1
|
Meriç N, Erkan PÇ, Kocabaş F. Deciphering avian hematopoietic stem cells by surface marker screening and gene expression profiling. Mol Immunol 2024; 175:20-30. [PMID: 39288684 DOI: 10.1016/j.molimm.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/29/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Avian species have played a pivotal role in developmental hematopoiesis research, leading to numerous critical discoveries. Avian influenza, particularly the H5N1 strain, poses a significant threat to poultry and has zoonotic potential for humans. Infections often result in abnormal hematologic profiles, highlighting the complex interplay between avian diseases and hematopoiesis. Many avian diseases can suppress immune cells in the bone marrow (BM), impacting immune responses. Studying hematopoietic stem cells (HSCs) in avian BM is crucial for understanding these processes and developing effective vaccines and protection strategies for both avian and human health. METHODS This study adapted methods from mouse studies to isolate avian HSCs as Lineage-negative (Lin-) cells. These isolated cells were further identified as Lin-Sca1+c-Kit+ (LSK) and were found to be more prevalent than in control groups. RT-PCR analyses were conducted, showing that genes like MEIS1 and TSC1 were upregulated, while SIRT1, FOXO1, and AHR were downregulated in these stem cells. Screening for LSK markers revealed ten unique surface antigens in the Sca1+c-Kit+ cell populations, including highly enriched antigens such as CD178, CD227, and CD184. Additionally, studies on quail HSCs demonstrated that similar labeling techniques were effective in quail BM. RESULTS The research demonstrated that the identification of avian HSC-specific surface antigens provides valuable insights into the pathogenesis of avian influenza and other diseases, enhancing our understanding of how these diseases suppress HSC function. Notably, the upregulation of MEIS1 and TSC1 genes in LSK cells underscores their critical roles in regulating hematopoietic processes. Conversely, the downregulation of SIRT1, FOXO1, and AHR genes provides important clues about their roles in differentiation and immune response mechanisms. DISCUSSION The findings of this study deepen our understanding of the effects of avian diseases on the immune system by identifying surface markers specific to avian HSCs. The suppression of HSC function by pathogens such as influenza highlights the importance of understanding these cells in developing targeted vaccines. These results represent a significant step towards improving global health security by mitigating risks associated with avian pathogens.
Collapse
Affiliation(s)
- Neslihan Meriç
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, Türkiye; Faculty of Engineering and Natural Sciences, Department of Molecular Biology and Genetics, Kütahya Health Sciences University, Kütahya, Türkiye.
| | - Pınar Çolakoğlu Erkan
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, Türkiye
| | - Fatih Kocabaş
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul, Türkiye.
| |
Collapse
|
2
|
Wang ZH, Zheng X, Rao GW, Zheng Q. Targeted small molecule therapy and inhibitors for lymphoma. Future Med Chem 2024; 16:1465-1484. [PMID: 39016063 PMCID: PMC11352716 DOI: 10.1080/17568919.2024.2359893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 05/21/2024] [Indexed: 07/18/2024] Open
Abstract
Lymphoma, a blood tumor, has become the ninth most common cancer in the world in 2020. Targeted inhibition is one of the important treatments for lymphoma. At present, there are many kinds of targeted drugs for the treatment of lymphoma. Studies have shown that Histone deacetylase, Bruton's tyrosine kinase and phosphoinositide 3-kinase all play an important role in the occurrence and development of tumors and become important and promising inhibitory targets. This article mainly expounds the important role of these target protein in tumors, and introduces the mechanism of action, structure-activity relationship and clinical research of listed small molecule inhibitors of these targets, hoping to provide new ideas for the treatment of lymphoma.
Collapse
Affiliation(s)
- Zhong-Hui Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou310014, P. R. China
| | - Xiang Zheng
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou310014, P. R. China
| | - Guo-Wu Rao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou310014, P. R. China
| | - Quan Zheng
- Core Facility,The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou324000, P. R. China
| |
Collapse
|
3
|
Odak I, Bayir LM, Riemann L, Sikora R, Schneider J, Xiao Y, Möhn N, Skripuletz T, Beutel G, Eder M, Ganser A, Förster R, Schultze-Florey CR, Koenecke C. Brief research report: in-depth immunophenotyping reveals stability of CD19 CAR T-cells over time. Front Immunol 2024; 15:1298598. [PMID: 38318174 PMCID: PMC10839090 DOI: 10.3389/fimmu.2024.1298598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Variability or stability might have an impact on treatment success and toxicity of CD19 CAR T-cells. We conducted a prospective observational study of 12 patients treated with Tisagenlecleucel for CD19+ B-cell malignancies. Using a 31-color spectral flow cytometry panel, we analyzed differentiation stages and exhaustion markers of CAR T-cell subsets prior to CAR T-cell infusion and longitudinally during 6 months of follow-up. The majority of activation markers on CAR T-cells showed stable expression patterns over time and were not associated with response to therapy or toxicity. Unsupervised cluster analysis revealed an immune signature of CAR T-cell products associated with the development of immune cell-associated neurotoxicity syndrome. Warranting validation in an independent patient cohort, in-depth phenotyping of CAR T-cell products as well as longitudinal monitoring post cell transfer might become a valuable tool to increase efficacy and safety of CAR T-cell therapy.
Collapse
Affiliation(s)
- Ivan Odak
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Lâle M. Bayir
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Lennart Riemann
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Ruth Sikora
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Jessica Schneider
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Yankai Xiao
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Nora Möhn
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | | | - Gernot Beutel
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Matthias Eder
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Christian R. Schultze-Florey
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Christian Koenecke
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| |
Collapse
|
4
|
Zhao Y, Guo R, Cao X, Zhang Y, Sun R, Lu W, Zhao M. Role of chemokines in T-cell acute lymphoblastic Leukemia: From pathogenesis to therapeutic options. Int Immunopharmacol 2023; 121:110396. [PMID: 37295031 DOI: 10.1016/j.intimp.2023.110396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/11/2023]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a highly heterogeneous and aggressive subtype of hematologic malignancy, with limited therapeutic options due to the complexity of its pathogenesis. Although high-dose chemotherapy and allogeneic hematopoietic stem cell transplantation have improved outcomes for T-ALL patients, there remains an urgent need for novel treatments in cases of refractory or relapsed disease. Recent research has demonstrated the potential of targeted therapies aimed at specific molecular pathways to improve patient outcomes. Chemokine-related signals, both upstream and downstream, modulate the composition of distinct tumor microenvironments, thereby regulating a multitude of intricate cellular processes such as proliferation, migration, invasion and homing. Furthermore, the progress in research has made significant contributions to precision medicine by targeting chemokine-related pathways. This review article summarizes the crucial roles of chemokines and their receptors in T-ALL pathogenesis. Moreover, it explores the advantages and disadvantages of current and potential therapeutic options that target chemokine axes, including small molecule antagonists, monoclonal antibodies, and chimeric antigen receptor T-cells.
Collapse
Affiliation(s)
- YiFan Zhao
- First Center Clinic College of Tianjin Medical University, Tianjin 300192, China
| | - RuiTing Guo
- First Center Clinic College of Tianjin Medical University, Tianjin 300192, China
| | - XinPing Cao
- First Center Clinic College of Tianjin Medical University, Tianjin 300192, China
| | - Yi Zhang
- First Center Clinic College of Tianjin Medical University, Tianjin 300192, China
| | - Rui Sun
- School of Medicine, Nankai University, Tianjin 300192, China
| | - WenYi Lu
- Department of Hematology, Tianjin First Central Hospital, Tianjin 300192, China
| | - MingFeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin 300192, China.
| |
Collapse
|
5
|
Han L, Zhang L. CCL21/CCR7 axis as a therapeutic target for autoimmune diseases. Int Immunopharmacol 2023; 121:110431. [PMID: 37331295 DOI: 10.1016/j.intimp.2023.110431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/19/2023] [Accepted: 05/30/2023] [Indexed: 06/20/2023]
Abstract
Chemokine receptor 7 (CCR7) is a G protein-coupled receptor containing 7 transmembrane domains that is expressed on various cells, such as naive T/B cells, central memory T cells, regulatory T cells, immature/mature dendritic cells (DCs), natural killer cells, and a minority of tumor cells. Chemokine ligand 21 (CCL21) is the known high-affinity ligand that binds to CCR7 and drives cell migration in tissues. CCL21 is mainly produced by stromal cells and lymphatic endothelial cells, and its expression is significantly increased under inflammatory conditions. Genome-wide association studies (GWAS) have shown a strong association between CCL21/CCR7 axis and disease severity in patients with rheumatoid arthritis, sjogren's syndrome, systemic lupus erythematosus, polymyositis, ankylosing spondylitis, and asthma. Disrupting CCL21/CCR7 interaction with antibodies or inhibitors prevents the migration of CCR7-expressing immune and non-immune cells at the site of inflammation and reduces disease severity. This review emphasizes the importance of the CCL21 /CCR7 axis in autoimmune diseases and evaluates its potential as a novel therapeutic target for these conditions.
Collapse
Affiliation(s)
- Le Han
- Department of Pharmacy, The Affiliated Jiangyin Hospital of Southeast University Medical College, Jiangyin 214400, China
| | - Lingling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
6
|
Xin S, Liu X, Li Z, Sun X, Wang R, Zhang Z, Feng X, Jin L, Li W, Tang C, Mei W, Cao Q, Wang H, Zhang J, Feng L, Ye L. ScRNA-seq revealed an immunosuppression state and tumor microenvironment heterogeneity related to lymph node metastasis in prostate cancer. Exp Hematol Oncol 2023; 12:49. [PMID: 37221625 PMCID: PMC10204220 DOI: 10.1186/s40164-023-00407-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 04/25/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Metastasis is a crucial aspect of disease progression leading to death in patients with prostate cancer (PCa). However, its mechanism remains unclear. We aimed to explore the mechanism of lymph node metastasis (LNM) by analyzing the heterogeneity of tumor microenvironment (TME) in PCa using scRNA-seq. METHODS A total of 32,766 cells were obtained from four PCa tissue samples for scRNA-seq, annotated, and grouped. InferCNV, GSVA, DEG functional enrichment analysis, trajectory analysis, intercellular network evaluation, and transcription factor analysis were carried out for each cell subgroup. Furthermore, validation experiments targeting luminal cell subgroups and CXCR4 + fibroblast subgroup were performed. RESULTS The results showed that only EEF2 + and FOLH1 + luminal subgroups were present in LNM, and they appeared at the initial stage of luminal cell differentiation, which were comfirmed by verification experiments. The MYC pathway was enriched in the EEF2 + and FOLH1 + luminal subgroups, and MYC was associated with PCa LNM. Moreover, MYC did not only promote the progression of PCa, but also led to immunosuppression in TME by regulating PDL1 and CD47. The proportion of CD8 + T cells in TME and among NK cells and monocytes was lower in LNM than in the primary lesion, while the opposite was true for Th and Treg cells. Furthermore, these immune cells in TME underwent transcriptional reprogramming, including CD8 + T subgroups of CCR7 + and IL7R+, as well as M2-like monocyte subgroups expressing tumor-associated signature genes, like CCR7, SGKI, and RPL31. Furthermore, STEAP4+, ADGRF5 + and CXCR4+, and SRGNC + fibroblast subgroups were closely related to tumor progression, tumor metabolism, and immunosuppression, indicating their contributions in PCa metastasis. Meanwhile, The presence of CXCR4 + Fibroblasts in PCa was confirmed by polychromatic immunofluorescence. CONCLUSIONS The significant heterogeneity of luminal, immune, and interstitial cells in PCa LNM may not only directly contribute to tumor progression, but also indirectly result in TME immunosuppression, which may be the cause of metastasis in PCa and in which MYC played an role.
Collapse
Affiliation(s)
- Shiyong Xin
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Ji-mo Rd, Pu-dong new District, Shanghai, 200120, China
- Department of Urology, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Xiang Liu
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Ji-mo Rd, Pu-dong new District, Shanghai, 200120, China
- Department of Urology, Putuo People's Hospital, School of Medicine, Shanghai, China
| | - Ziyao Li
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Ji-mo Rd, Pu-dong new District, Shanghai, 200120, China
| | - Xianchao Sun
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Ji-mo Rd, Pu-dong new District, Shanghai, 200120, China
| | - Rong Wang
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, 010000, China
| | - Zhenhua Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Xinwei Feng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Liang Jin
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Ji-mo Rd, Pu-dong new District, Shanghai, 200120, China
| | - Weiyi Li
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Ji-mo Rd, Pu-dong new District, Shanghai, 200120, China
| | - Chaozhi Tang
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Ji-mo Rd, Pu-dong new District, Shanghai, 200120, China
| | - Wangli Mei
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Ji-mo Rd, Pu-dong new District, Shanghai, 200120, China
| | - Qiong Cao
- Department of Pathology, The Third Affiliated Hospital of Henan University of Science and Technology, Henan, 471003, China
| | - Haojie Wang
- Department of Central Laboratory, Zhengzhou University, Luoyang Central Hospital, Luoyang, 471003, China
| | - Jianguo Zhang
- Department of Urology, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Lijin Feng
- Department of Pathology, Jing'an District Zhabei Central Hospital, No.619, Zhonghuaxin Road, Shanghai, 200070, China.
| | - Lin Ye
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Ji-mo Rd, Pu-dong new District, Shanghai, 200120, China.
| |
Collapse
|
7
|
Bordeaux ZA, Reddy SV, Lee K, Lu W, Choi J, Miller M, Roberts C, Pollizzi A, Kwatra SG, Kwatra MM. Differential Response of Mycosis Fungoides Cells to Vorinostat. Int J Mol Sci 2023; 24:ijms24098075. [PMID: 37175780 PMCID: PMC10179468 DOI: 10.3390/ijms24098075] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/23/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Mycosis fungoides (MF) is the most common form of cutaneous T-cell lymphoma (CTCL) and is characterized by epidermotrophism of malignant CD4+ T-lymphocytes. When MF advances to a recurrent stage, patients require treatment with systemic therapies such as vorinostat, a histone deacetylase inhibitor. While vorinostat has been shown to exhibit anti-tumor activity in MF, its exact molecular mechanism has yet to be fully discerned. In the present study, we examined the transcriptomic and proteomic profiles of vorinostat treatment in two MF cell lines, Myla 2059 and HH. We find that vorinostat downregulates CTLA-4, CXCR4, and CCR7 in both cell lines, but its effect on several key pathways differs between the two MF cell lines. For example, vorinostat upregulates CCL5, CCR5, and CXCL10 expression in Myla cells but downregulates CCL5 and CXCL10 expression in HH cells. Furthermore, vorinostat upregulates IFN-γ and IL-23 signaling and downregulates IL-6, IL-7, and IL-15 signaling in Myla cells but does not affect these pathways in HH cells. Although Myla and HH represent established MF cell lines, their distinct tumor origin from separate patients demonstrates that inherent phenotypic variations within the disease persist, underscoring the importance of using a variety of MF cells in the preclinical development of MF therapeutics.
Collapse
Affiliation(s)
- Zachary A Bordeaux
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sriya V Reddy
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kevin Lee
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Weiying Lu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Justin Choi
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Meghan Miller
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Callie Roberts
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Anthony Pollizzi
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Shawn G Kwatra
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Madan M Kwatra
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|