were collected for response evaluation, and blood was collected for gas exchange and gene expression assays at 06:00, 10:00 and 14:00 continuously for 1 week. Lymphocytes were isolated from whole-blood samples for mRNA-seq and expression analysis of key pathway genes/proteins. The cattle respiration rates differed with time, averaging 51 bpm at 06:00, 76 bpm at 10:00, and 121 bpm at 14:00 (p < 0.05). Gas exchange analysis showed that both pH and pCO2 differed with time: they were 7.41 and 41 mmHg at 06:00, 7.45 and 37.5 mmHg at 10:00, and 7.49 and 33 mmHg at 14:00, respectively (p < 0.01). Sixteen heat-related differentially expressed genes (DEGs; 13 upregulated and 3 downregulated) were screened between 212 DEGs and 1370 heat stress-affected genes. Kyoto Encyclopedia of Genes and Genomes (KEGG) hub gene functional analysis annotated eleven genes to signal transduction, six genes to the immune response, and five genes to the endocrine response, including both prostaglandin-endoperoxide synthase 2 (PTGS2) and VEGF. Gene Ontology (GO) functional enrichment analysis revealed that oxygen regulation was associated with the phosphorus metabolic process, response to oxygen levels, response to decreased oxygen levels, response to hypoxia and cytokine activity terms. The main signaling pathways were the VEGF, hypoxia inducible factor-1(HIF-1), cytokine-cytokine receptor interaction and TNF pathways. Four genes involved Integrin beta 3 (ITBG3), PTGS2, VEGF, and myosin light chain 9 (MYL9) among the 16 genes related to immunosuppression, oxidative stress, and endocrine dysfunction were identified as participants in the VEGF signaling pathway and oxygenation. CONCLUSION These findings help elucidate the underlying immune and oxygen regulation mechanisms associated with the VEGF signaling pathway in heat-stressed dairy cattle.
Collapse
Affiliation(s)
- Jian Wang
- Faculty of Veterinary Medicine, Southwest University, Chongqing, 400700, China.
| | - Yang Xiang
- Faculty of Veterinary Medicine, Southwest University, Chongqing, 400700, China
| | - Shisong Jiang
- Department of Oncology, Oxford University, Oxford, OX3 7DQ, UK
| | - Hongchang Li
- Faculty of Veterinary Medicine, Southwest University, Chongqing, 400700, China
| | - Flurin Caviezel
- Department of Oncology, Oxford University, Oxford, OX3 7DQ, UK
| | - Suporn Katawatin
- Department of Animal Science, Khon Kaen University, Kaen, 40002, Thailand
| | - Monchai Duangjinda
- Department of Animal Science, Khon Kaen University, Kaen, 40002, Thailand
| |
Collapse
|
34
|
Yokoyama M, Kimura MY, Ito T, Hayashizaki K, Endo Y, Wang Y, Yagi R, Nakagawa T, Kato N, Matsubara H, Nakayama T. Myosin Light Chain 9/12 Regulates the Pathogenesis of Inflammatory Bowel Disease. Front Immunol 2021; 11:594297. [PMID: 33584659 PMCID: PMC7878395 DOI: 10.3389/fimmu.2020.594297] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/15/2020] [Indexed: 12/30/2022] Open
Abstract
The numbers of patients with inflammatory bowel disease (IBD), such as ulcerative colitis (UC) and Crohn’s disease (CD), have been increasing over time, worldwide; however, the pathogenesis of IBD is multifactorial and has not been fully understood. Myosin light chain 9 and 12a and 12b (Myl9/12) are known as ligands of the CD69 molecule. They create “Myl9 nets” that are often detected in inflamed site, which play a crucial role in regulating the recruitment and retention of CD69-expressing effector cells in inflamed tissues. We demonstrated the strong expression of Myl9/12 in the inflamed gut of IBD patients and mice with DSS-induced colitis. The administration of anti-Myl9/12 Ab to mice with DSS-induced colitis ameliorated the inflammation and prolonged their survival. The plasma Myl9 levels in the patients with active UC and CD were significantly higher than those in patients with disease remission, and may depict the disease severity of IBD patients, especially those with UC. Thus, our results indicate that Myl9/12 are involved in the pathogenesis of IBD, and are likely to be a new therapeutic target for patients suffering from IBD.
Collapse
Affiliation(s)
- Masaya Yokoyama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.,Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Motoko Y Kimura
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshihiro Ito
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Koji Hayashizaki
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yukihiro Endo
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yangsong Wang
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ryoji Yagi
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tomoo Nakagawa
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Naoya Kato
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.,Advanced Research and Development Programs for Medical Innovation (AMED-CREST), Japan Agency for Medical Research and Development (AMED), Chiba, Japan
| |
Collapse
|
35
|
Gómez-Mendoza DP, Marques FD, Melo-Braga MN, Sprenger RR, Sinisterra RD, Kjeldsen F, Santos RA, Verano-Braga T. Angiotensin-(1-7) oral treatment after experimental myocardial infarction leads to downregulation of CXCR4. J Proteomics 2019; 208:103486. [DOI: 10.1016/j.jprot.2019.103486] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/05/2019] [Accepted: 08/10/2019] [Indexed: 11/27/2022]
|
36
|
Müller LME, Holmes M, Michael JL, Scott GB, West EJ, Scott KJ, Parrish C, Hall K, Stäble S, Jennings VA, Cullen M, McConnell S, Langton C, Tidswell EL, Shafren D, Samson A, Harrington KJ, Pandha H, Ralph C, Kelly RJ, Cook G, Melcher AA, Errington-Mais F. Plasmacytoid dendritic cells orchestrate innate and adaptive anti-tumor immunity induced by oncolytic coxsackievirus A21. J Immunother Cancer 2019; 7:164. [PMID: 31262361 PMCID: PMC6604201 DOI: 10.1186/s40425-019-0632-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 06/06/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The oncolytic virus, coxsackievirus A21 (CVA21), has shown promise as a single agent in several clinical trials and is now being tested in combination with immune checkpoint blockade. Combination therapies offer the best chance of disease control; however, the design of successful combination strategies requires a deeper understanding of the mechanisms underpinning CVA21 efficacy, in particular, the role of CVA21 anti-tumor immunity. Therefore, this study aimed to examine the ability of CVA21 to induce human anti-tumor immunity, and identify the cellular mechanism responsible. METHODS This study utilized peripheral blood mononuclear cells from i) healthy donors, ii) Acute Myeloid Leukemia (AML) patients, and iii) patients taking part in the STORM clinical trial, who received intravenous CVA21; patients receiving intravenous CVA21 were consented separately in accordance with local institutional ethics review and approval. Collectively, these blood samples were used to characterize the development of innate and adaptive anti-tumor immune responses following CVA21 treatment. RESULTS An Initial characterization of peripheral blood mononuclear cells, collected from cancer patients following intravenous infusion of CVA21, confirmed that CVA21 activated immune effector cells in patients. Next, using hematological disease models which were sensitive (Multiple Myeloma; MM) or resistant (AML) to CVA21-direct oncolysis, we demonstrated that CVA21 stimulated potent anti-tumor immune responses, including: 1) cytokine-mediated bystander killing; 2) enhanced natural killer cell-mediated cellular cytotoxicity; and 3) priming of tumor-specific cytotoxic T lymphocytes, with specificity towards known tumor-associated antigens. Importantly, immune-mediated killing of both MM and AML, despite AML cells being resistant to CVA21-direct oncolysis, was observed. Upon further examination of the cellular mechanisms responsible for CVA21-induced anti-tumor immunity we have identified the importance of type I IFN for NK cell activation, and demonstrated that both ICAM-1 and plasmacytoid dendritic cells were key mediators of this response. CONCLUSION This work supports the development of CVA21 as an immunotherapeutic agent for the treatment of both AML and MM. Additionally, the data presented provides an important insight into the mechanisms of CVA21-mediated immunotherapy to aid the development of clinical biomarkers to predict response and rationalize future drug combinations.
Collapse
Affiliation(s)
- Louise M. E. Müller
- Section of Infection and Immunity, Leeds Institute of Medical Research (LIMR), University of Leeds, St. James’s University Hospital, Level 5, Wellcome Trust Brenner Building (WTBB), Leeds, LS9 7TF UK
| | - Matthew Holmes
- Section of Infection and Immunity, Leeds Institute of Medical Research (LIMR), University of Leeds, St. James’s University Hospital, Level 5, Wellcome Trust Brenner Building (WTBB), Leeds, LS9 7TF UK
| | - Joanne L. Michael
- Section of Infection and Immunity, Leeds Institute of Medical Research (LIMR), University of Leeds, St. James’s University Hospital, Level 5, Wellcome Trust Brenner Building (WTBB), Leeds, LS9 7TF UK
| | - Gina B. Scott
- Section of Infection and Immunity, Leeds Institute of Medical Research (LIMR), University of Leeds, St. James’s University Hospital, Level 5, Wellcome Trust Brenner Building (WTBB), Leeds, LS9 7TF UK
| | - Emma J. West
- Section of Infection and Immunity, Leeds Institute of Medical Research (LIMR), University of Leeds, St. James’s University Hospital, Level 5, Wellcome Trust Brenner Building (WTBB), Leeds, LS9 7TF UK
| | - Karen J. Scott
- Section of Infection and Immunity, Leeds Institute of Medical Research (LIMR), University of Leeds, St. James’s University Hospital, Level 5, Wellcome Trust Brenner Building (WTBB), Leeds, LS9 7TF UK
| | | | - Kathryn Hall
- Section of Infection and Immunity, Leeds Institute of Medical Research (LIMR), University of Leeds, St. James’s University Hospital, Level 5, Wellcome Trust Brenner Building (WTBB), Leeds, LS9 7TF UK
| | - Sina Stäble
- Section of Infection and Immunity, Leeds Institute of Medical Research (LIMR), University of Leeds, St. James’s University Hospital, Level 5, Wellcome Trust Brenner Building (WTBB), Leeds, LS9 7TF UK
| | - Victoria A. Jennings
- Translational Immunotherapy Team, The Institute of Cancer Research and Royal Marsden Hospital/Institute of Cancer Research NIHR Biomedical Research Centre, London, UK
| | - Matthew Cullen
- Haematological Malignancy Diagnostics Service, St. James’s University Hospital, Leeds, UK
| | - Stewart McConnell
- Department of Haematology, St. James’s University Hospital, Leeds, UK
| | - Catherine Langton
- Department of Haematology, St. James’s University Hospital, Leeds, UK
| | - Emma L. Tidswell
- Section of Infection and Immunity, Leeds Institute of Medical Research (LIMR), University of Leeds, St. James’s University Hospital, Level 5, Wellcome Trust Brenner Building (WTBB), Leeds, LS9 7TF UK
| | - Darren Shafren
- School of Biomedical Science and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Adel Samson
- Section of Infection and Immunity, Leeds Institute of Medical Research (LIMR), University of Leeds, St. James’s University Hospital, Level 5, Wellcome Trust Brenner Building (WTBB), Leeds, LS9 7TF UK
| | - Kevin J. Harrington
- Translational Immunotherapy Team, The Institute of Cancer Research and Royal Marsden Hospital/Institute of Cancer Research NIHR Biomedical Research Centre, London, UK
| | - Hardev Pandha
- Surrey Cancer Research Institute, Leggett Building, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Christy Ralph
- Section of Infection and Immunity, Leeds Institute of Medical Research (LIMR), University of Leeds, St. James’s University Hospital, Level 5, Wellcome Trust Brenner Building (WTBB), Leeds, LS9 7TF UK
| | - Richard J. Kelly
- Department of Haematology, St. James’s University Hospital, Leeds, UK
| | - Gordon Cook
- Section of Experimental Haematology, LIMR, University of Leeds, St. James’s University Hospital, Leeds, UK
| | - Alan A. Melcher
- Translational Immunotherapy Team, The Institute of Cancer Research and Royal Marsden Hospital/Institute of Cancer Research NIHR Biomedical Research Centre, London, UK
| | - Fiona Errington-Mais
- Section of Infection and Immunity, Leeds Institute of Medical Research (LIMR), University of Leeds, St. James’s University Hospital, Level 5, Wellcome Trust Brenner Building (WTBB), Leeds, LS9 7TF UK
| |
Collapse
|
37
|
Hirahara K. Pathogenicity of acquired immunity in human diseases. Semin Immunopathol 2019; 41:279-281. [PMID: 31065764 DOI: 10.1007/s00281-019-00739-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 03/18/2019] [Indexed: 10/26/2022]
Affiliation(s)
- Kiyoshi Hirahara
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan. .,AMED-PRIME, AMED, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.
| |
Collapse
|