1
|
Bellavia D, Costa V, De Luca A, Maglio M, Pagani S, Fini M, Giavaresi G. Vitamin D Level Between Calcium-Phosphorus Homeostasis and Immune System: New Perspective in Osteoporosis. Curr Osteoporos Rep 2024; 22:599-610. [PMID: 27734322 DOI: 10.1007/s11914-016-0331-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Vitamin D is a key molecule in calcium and phosphate homeostasis; however, increasing evidence has recently shown that it also plays a crucial role in the immune system, both innate and adaptive. A deregulation of vitamin D levels, due also to mutations and polymorphisms in the genes of the vitamin D pathway, determines severe alterations in the homeostasis of the organism, resulting in a higher risk of onset of some diseases, including osteoporosis. This review gives an overview of the influence of vitamin D levels on the pathogenesis of osteoporosis, between bone homeostasis and immune system.
Collapse
Affiliation(s)
- Daniele Bellavia
- Innovative Technology Platforms for Tissue Engineering, Theranostics and Oncology, Rizzoli Orthopaedic Institute, Via Divisi, 83, 90100, Palermo, Italy
| | - Viviana Costa
- Innovative Technology Platforms for Tissue Engineering, Theranostics and Oncology, Rizzoli Orthopaedic Institute, Via Divisi, 83, 90100, Palermo, Italy
| | - Angela De Luca
- Innovative Technology Platforms for Tissue Engineering, Theranostics and Oncology, Rizzoli Orthopaedic Institute, Via Divisi, 83, 90100, Palermo, Italy
| | - Melania Maglio
- Laboratory of Biocompatibility, Technological Innovations and Advanced Therapies, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Stefania Pagani
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Milena Fini
- Laboratory of Biocompatibility, Technological Innovations and Advanced Therapies, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Gianluca Giavaresi
- Innovative Technology Platforms for Tissue Engineering, Theranostics and Oncology, Rizzoli Orthopaedic Institute, Via Divisi, 83, 90100, Palermo, Italy.
| |
Collapse
|
2
|
Niu Q, Hao J, Li Z, Zhang H. Helper T cells: A potential target for sex hormones to ameliorate rheumatoid arthritis? (Review). Mol Med Rep 2024; 30:215. [PMID: 39370806 PMCID: PMC11450432 DOI: 10.3892/mmr.2024.13339] [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: 07/17/2024] [Accepted: 09/06/2024] [Indexed: 10/08/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease whose etiology is not fully understood. Defective peripheral immune tolerance and subsequent mis‑differentiation and aberrant infiltration of synovium by various immune cells, especially helper T (Th) cells, play an important role in the development of RA. There are significant sex differences in RA, but the results of studies on the effects of sex hormones on RA have been difficult to standardize and hormone replacement therapy has been limited by the potential for serious side effects. Existing research has amply demonstrated that cellular immune responses are largely determined by sex and that sex hormones play a key role in Th cell responses. Based on the aforementioned background and the plasticity of Th cells, it is reasonable to hypothesize that the action of sex hormones on Th cells will hopefully become a therapeutic target for RA. The present review discussed the role of various Th cell subsets in the pathogenesis of RA and also explored the role of sex hormones on the phenotype and function of these aberrantly regulated immune cells in RA as well as other pathologic effects on RA.
Collapse
Affiliation(s)
- Quanjun Niu
- Department of Orthopedics IV, Handan Hospital of Traditional Chinese Medicine, Handan, Hebei 056001, P.R. China
| | - Junhang Hao
- Department of Orthopedics IV, Handan Hospital of Traditional Chinese Medicine, Handan, Hebei 056001, P.R. China
| | - Zhen Li
- Department of Orthopedics IV, Handan Hospital of Traditional Chinese Medicine, Handan, Hebei 056001, P.R. China
| | - Huiping Zhang
- Department of Orthopedics IV, Handan Hospital of Traditional Chinese Medicine, Handan, Hebei 056001, P.R. China
| |
Collapse
|
3
|
Çebi M, Çakar A, Durmuş H, Akan O, Aysal F, Parman Y, Saruhan-Direskeneli G. In vitro modulation of T cells in myasthenia gravis by low-dose IL-2. Eur J Immunol 2024; 54:e2451268. [PMID: 39285833 DOI: 10.1002/eji.202451268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 11/08/2024]
Abstract
Follicular helper (Tfh), peripheral helper (Tph), and regulatory (Treg) T cells are involved in myasthenia gravis (MG) pathogenesis, an autoimmune disorder arising from autoantibodies targeting neuromuscular junction proteins. This study explores the impact of low-dose IL-2 on Tfh, Tph, and Treg cells in vitro in MG. Acetylcholine-receptor antibody-positive MG (AChR-MG), muscle-specific kinase antibody-positive MG (MuSK-MG) patients, and healthy controls (HC) were studied. Blood cells were cultured with/without IL-2 and compared by the ratios of IL-2 stimulated/unstimulated cultures. In both AChR-MG and MuSK-MG patients, CD25+FoxP3+Tregs were lower, while CXCR5+PD-1+ or ICOS+Tfh and CXCR5-PD-1+ or ICOS+Tph cells were higher compared with HC. Among the MG group, the FoxP3+ Treg cells in AChR-MG patients were even lower compared with MuSK-MG patients. In vitro IL-2 stimulation increased Tregs in all groups while decreasing PD-1+/ICOS+Tfh and PD-1+/ICOS+Tph populations. The fold-increase ratio of Tregs and the fold-decrease ratio of PD-1+ or ICOS+Tfh and ICOS+Tph cells in AChR-MG and MuSK-MG patients were greater than in HCs. Low-dose IL-2 treatment may balance Tfh, Tph, and Treg cells in MG patients, offering a potential opportunity for disease modulation.
Collapse
Affiliation(s)
- Merve Çebi
- Department of Physiology, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
- Department of Immunology, Institute of Health Sciences, Istanbul University, Istanbul, Turkey
| | - Arman Çakar
- Department of Neurology, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Hacer Durmuş
- Department of Neurology, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Onur Akan
- Department of Neurology, Prof. Dr. Cemil Tascioglu City Hospital, University of Health Sciences, Istanbul, Turkey
| | | | - Yeşim Parman
- Department of Neurology, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | | |
Collapse
|
4
|
Pamuk S, Ertugrul B, Kasarci G, Bireller S, Ergen A, Cakmakoglu B, Ulusan M. Focusing on tumor and it's microenvironmental immune members for head and neck cancer patients. Pathol Res Pract 2024; 263:155575. [PMID: 39236499 DOI: 10.1016/j.prp.2024.155575] [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: 07/08/2024] [Revised: 08/09/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
OBJECTIVE Immune-related gene expression levels in the tumor microenvironment (TM) of head and neck squamous cell carcinoma (HNSCC) patients was compared. MATERIALS AND METHODS The CD163, CD274, CD86, FUT4, FOXP3, and ITGAX levels of HNSCC patients in their tumor tissues (n =76) and surrounding tissues adjacent to the tumor (n =76) were determined using quantitative real-time PCR (qRT-PCR). Changes in these genes were also evaluated by associating with demographical data of the patients. RESULTS CD163, CD274, FUT4, and FOXP3 gene expression levels were significantly higher in tumor tissue than in surrounding tissue. FUT4 fold change was statistically higher in patients with lymph node involvement. CD86 expression was statistically lower in smokers of 50 boxes per year or more. CD163, CD274, and FUT4 expressions were increased in response to the presence of extranodal extension (ENE). CONCLUSIONS These preliminary results demonstrate the alterations in expression levels of immunologic markers are associated with the clinical presentations of HNSCC. AVAILABILITY OF DATA AND MATERIALS The datasets used and/or analysed during the current study available from the corresponding author on reasonable request.
Collapse
Affiliation(s)
- Saim Pamuk
- Department of Otorhinolaryngology & Head and Neck Surgery, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Baris Ertugrul
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Goksu Kasarci
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey; Graduate School of Health Sciences, Istanbul University, Istanbul, Turkey
| | - Sinem Bireller
- Department of Biochemistry, Faculty of Pharmacy, Acibadem Mehmet Ali Aydınlar University, Istanbul, Turkey
| | - Arzu Ergen
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Bedia Cakmakoglu
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey.
| | - Murat Ulusan
- Department of Otorhinolaryngology & Head and Neck Surgery, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
5
|
Anvar MT, Rashidan K, Arsam N, Rasouli-Saravani A, Yadegari H, Ahmadi A, Asgari Z, Vanan AG, Ghorbaninezhad F, Tahmasebi S. Th17 cell function in cancers: immunosuppressive agents or anti-tumor allies? Cancer Cell Int 2024; 24:355. [PMID: 39465401 PMCID: PMC11514949 DOI: 10.1186/s12935-024-03525-9] [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: 08/13/2024] [Accepted: 10/08/2024] [Indexed: 10/29/2024] Open
Abstract
T helper (Th) 17 cells, a distinct subset of Th lymphocytes, are known for their prominent interleukin (IL)-17 production and other pro-inflammatory cytokines. These cells exhibit remarkable plasticity, allowing them to exhibit different phenotypes in the cancer microenvironment. This adaptability enables Th17 cells to promote tumor progression by immunosuppressive activities and angiogenesis, but also mediate anti-tumor immune responses through employing immune cells in tumor setting or even by directly converting toward Th1 phenotype and producing interferon-gamma (IFN-γ). This dual role of Th17 cells in cancer makes it a double-edged sword in encountering cancer. In this review, we aim to elucidate the complexities of Th17 cell function in cancer by summarizing recent studies and, ultimately, to design novel therapeutic strategies, especially targeting Th17 cells in the tumor milieu, which could pave the way for more effective cancer treatments.
Collapse
Affiliation(s)
- Milad Taghizadeh Anvar
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimiya Rashidan
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nima Arsam
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ashkan Rasouli-Saravani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamidreza Yadegari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Ahmadi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeynab Asgari
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Ghorbani Vanan
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Farid Ghorbaninezhad
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Safa Tahmasebi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Huang Q, Zhu J. Regulatory T cell-based therapy in type 1 diabetes: Latest breakthroughs and evidence. Int Immunopharmacol 2024; 140:112724. [PMID: 39098233 DOI: 10.1016/j.intimp.2024.112724] [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: 06/07/2024] [Revised: 07/10/2024] [Accepted: 07/16/2024] [Indexed: 08/06/2024]
Abstract
Autoimmune diseases (ADs) are among the most significant health complications, with their incidence rising in recent years. Type 1 diabetes (T1D), an AD, targets the insulin-producing β cells in the pancreas, leading to chronic insulin deficiency in genetically susceptible individuals. Regulatory immune cells, particularly T-cells (Tregs), have been shown to play a crucial role in the pathogenesis of diabetes by modulating immune responses. In diabetic patients, Tregs often exhibit diminished effectiveness due to various factors, such as instability in forkhead box P3 (Foxp3) expression or abnormal production of the proinflammatory cytokine interferon-gamma (IFN-γ) by autoreactive T-cells. Consequently, Tregs represent a potential therapeutic target for diabetes treatment. Building on the successful clinical outcomes of chimeric antigen receptor (CAR) T-cell therapy in cancer treatment, particularly in leukemias, the concept of designing and utilizing CAR Tregs for ADs has emerged. This review summarizes the findings on Treg targeting in T1D and discusses the benefits and limitations of this treatment approach for patients suffering from T1D.
Collapse
Affiliation(s)
- Qiongxiao Huang
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China
| | - Jing Zhu
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China.
| |
Collapse
|
7
|
Kaur P, Singh SK, Mishra MK, Singh S, Singh R. Nanotechnology for boosting ovarian cancer immunotherapy. J Ovarian Res 2024; 17:202. [PMID: 39402681 PMCID: PMC11475952 DOI: 10.1186/s13048-024-01507-z] [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: 08/04/2024] [Accepted: 08/28/2024] [Indexed: 10/19/2024] Open
Abstract
Ovarian cancer, often referred to as the "silent killer," is notoriously difficult to detect in its early stages, leading to a poor prognosis for many patients. Diagnosis is often delayed until the cancer has advanced, primarily due to its ambiguous and frequently occurring clinical symptoms. Ovarian cancer leads to more deaths than any other cancer of the female reproductive system. The main reasons for the high mortality rates include delayed diagnosis and resistance to treatment. As a result, there is an urgent need for improved diagnostic and treatment options for ovarian cancer. The standard treatments typically involve debulking surgery along with platinum-based chemotherapies. Among patients with advanced-stage cancer who initially respond to current therapies, 50-75% experience a recurrence. Recently, immunotherapy-based approaches to enhance the body's immune response to combat tumor growth have shown promise. Immune checkpoint inhibitors have shown promising results in treating other types of tumors. However, in ovarian cancer, only a few of these inhibitors have been effective because the tumor's environment suppresses the immune system and creates barriers for treatment. This hampers the effectiveness of existing immunotherapies. Nonetheless, advanced immunotherapy techniques and delivery systems based on nanotechnology hold promise for overcoming these challenges.
Collapse
Affiliation(s)
- Prabhjot Kaur
- Department of Microbiology, Biochemistry and Immunology, Cancer Health Equity Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA, 30310, USA
| | - Santosh Kumar Singh
- Department of Microbiology, Biochemistry and Immunology, Cancer Health Equity Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA, 30310, USA
| | - Manoj K Mishra
- Cancer Biology Research and Training, Department of Biological Sciences, Alabama State University, Montgomery, AL, 36014, USA
| | - Shailesh Singh
- Department of Microbiology, Biochemistry and Immunology, Cancer Health Equity Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA, 30310, USA
- Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Rajesh Singh
- Department of Microbiology, Biochemistry and Immunology, Cancer Health Equity Institute, Morehouse School of Medicine, 720 Westview Drive SW, Atlanta, GA, 30310, USA.
- Cancer Health Equity Institute, Morehouse School of Medicine, Atlanta, GA, 30310, USA.
| |
Collapse
|
8
|
Chandwaskar R, Dalal R, Gupta S, Sharma A, Parashar D, Kashyap VK, Sohal JS, Tripathi SK. Dysregulation of T cell response in the pathogenesis of inflammatory bowel disease. Scand J Immunol 2024:e13412. [PMID: 39394898 DOI: 10.1111/sji.13412] [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: 11/20/2023] [Revised: 08/26/2024] [Accepted: 09/17/2024] [Indexed: 10/14/2024]
Abstract
Inflammatory bowel disease (IBD), comprised of Crohn's disease (CD) and ulcerative colitis (UC), are gut inflammatory diseases that were earlier prevalent in the Western Hemisphere but now are on the rise in the East, with India standing second highest in the incidence rate in the world. Inflammation in IBD is a cause of dysregulated immune response, wherein helper T (Th) cell subsets and their cytokines play a major role in the pathogenesis of IBD. In addition, gut microbiota, environmental factors such as dietary factors and host genetics influence the outcome and severity of IBD. Dysregulation between effector and regulatory T cells drives gut inflammation, as effector T cells like Th1, Th17 and Th9 subsets Th cell lineages were found to be increased in IBD patients. In this review, we attempted to discuss the role of different Th cell subsets together with other T cells like CD8+ T cells, NKT and γδT cells in the outcome of gut inflammation in IBD. We also highlighted the potential therapeutic candidates for IBD.
Collapse
Affiliation(s)
- Rucha Chandwaskar
- Amity Institute of Microbial Technology (AIMT), Amity University Jaipur, Rajasthan, India
| | - Rajdeep Dalal
- Infection and Immunology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, India
| | - Saurabh Gupta
- Centre for Vaccines and Diagnostic Research, GLA University, Mathura, Uttar Pradesh, India
| | - Aishwarya Sharma
- Sri Siddhartha Medical College and Research Center, Tumkur, Karnataka, India
| | - Deepak Parashar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Vivek K Kashyap
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
| | - Jagdip Singh Sohal
- Centre for Vaccines and Diagnostic Research, GLA University, Mathura, Uttar Pradesh, India
| | - Subhash K Tripathi
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, Washington, USA
| |
Collapse
|
9
|
Sun Y, Pan Z, Wang Z, Wang H, Wei L, Cui F, Zou Q, Zhang Z. Single-cell transcriptome analysis reveals immune microenvironment changes and insights into the transition from DCIS to IDC with associated prognostic genes. J Transl Med 2024; 22:894. [PMID: 39363164 PMCID: PMC11448450 DOI: 10.1186/s12967-024-05706-6] [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: 06/27/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Ductal carcinoma in situ (DCIS) of the breast is an early stage of breast cancer, and preventing its progression to invasive ductal carcinoma (IDC) is crucial for the early detection and treatment of breast cancer. Although single-cell transcriptome analysis technology has been widely used in breast cancer research, the biological mechanisms underlying the transition from DCIS to IDC remain poorly understood. RESULTS We identified eight cell types through cell annotation, finding significant differences in T cell proportions between DCIS and IDC. Using this as a basis, we performed pseudotime analysis on T cell subpopulations, revealing that differentially expressed genes primarily regulate immune cell migration and modulation. By intersecting WGCNA results of T cells highly correlated with the subtypes and the differentially expressed genes, we identified six key genes: FGFBP2, GNLY, KLRD1, TYROBP, PRF1, and NKG7. Excluding PRF1, the other five genes were significantly associated with overall survival in breast cancer, highlighting their potential as prognostic biomarkers. CONCLUSIONS We identified immune cells that may play a role in the progression from DCIS to IDC and uncovered five key genes that can serve as prognostic markers for breast cancer. These findings provide insights into the mechanisms underlying the transition from DCIS to IDC, offering valuable perspectives for future research. Additionally, our results contribute to a better understanding of the biological processes involved in breast cancer progression.
Collapse
MESH Headings
- Humans
- Single-Cell Analysis
- Female
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
- Gene Expression Profiling
- Prognosis
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Carcinoma, Intraductal, Noninfiltrating/immunology
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Breast Neoplasms/genetics
- Breast Neoplasms/immunology
- Breast Neoplasms/pathology
- Gene Expression Regulation, Neoplastic
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/immunology
- Disease Progression
- Transcriptome/genetics
- Single-Cell Gene Expression Analysis
Collapse
Affiliation(s)
- Yidi Sun
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China
| | - Zhuoyu Pan
- International Business School, Hainan University, Haikou, 570228, China
| | - Ziyi Wang
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China
| | - Haofei Wang
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China
| | - Leyi Wei
- Centre for Artificial Intelligence driven Drug Discovery, Faculty of Applied Science, Macao Polytechnic University, Macao SAR, China
- School of Informatics, Xiamen University, Xiamen, China
| | - Feifei Cui
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China.
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610054, China.
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, 324000, China.
| | - Zilong Zhang
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China.
| |
Collapse
|
10
|
Rubino V, Carriero F, Palatucci AT, Giovazzino A, Salemi F, Carrano R, Sabbatini M, Ruggiero G, Terrazzano G. T R3-56 and Treg Regulatory T Cell Subsets as Potential Indicators of Graft Tolerance Control in Kidney Transplant Recipients. Int J Mol Sci 2024; 25:10610. [PMID: 39408939 PMCID: PMC11477056 DOI: 10.3390/ijms251910610] [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: 08/02/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Identification of early signatures of immune rejection represents a key challenge in the clinical management of kidney transplant. To address such an issue, we enrolled 53 kidney transplant recipients without signs of graft rejection, no infectious episodes and no change in the immunosuppressive regimen in the last 6 months. An extensive immune profile revealed increased activation of the T cells, a decreased amount and growth ability of the Treg and a higher level of the TR3-56 regulatory T cell subset, described by us as involved in the preferential control of cytotoxic T lymphocytes. In renal transplant recipients, the high level of the TR3-56 cells associates with a reduction in both the amount and the growth ability of the Treg. Moreover, when the transplanted subjects were categorised according to their stable or unstable disease status, as defined by changes in serum creatinine ≥0.2 mg/dL in two consecutive detections, a higher TR3-56 level and defective Treg growth ability were observed to characterise patients with unstable graft control. Further studies are required to substantiate the hypothesis that immune profiling, including TR3-56 evaluation, might represent a valuable diagnostic tool to identify patients at risk of developing significant anti-donor allo-immune responses.
Collapse
Affiliation(s)
- Valentina Rubino
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli “Federico II”, 80131 Napoli, Italy; (V.R.); (A.G.)
| | - Flavia Carriero
- Dipartimento di Scienze Della Salute, Università Della Basilicata, 85100 Potenza, Italy; (F.C.); (A.T.P.); (G.T.)
| | - Anna Teresa Palatucci
- Dipartimento di Scienze Della Salute, Università Della Basilicata, 85100 Potenza, Italy; (F.C.); (A.T.P.); (G.T.)
| | - Angela Giovazzino
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli “Federico II”, 80131 Napoli, Italy; (V.R.); (A.G.)
| | - Fabrizio Salemi
- Percorso Clinico Assistenziale in Nefrologia e Trapianto Renale, Azienda Ospedaliera Universitaria “Federico II”, 80131 Napoli, Italy; (F.S.); (R.C.)
| | - Rosa Carrano
- Percorso Clinico Assistenziale in Nefrologia e Trapianto Renale, Azienda Ospedaliera Universitaria “Federico II”, 80131 Napoli, Italy; (F.S.); (R.C.)
| | - Massimo Sabbatini
- Dipartimento di Sanità Pubblica, Sezione di Nefrologia, Università di Napoli “Federico II”, 80131 Napoli, Italy;
| | - Giuseppina Ruggiero
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli “Federico II”, 80131 Napoli, Italy; (V.R.); (A.G.)
| | - Giuseppe Terrazzano
- Dipartimento di Scienze Della Salute, Università Della Basilicata, 85100 Potenza, Italy; (F.C.); (A.T.P.); (G.T.)
| |
Collapse
|
11
|
Roškar Z, Dreisinger M, Homšak E, Avčin T, Bevc S, Goropevšek A. Increased Frequency of Circulating Activated FOXP3 + Regulatory T Cell Subset in Patients with Chronic Lymphocytic Leukemia Is Associated with the Estimate of the Size of the Tumor Mass, STAT5 Signaling and Disease Course during Follow-Up of Patients on Therapy. Cancers (Basel) 2024; 16:3228. [PMID: 39335199 PMCID: PMC11430700 DOI: 10.3390/cancers16183228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
INTRODUCTION Advanced chronic lymphocytic leukemia (CLL) is accompanied by increased circulating regulatory T cells (Tregs) and increased susceptibility to severe infections, which were also shown to entail a striking induction of FOXP3 expression in Tregs. As homeostasis of the most suppressive CD45RA-FOXP3high activated Treg (aTreg) subset differs, it is critical to analyse homeostatic signalling in Treg subsets. MATERIALS AND METHODS In this study, by using conventional and imaging flow cytometry, we monitored STAT5 signalling/phosphorylation (pSTAT5) and investigated Treg subsets in relation to the Binet stage, the total tumor mass score (TTM) and the disease course during a follow-up of 37 patients with CLL. RESULTS The aTreg percentage was significantly increased among CD4+ T cells from patients with advanced disease and significantly correlated with the TTM. A subgroup of patients with higher aTreg percentages among CD4+FOXP3+ T cells at the start of therapy was characterised by more frequent episodes of severe infections during follow-up. CONCLUSIONS The results suggesting that an aTreg fraction could represent a possible marker of a severe disease course with infectious complications. Augmented homeostatic STAT5 signalling could support aTreg expansion, as higher pSTAT5 levels were significantly correlated with an increased aTreg frequency among CD4+FOXP3+ T cells during the follow-up of patients on therapy, as well as following SARS-CoV-2 antigen-specific stimulation in vitro.
Collapse
Affiliation(s)
- Zlatko Roškar
- Department of Haematology, University Medical Centre Maribor, 2000 Maribor, Slovenia
| | - Mojca Dreisinger
- Department of Haematology, University Medical Centre Maribor, 2000 Maribor, Slovenia
| | - Evgenija Homšak
- Department of Laboratory Diagnostics, University Medical Centre Maribor, 2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Tadej Avčin
- Department of Allergology, Rheumatology and Clinical Immunology, Children's Hospital, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Department of Pediatrics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Sebastjan Bevc
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
- Department of Nephrology, University Medical Centre Maribor, 2000 Maribor, Slovenia
| | - Aleš Goropevšek
- Department of Laboratory Diagnostics, University Medical Centre Maribor, 2000 Maribor, Slovenia
| |
Collapse
|
12
|
Antony F, Kinha D, Nowińska A, Rouse BT, Suryawanshi A. The immunobiology of corneal HSV-1 infection and herpetic stromal keratitis. Clin Microbiol Rev 2024; 37:e0000624. [PMID: 39078136 PMCID: PMC11391706 DOI: 10.1128/cmr.00006-24] [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] [Indexed: 07/31/2024] Open
Abstract
SUMMARYHuman alphaherpesvirus 1 (HSV-1) is a highly successful neurotropic pathogen that primarily infects the epithelial cells lining the orofacial mucosa. After primary lytic replication in the oral, ocular, and nasal mucosal epithelial cells, HSV-1 establishes life-long latency in neurons within the trigeminal ganglion. Patients with compromised immune systems experience frequent reactivation of HSV-1 from latency, leading to virus entry in the sensory neurons, followed by anterograde transport and lytic replication at the innervated mucosal epithelial surface. Although recurrent infection of the corneal mucosal surface is rare, it can result in a chronic immuno-inflammatory condition called herpetic stromal keratitis (HSK). HSK leads to gradual vision loss and can cause permanent blindness in severe untreated cases. Currently, there is no cure or successful vaccine to prevent latent or recurrent HSV-1 infections, posing a significant clinical challenge to managing HSK and preventing vision loss. The conventional clinical management of HSK primarily relies on anti-virals to suppress HSV-1 replication, anti-inflammatory drugs (such as corticosteroids) to provide symptomatic relief from pain and inflammation, and surgical interventions in more severe cases to replace damaged cornea. However, each clinical treatment strategy has limitations, such as local and systemic drug toxicities and the emergence of anti-viral-resistant HSV-1 strains. In this review, we summarize the factors and immune cells involved in HSK pathogenesis and highlight alternate therapeutic strategies for successful clinical management of HSK. We also discuss the therapeutic potential of immunoregulatory cytokines and immunometabolism modulators as promising HSK therapies against emerging anti-viral-resistant HSV-1 strains.
Collapse
Affiliation(s)
- Ferrin Antony
- Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
| | - Divya Kinha
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Anna Nowińska
- Clinical Department of Ophthalmology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
- Ophthalmology Department, Railway Hospital in Katowice, Katowice, Poland
| | - Barry T Rouse
- College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Amol Suryawanshi
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
13
|
Zhou Z, Xu J, Liu S, Lv Y, Zhang R, Zhou X, Zhang Y, Weng S, Xu H, Ba Y, Zuo A, Han X, Liu Z. Infiltrating treg reprogramming in the tumor immune microenvironment and its optimization for immunotherapy. Biomark Res 2024; 12:97. [PMID: 39227959 PMCID: PMC11373505 DOI: 10.1186/s40364-024-00630-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/31/2024] [Indexed: 09/05/2024] Open
Abstract
Immunotherapy has shown promising anti-tumor effects across various tumors, yet it encounters challenges from the inhibitory tumor immune microenvironment (TIME). Infiltrating regulatory T cells (Tregs) are important contributors to immunosuppressive TIME, limiting tumor immunosurveillance and blocking effective anti-tumor immune responses. Although depletion or inhibition of systemic Tregs enhances the anti-tumor immunity, autoimmune sequelae have diminished expectations for the approach. Herein, we summarize emerging strategies, specifically targeting tumor-infiltrating (TI)-Tregs, that elevate the capacity of organisms to resist tumors by reprogramming their phenotype. The regulatory mechanisms of Treg reprogramming are also discussed as well as how this knowledge could be utilized to develop novel and effective cancer immunotherapies.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Henan, 450052, China
| | - Jiaxin Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Department of Human Anatomy, School of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yingying Lv
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ruiqi Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xing Zhou
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
14
|
He M, Zong X, Xu B, Qi W, Huang W, Djekidel MN, Zhang Y, Pagala VR, Li J, Hao X, Guy C, Bai L, Cross R, Li C, Peng J, Feng Y. Dynamic Foxp3-chromatin interaction controls tunable Treg cell function. J Exp Med 2024; 221:e20232068. [PMID: 38935023 PMCID: PMC11211070 DOI: 10.1084/jem.20232068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 04/11/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Nuclear factor Foxp3 determines regulatory T (Treg) cell fate and function via mechanisms that remain unclear. Here, we investigate the nature of Foxp3-mediated gene regulation in suppressing autoimmunity and antitumor immune response. Contrasting with previous models, we find that Foxp3-chromatin binding is regulated by Treg activation states, tumor microenvironment, and antigen and cytokine stimulations. Proteomics studies uncover dynamic proteins within Foxp3 proximity upon TCR or IL-2 receptor signaling in vitro, reflecting intricate interactions among Foxp3, signal transducers, and chromatin. Pharmacological inhibition and genetic knockdown experiments indicate that NFAT and AP-1 protein Batf are required for enhanced Foxp3-chromatin binding in activated Treg cells and tumor-infiltrating Treg cells to modulate target gene expression. Furthermore, mutations at the Foxp3 DNA-binding domain destabilize the Foxp3-chromatin association. These representative settings delineate context-dependent Foxp3-chromatin interaction, suggesting that Foxp3 associates with chromatin by hijacking DNA-binding proteins resulting from Treg activation or differentiation, which is stabilized by direct Foxp3-DNA binding, to dynamically regulate Treg cell function according to immunological contexts.
Collapse
Affiliation(s)
- Minghong He
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Xinying Zong
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Beisi Xu
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Wenjie Qi
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Wenjun Huang
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | | | - Yang Zhang
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Vishwajeeth R. Pagala
- Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Jun Li
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Xiaolei Hao
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Clifford Guy
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Lu Bai
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Richard Cross
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Chunliang Li
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Junmin Peng
- Department of Structure Biology and Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Yongqiang Feng
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
15
|
Gharib E, Robichaud GA. From Crypts to Cancer: A Holistic Perspective on Colorectal Carcinogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:9463. [PMID: 39273409 PMCID: PMC11395697 DOI: 10.3390/ijms25179463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Gilles A Robichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
16
|
Akimova T, Wang L, Bartosh Z, Christensen LM, Eruslanov E, Singhal S, Aishwarya V, Hancock WW. Antisense targeting of FOXP3+ Tregs to boost anti-tumor immunity. Front Immunol 2024; 15:1426657. [PMID: 39234236 PMCID: PMC11371716 DOI: 10.3389/fimmu.2024.1426657] [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: 05/01/2024] [Accepted: 07/29/2024] [Indexed: 09/06/2024] Open
Abstract
Our goal is to improve the outcomes of cancer immunotherapy by targeting FOXP3+ T-regulatory (Treg) cells with a next generation of antisense oligonucleotides (ASO), termed FOXP3 AUMsilence ASO. We performed in vitro experiments with human healthy donor PBMC and clinical samples from patients with lung cancer, mesothelioma and melanoma, and tested our approach in vivo using ASO FOXP3 in syngeneic murine cancer models and in humanized mice. ASO FOXP3 had no effects on cell viability or cell division, did not affect expression of other FOXP members, but decreased expression of FOXP3 mRNA in PBMC by 54.9% and in cancer samples by 64.7%, with corresponding 41.0% (PBMC) and 60.0% (cancer) decreases of Treg numbers (all p<0.0001). Hence, intratumoral Treg were more sensitive to the effects of ASO FOXP3 than peripheral blood Tregs. Isolated human Treg, incubated with ASO FOXP3 for 3.5 hours, had significantly impaired suppressive function (66.4%) versus Scramble control. In murine studies, we observed a significant inhibition of tumor growth, while 13.6% (MC38) to 22% (TC1) of tumors were completely resorbed, in conjunction with ~50% decrease of Foxp3 mRNA by qPCR and decreased numbers of intratumoral Tregs. In addition, there were no changes in FOXP3 mRNA expression or in the numbers of Tregs in draining lymph nodes and in spleens of tumor bearing mice, confirming that intratumoral Treg had enhanced sensitivity to ASO FOXP3 in vivo compared to other Treg populations. ASO FOXP3 Treg targeting in vivo and in vitro was accompanied by significant downregulation of multiple exhaustion markers, and by increased expression of perforin and granzyme-B by intratumoral T cells. To conclude, we report that targeting the key Treg transcription factor FOXP3, with ASO FOXP3, has a powerful anti-tumoral effect and enhances T cell response in vitro and in vivo.
Collapse
Affiliation(s)
- Tatiana Akimova
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- AUM Biotech, LLC., Philadelphia, PA, United States
| | - Liqing Wang
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Zhanna Bartosh
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- AUM Biotech, LLC., Philadelphia, PA, United States
| | - Lanette M. Christensen
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Evgeniy Eruslanov
- Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Sunil Singhal
- Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | | | - Wayne W. Hancock
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
17
|
Kamaraj US, Gautam P, Cheng T, Chin TS, Tay SK, Ho TH, Nadarajah R, Goh RCH, Wong SL, Mantoo S, Busmanis I, Li H, Le MT, Li QJ, Lim EH, Loh YH. Deciphering tumour microenvironment and elucidating the origin of cancer cells in ovarian clear cell carcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.06.606821. [PMID: 39149248 PMCID: PMC11326226 DOI: 10.1101/2024.08.06.606821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Ovarian clear cell carcinoma (CCC) has an East Asian preponderance. It is associated with endometriosis, a benign condition where endometrial (inner lining of the uterus) tissue is found outside the uterus and on the peritoneal surface, in the abdominal or pelvic space. CCC is relatively more resistant to conventional chemotherapy compared to other ovarian cancer subtypes and is associated with a poorer prognosis. In this study, we recruited and obtained tumour tissues from seven patients across the four stages of CCC. The tumour and the tumour microenvironment (TME) from 7 CCC patients spanning clinical stages 1-4 were transcriptionally profiled using high-resolution scRNA-seq to gain insight into CCC's biological mechanisms. Firstly, we built a scRNA-seq resource for the CCC tumour microenvironment (TME). Secondly, we identified the different cell type proportions and found high levels of immune infiltration in CCC. Thirdly, since CCC is associated with endometriosis, we compared CCC with two publicly available endometriosis scRNA-seq datasets. The CCC malignant cells showed similarities with glandular secretory and ciliated epithelial cells found in endometriosis. Finally, we determined the differences in cell-cell communication between various cell types present in CCC TME and endometriosis conditions to gain insights into the transformations in CCC.
Collapse
Affiliation(s)
- Uma S Kamaraj
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Pradeep Gautam
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Terence Cheng
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Tham Su Chin
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Sun Kuie Tay
- Department of Obstetrics & Gynaecology, Singapore General Hospital, Outram Road, Singapore 169608
| | - Tew Hong Ho
- Department of Obstetrics & Gynaecology, Singapore General Hospital, Outram Road, Singapore 169608
| | - Ravichandran Nadarajah
- Department of Obstetrics & Gynaecology, Singapore General Hospital, Outram Road, Singapore 169608
| | - Ronald Chin Hong Goh
- Department of Anatomical Pathology, Singapore General Hospital, Academia, College Road, Singapore 169856
| | - Shing Lih Wong
- Department of Anatomical Pathology, Singapore General Hospital, Academia, College Road, Singapore 169856
| | - Sangeeta Mantoo
- Department of Anatomical Pathology, Singapore General Hospital, Academia, College Road, Singapore 169856
| | - Inny Busmanis
- Department of Anatomical Pathology, Singapore General Hospital, Academia, College Road, Singapore 169856
| | - Hu Li
- Center for Individualized Medicine, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Minh Tn Le
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Qi-Jing Li
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Elaine Hsuen Lim
- Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Boulevard, Singapore 168583
| | - Yuin-Han Loh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
- Department of Physiology, NUS Yong Loo Lin School of Medicine, 2 Medical Drive, MD9, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- NUS Graduate School's Integrative Sciences and Engineering Programme, National University of Singapore, 28 Medical Drive, Singapore, Singapore
| |
Collapse
|
18
|
Christensen LM, Akimova T, Wang L, Han R, Samanta A, Di Giorgio E, Hancock WW. T-regulatory cells require Sin3a for stable expression of Foxp3. Front Immunol 2024; 15:1444937. [PMID: 39156895 PMCID: PMC11327135 DOI: 10.3389/fimmu.2024.1444937] [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: 06/06/2024] [Accepted: 07/08/2024] [Indexed: 08/20/2024] Open
Abstract
Histone deacetylases 1 and 2 play a major role in the transcriptional regulation of T-regulatory (Treg) cells via interactions with a myriad of coregulatory factors. Sin3a has been well established as a Hdac1/2 cofactor, while its role within Tregs has not been established. In this study, the effects of conditional deletion of Sin3a within Foxp3+ Tregs were evaluated. Developmental deletion of Sin3a from Foxp3+ Tregs resulted in the rapid onset of fatal autoimmunity. Treg numbers were greatly reduced, while residual Tregs had impaired suppressive function. Mice also showed effector T-cell activation, autoantibody production, and widespread tissue injury. Mechanistically, Sin3a deletion resulted in decreased transcription of Foxp3 with a complete lack of CNS2 CpG demethylation. In addition, Foxp3 protein stability was impaired with an increased ex-Treg population. Thus, Sin3a plays a critical role in the maintenance of Treg identity and function and is essential for the expression and stability of Foxp3.
Collapse
Affiliation(s)
- Lanette M. Christensen
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Tatiana Akimova
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Liqing Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Rongxiang Han
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Arabinda Samanta
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | | | - Wayne W. Hancock
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
19
|
Younis N, Puigmal N, Kurdi AE, Badaoui A, Zhang D, Morales-Garay C, Saad A, Cruz D, Rahy NA, Daccache A, Huerta T, Deban C, Halawi A, Choi J, Dosta P, Guo Lian C, Artzi N, Azzi JR. Microneedle-Mediated Delivery of Immunomodulators Restores Immune Privilege in Hair Follicles and Reverses Immune-Mediated Alopecia. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312088. [PMID: 38638030 DOI: 10.1002/adma.202312088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 04/05/2024] [Indexed: 04/20/2024]
Abstract
Disorders in the regulatory arm of the adaptive immune system result in autoimmune-mediated diseases. While systemic immunosuppression is the prevailing approach to manage them, it fails to achieve long-lasting remission due to concomitant suppression of the regulatory arm and carries the risk of heightened susceptibility to infections and malignancies. Alopecia areata is a condition characterized by localized hair loss due to autoimmunity. The accessibility of the skin allows local rather than systemic intervention to avoid broad immunosuppression. It is hypothesized that the expansion of endogenous regulatory T cells (Tregs) at the site of antigen encounter can restore the immune balance and generate a long-lasting tolerogenic response. A hydrogel microneedle (MN) patch is therefore utilized for delivery of CCL22, a Treg-chemoattractant, and IL-2, a Treg survival factor to amplify them. In an immune-mediated murine model of alopecia, local bolstering of Treg numbers is shown, leading to sustained hair regrowth and attenuation of inflammatory pathways. In a humanized skin transplant mouse model, expansion of Tregs within human skin is confirmed without engendering peripheral immunosuppression. The patch offers high-loading capacity and shelf-life stability for prospective clinical translation. By harmonizing immune responses locally, the aim is to reshape the landscape of autoimmune skin disease management.
Collapse
Affiliation(s)
- Nour Younis
- Brigham and Woman's Hospital, Department of Medicine, Renal Division, Harvard Medical School, Boston, MA, 02115, USA
| | - Núria Puigmal
- Brigham and Woman's Hospital, Department of Medicine, Division of Engineering in Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Abdallah El Kurdi
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, 11-0236, Lebanon
| | - Andrew Badaoui
- Brigham and Woman's Hospital, Department of Medicine, Renal Division, Harvard Medical School, Boston, MA, 02115, USA
| | - Dongliang Zhang
- Brigham and Woman's Hospital, Department of Medicine, Renal Division, Harvard Medical School, Boston, MA, 02115, USA
| | - Claudia Morales-Garay
- Brigham and Woman's Hospital, Department of Medicine, Division of Engineering in Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Anis Saad
- Brigham and Woman's Hospital, Department of Medicine, Renal Division, Harvard Medical School, Boston, MA, 02115, USA
| | - Diane Cruz
- Brigham and Woman's Hospital, Department of Medicine, Division of Engineering in Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Nadim Al Rahy
- Brigham and Woman's Hospital, Department of Medicine, Renal Division, Harvard Medical School, Boston, MA, 02115, USA
| | - Andrea Daccache
- Brigham and Woman's Hospital, Department of Medicine, Renal Division, Harvard Medical School, Boston, MA, 02115, USA
| | - Triana Huerta
- Brigham and Woman's Hospital, Department of Medicine, Division of Engineering in Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Christa Deban
- Brigham and Woman's Hospital, Department of Medicine, Renal Division, Harvard Medical School, Boston, MA, 02115, USA
| | - Ahmad Halawi
- Brigham and Woman's Hospital, Department of Medicine, Renal Division, Harvard Medical School, Boston, MA, 02115, USA
| | - John Choi
- Brigham and Woman's Hospital, Department of Medicine, Renal Division, Harvard Medical School, Boston, MA, 02115, USA
| | - Pere Dosta
- Brigham and Woman's Hospital, Department of Medicine, Division of Engineering in Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Christine Guo Lian
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Natalie Artzi
- Brigham and Woman's Hospital, Department of Medicine, Division of Engineering in Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Jamil R Azzi
- Brigham and Woman's Hospital, Department of Medicine, Renal Division, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
20
|
Nguyen BA, Alexander MR, Harrison DG. Immune mechanisms in the pathophysiology of hypertension. Nat Rev Nephrol 2024; 20:530-540. [PMID: 38658669 DOI: 10.1038/s41581-024-00838-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2024] [Indexed: 04/26/2024]
Abstract
Hypertension is a leading risk factor for morbidity and mortality worldwide. Despite current anti-hypertensive therapies, most individuals with hypertension fail to achieve adequate blood pressure control. Moreover, even with adequate control, a residual risk of cardiovascular events and associated organ damage remains. These findings suggest that current treatment modalities are not addressing a key element of the underlying pathology. Emerging evidence implicates immune cells as key mediators in the development and progression of hypertension. In this Review, we discuss our current understanding of the diverse roles of innate and adaptive immune cells in hypertension, highlighting key findings from human and rodent studies. We explore mechanisms by which these immune cells promote hypertensive pathophysiology, shedding light on their multifaceted involvement. In addition, we highlight advances in our understanding of autoimmunity, HIV and immune checkpoints that provide valuable insight into mechanisms of chronic and dysregulated inflammation in hypertension.
Collapse
Affiliation(s)
- Bianca A Nguyen
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Matthew R Alexander
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN, USA
| | - David G Harrison
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN, USA.
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
21
|
Passelli K, Repáraz D, Kinj R, Herrera FG. Strategies for overcoming tumour resistance to immunotherapy: harnessing the power of radiation therapy. Br J Radiol 2024; 97:1378-1390. [PMID: 38833685 PMCID: PMC11256940 DOI: 10.1093/bjr/tqae100] [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: 01/11/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 06/06/2024] Open
Abstract
Immune checkpoint inhibitors (ICI) have revolutionized cancer treatment; yet their efficacy remains variable across patients. This review delves into the intricate interplay of tumour characteristics contributing to resistance against ICI therapy and suggests that combining with radiotherapy holds promise. Radiation, known for its ability to trigger immunogenic cell death and foster an in situ vaccination effect, may counteract these resistance mechanisms, enhancing ICI response and patient outcomes. However, particularly when delivered at high-dose, it may trigger immunosuppressive mechanism and consequent side-effects. Notably, low-dose radiotherapy (LDRT), with its capacity for tumour reprogramming and reduced side effects, offers the potential for widespread application. Preclinical and clinical studies have shown encouraging results in this regard.
Collapse
Affiliation(s)
- Katiuska Passelli
- Centre Hospitalier Universitaire Vaudoise, Service of Radiation Oncology, Department of Oncology, University of Lausanne, AGORA Center for Cancer Research, Swiss Cancer Center Leman, 1012-Lausanne, Switzerland
| | - David Repáraz
- Centre Hospitalier Universitaire Vaudoise, Service of Radiation Oncology, Department of Oncology, University of Lausanne, AGORA Center for Cancer Research, Swiss Cancer Center Leman, 1012-Lausanne, Switzerland
| | - Remy Kinj
- Centre Hospitalier Universitaire Vaudoise, Service of Radiation Oncology, Department of Oncology, University of Lausanne, 1012-Lausanne, Switzerland
| | - Fernanda G Herrera
- Centre Hospitalier Universitaire Vaudois, Service of Radiation Oncology and Service of Immuno-oncology, Department of Oncology, University of Lausanne, Ludwig Institute for Cancer Research, Agora Center for Cancer Research, Swiss Cancer Center Leman, 1012-Lausanne, Switzerland
| |
Collapse
|
22
|
Cheng W, Kang K, Zhao A, Wu Y. Dual blockade immunotherapy targeting PD-1/PD-L1 and CTLA-4 in lung cancer. J Hematol Oncol 2024; 17:54. [PMID: 39068460 PMCID: PMC11283714 DOI: 10.1186/s13045-024-01581-2] [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: 06/06/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
Cancer immunotherapies, represented by immune checkpoint inhibitors (ICIs), have reshaped the treatment paradigm for both advanced non-small cell lung cancer and small cell lung cancer. Programmed death receptor-1/programmed death receptor ligand-1 (PD-1/PD-L1) and cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) are some of the most common and promising targets in ICIs. Compared to ICI monotherapy, which occasionally demonstrates treatment resistance and limited efficacy, the dual blockade immunotherapy targeting PD-1/PD-L1 and CTLA-4 operates at different stages of T cell activation with synergistically enhancing immune responses against cancer cells. This emerging dual therapy heralds a new direction for cancer immunotherapy, which, however, may increase the risk of drug-related adverse reactions while improving efficacy. Previous clinical trials have explored combination therapy strategy of anti-PD-1/PD-L1 and anti-CTLA-4 agents in lung cancer, yet its efficacy remains to be unclear with the inevitable incidence of immune-related adverse events. The recent advent of bispecific antibodies has made this sort of dual targeting more feasible, aiming to alleviate toxicity without compromising efficacy. Thus, this review highlights the role of dual blockade immunotherapy targeting PD-1/PD-L1 and CTLA-4 in treating lung cancer, and further elucidates its pre-clinical mechanisms and current advancements in clinical trials. Besides, we also provide novel insights into the potential combinations of dual blockade therapies with other strategies to optimize the future treatment mode for lung cancer.
Collapse
Affiliation(s)
- Weishi Cheng
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Kai Kang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ailin Zhao
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Yijun Wu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
23
|
Zhang X, Greve PF, Minh TTN, Wubbolts R, Demir AY, Zaal EA, Berkers CR, Boes M, Stoorvogel W. Extracellular vesicles from seminal plasma interact with T cells in vitro and drive their differentiation into regulatory T-cells. J Extracell Vesicles 2024; 13:e12457. [PMID: 39007430 PMCID: PMC11247398 DOI: 10.1002/jev2.12457] [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: 08/31/2023] [Revised: 04/04/2024] [Accepted: 05/06/2024] [Indexed: 07/16/2024] Open
Abstract
Seminal plasma induces immune tolerance towards paternal allogenic antigens within the female reproductive tract and during foetal development. Recent evidence suggests a role for extracellular vesicles in seminal plasma (spEVs). We isolated spEVs from seminal plasma that was donated by vasectomized men, thereby excluding any contributions from the testis or epididymis. Previous analysis demonstrated that such isolated spEVs originate mainly from the prostate. Here we observed that when isolated fluorescently labelled spEVs were mixed with peripheral blood mononuclear cells, they were endocytosed predominantly by monocytes, and to a lesser extent also by T-cells. In a mixed lymphocyte reaction, T-cell proliferation was inhibited by spEVs. A direct effect of spEVs on T-cells was demonstrated when isolated T cells were activated by anti-CD3/CD28 coated beads. Again, spEVs interfered with T cell proliferation, as well as with the expression of CD25 and the release of IFN-γ, TNF, and IL-2. Moreover, spEVs stimulated the expression of Foxp3 and IL-10 by CD4+CD25+CD127- T cells, indicating differentiation into regulatory T-cells (Tregs). Prior treatment of spEVs with proteinase K revoked their effects on T-cells, indicating a requirement for surface-exposed spEV proteins. The adenosine A2A receptor-specific antagonist CPI-444 also reduced effects of spEVs on T-cells, consistent with the notion that the development of Tregs and their immune suppressive functions are under the influence of adenosine-A2A receptor signalling. We found that adenosine is highly enriched in spEVs and propose that spEVs are targeted to and endocytosed by T-cells, after which they may release their adenosine content into the lumen of endosomes, thus allowing endosome-localized A2A receptor signalling in spEVs targeted T-cells. Collectively, these data support the idea that spEVs can prime T cells directly for differentiation into Tregs.
Collapse
Affiliation(s)
- Xiaogang Zhang
- Department of Biomolecular Health SciencesFaculty of Veterinary ScienceUtrecht UniversityUtrechtThe Netherlands
| | - Patrick F. Greve
- Department of Pediatrics and Center for Translational ImmunologyUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Thi Tran Ngoc Minh
- Department of Biomolecular Health SciencesFaculty of Veterinary ScienceUtrecht UniversityUtrechtThe Netherlands
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
| | - Richard Wubbolts
- Department of Biomolecular Health SciencesFaculty of Veterinary ScienceUtrecht UniversityUtrechtThe Netherlands
| | - Ayşe Y. Demir
- Department of Clinical Chemistry and HematologyMeander Medical CentreAmersfoortThe Netherlands
| | - Esther A. Zaal
- Department of Biomolecular Health SciencesFaculty of Veterinary ScienceUtrecht UniversityUtrechtThe Netherlands
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
| | - Celia R. Berkers
- Department of Biomolecular Health SciencesFaculty of Veterinary ScienceUtrecht UniversityUtrechtThe Netherlands
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
| | - Marianne Boes
- Department of Pediatrics and Center for Translational ImmunologyUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Willem Stoorvogel
- Department of Biomolecular Health SciencesFaculty of Veterinary ScienceUtrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
24
|
Pourbagheri-Sigaroodi A, Momeny M, Rezaei N, Fallah F, Bashash D. Immune landscape of hepatocellular carcinoma: From dysregulation of the immune responses to the potential immunotherapies. Cell Biochem Funct 2024; 42:e4098. [PMID: 39034646 DOI: 10.1002/cbf.4098] [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: 05/25/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/23/2024]
Abstract
Hepatocellular carcinoma (HCC) presents a considerable global health burden due to its late diagnosis and high morbidity. The liver's specific anatomical and physiological features expose it to various antigens, requiring precise immune regulation. To the best of our knowledge, this is the first time that a comprehensive overview of the interactions between the immune system and gut microbiota in the development of HCC, as well as the relevant therapeutic approaches are discussed. Dysregulation of immune compartments within the liver microenvironment drives HCC pathogenesis, characterized by elevated regulatory cells such as regulatory T cells (Tregs), myeloid-derived suppressor cells, and M2 macrophages as well as suppressive molecules, alongside reduced number of effector cells like T cells, natural killer cells, and M1 macrophages. Dysbiosis of gut microbiota also contributes to HCC by disrupting intestinal barrier integrity and triggering overactivated immune responses. Immunotherapy approaches, particularly immune checkpoint inhibitors, have exhibited promise in HCC management, yet adoptive cell therapy and cancer vaccination research are in the early steps with relatively less favorable outcomes. Further understanding of immune dysregulation, gut microbiota involvement, and therapeutic combination strategies are essential for advancing precision immunotherapy in HCC.
Collapse
Affiliation(s)
- Atieh Pourbagheri-Sigaroodi
- Pediatric Infections Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Momeny
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Fallah
- Pediatric Infections Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Huang D, Jiao X, Huang S, Liu J, Si H, Qi D, Pei X, Lu D, Wang Y, Li Z. Analysis of the heterogeneity and complexity of murine extraorbital lacrimal gland via single-cell RNA sequencing. Ocul Surf 2024; 34:60-95. [PMID: 38945476 DOI: 10.1016/j.jtos.2024.06.005] [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: 08/26/2022] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024]
Abstract
PURPOSE The lacrimal gland is essential for maintaining ocular surface health and avoiding external damage by secreting an aqueous layer of the tear film. However, a healthy lacrimal gland's inventory of cell types and heterogeneity remains understudied. METHODS Here, 10X Genome-based single-cell RNA sequencing was used to generate an unbiased classification of cellular diversity in the extraorbital lacrimal gland (ELG) of C57BL/6J mice. From 43,850 high-quality cells, we produced an atlas of cell heterogeneity and defined cell types using classic marker genes. The possible functions of these cells were analyzed through bioinformatics analysis. Additionally, the CellChat was employed for a preliminary analysis of the cell-cell communication network in the ELG. RESULTS Over 37 subclasses of cells were identified, including seven types of glandular epithelial cells, three types of fibroblasts, ten types of myeloid-derived immune cells, at least eleven types of lymphoid-derived immune cells, and five types of vascular-associated cell subsets. The cell-cell communication network analysis revealed that fibroblasts and immune cells play a pivotal role in the dense intercellular communication network within the mouse ELG. CONCLUSIONS This study provides a comprehensive transcriptome atlas and related database of the mouse ELG.
Collapse
Affiliation(s)
- Duliurui Huang
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Xinwei Jiao
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Shenzhen Huang
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Jiangman Liu
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Hongli Si
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Di Qi
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Xiaoting Pei
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Dingli Lu
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Yimian Wang
- Division of Medicine, Faculty of Medical Sciences, University College London, Gower Street, London, WC1E 6BT, UK
| | - Zhijie Li
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China; Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China.
| |
Collapse
|
26
|
Ruocco MR, Gisonna A, Acampora V, D’Agostino A, Carrese B, Santoro J, Venuta A, Nasso R, Rocco N, Russo D, Cavaliere A, Altobelli GG, Masone S, Avagliano A, Arcucci A, Fiume G. Guardians and Mediators of Metastasis: Exploring T Lymphocytes, Myeloid-Derived Suppressor Cells, and Tumor-Associated Macrophages in the Breast Cancer Microenvironment. Int J Mol Sci 2024; 25:6224. [PMID: 38892411 PMCID: PMC11172575 DOI: 10.3390/ijms25116224] [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: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Breast cancers (BCs) are solid tumors composed of heterogeneous tissues consisting of cancer cells and an ever-changing tumor microenvironment (TME). The TME includes, among other non-cancer cell types, immune cells influencing the immune context of cancer tissues. In particular, the cross talk of immune cells and their interactions with cancer cells dramatically influence BC dissemination, immunoediting, and the outcomes of cancer therapies. Tumor-infiltrating lymphocytes (TILs), tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs) represent prominent immune cell populations of breast TMEs, and they have important roles in cancer immunoescape and dissemination. Therefore, in this article we review the features of TILs, TAMs, and MDSCs in BCs. Moreover, we highlight the mechanisms by which these immune cells remodel the immune TME and lead to breast cancer metastasis.
Collapse
Affiliation(s)
- Maria Rosaria Ruocco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (M.R.R.); (A.G.)
| | - Armando Gisonna
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (M.R.R.); (A.G.)
| | - Vittoria Acampora
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Anna D’Agostino
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy; (A.D.); (B.C.); (J.S.)
| | - Barbara Carrese
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy; (A.D.); (B.C.); (J.S.)
| | - Jessie Santoro
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy; (A.D.); (B.C.); (J.S.)
| | - Alessandro Venuta
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Rosarita Nasso
- Department of Movement Sciences and Wellness, University of Naples “Parthenope”, 80133 Naples, Italy;
| | - Nicola Rocco
- Department of Advanced Biomedical Science, University of Naples Federico II, 80131 Naples, Italy; (N.R.); (D.R.); (G.G.A.)
| | - Daniela Russo
- Department of Advanced Biomedical Science, University of Naples Federico II, 80131 Naples, Italy; (N.R.); (D.R.); (G.G.A.)
| | | | - Giovanna Giuseppina Altobelli
- Department of Advanced Biomedical Science, University of Naples Federico II, 80131 Naples, Italy; (N.R.); (D.R.); (G.G.A.)
| | - Stefania Masone
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy;
| | - Angelica Avagliano
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Alessandro Arcucci
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Giuseppe Fiume
- Department of Experimental and Clinical Medicine, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy;
| |
Collapse
|
27
|
M Yusoff NNF, Ahmad S, Wan Abdul Rahman WF, Mohamud R, C Boer J, Plebanski M, Abdullah B, Chen X, Tengku Din TADAA. CD4+ Foxp3+ Regulatory T-cells in Modulating Inflammatory Microenvironment in Chronic Rhinosinusitis with Nasal Polyps: Progress and Future Prospect. Cytokine 2024; 178:156557. [PMID: 38452440 DOI: 10.1016/j.cyto.2024.156557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/26/2024] [Accepted: 02/20/2024] [Indexed: 03/09/2024]
Abstract
Chronic rhinosinusitis with nasal polyps (CRSwNP) is a subtype of chronic rhinosinusitis (CRS) characterized by the presence of nasal polyps (NP) in the paranasal mucosa. Despite the complex etiology, NP is believed to result from chronic inflammation. The long-term aftermath of the type 2 response is responsible for symptoms seen in NP patients, i.e. rhinorrhea, hyposmia, and nasal obstruction. Immune cellular tolerogenic mechanisms, particularly CD4 + Foxp3 + regulatory T cells (Tregs), are crucial to curtail inflammatory responses. Current evidence suggests impaired Treg activity is the main reason underlying the compromise of self-tolerance, contributing to the onset of CRSwNP. There is compelling evidence that tumor necrosis factor 2 (TNFR2) is preferentially expressed by Tregs, and TNFR2 is able to identify the most potent suppressive subset of Tregs. Tumor necrosis factor (TNF)-TNFR2 interaction plays a decisive role in the activation and expansion of Tregs. This review summarizes current understanding of Tregs biology, focusing on the discussion of the recent advances in the study of TNF-TNFR2 axis in the upregulation of Treg function as a negative feedback mechanism in the control of chronic inflammation. The role of dysregulation of Tregs in the immunopathogenesis of CRSwNP will be analyzed. The future perspective on the harnessing Tregs-mediated self-tolerant mechanism in the management of CRSwNP will be introduced.
Collapse
Affiliation(s)
- Nur Najwa Farahin M Yusoff
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Suhana Ahmad
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | | | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Jennifer C Boer
- Translational Immunology and Nanotechnology Unit, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia
| | - Magdalena Plebanski
- Translational Immunology and Nanotechnology Unit, School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia
| | - Baharudin Abdullah
- Department of Otorhinolaryngology-Head and Neck Surgery, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia.
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | | |
Collapse
|
28
|
Magni S, Sawlekar R, Capelle CM, Tslaf V, Baron A, Zeng N, Mombaerts L, Yue Z, Yuan Y, Hefeng FQ, Gonçalves J. Inferring upstream regulatory genes of FOXP3 in human regulatory T cells from time-series transcriptomic data. NPJ Syst Biol Appl 2024; 10:59. [PMID: 38811598 PMCID: PMC11137136 DOI: 10.1038/s41540-024-00387-9] [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: 02/08/2024] [Accepted: 05/10/2024] [Indexed: 05/31/2024] Open
Abstract
The discovery of upstream regulatory genes of a gene of interest still remains challenging. Here we applied a scalable computational method to unbiasedly predict candidate regulatory genes of critical transcription factors by searching the whole genome. We illustrated our approach with a case study on the master regulator FOXP3 of human primary regulatory T cells (Tregs). While target genes of FOXP3 have been identified, its upstream regulatory machinery still remains elusive. Our methodology selected five top-ranked candidates that were tested via proof-of-concept experiments. Following knockdown, three out of five candidates showed significant effects on the mRNA expression of FOXP3 across multiple donors. This provides insights into the regulatory mechanisms modulating FOXP3 transcriptional expression in Tregs. Overall, at the genome level this represents a high level of accuracy in predicting upstream regulatory genes of key genes of interest.
Collapse
Affiliation(s)
- Stefano Magni
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Rucha Sawlekar
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
- Robotics and Artificial Intelligence, Department of Computer Science, Electrical and Space Engineering, Luleå University of Technology, Luleå, Sweden
| | - Christophe M Capelle
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-Sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Vera Tslaf
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-Sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Alexandre Baron
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-Sur-Alzette, Luxembourg
| | - Ni Zeng
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-Sur-Alzette, Luxembourg
| | - Laurent Mombaerts
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Zuogong Yue
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Ye Yuan
- School of Artificial Intelligence and Automation, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Q Hefeng
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-Sur-Alzette, Luxembourg.
| | - Jorge Gonçalves
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg.
- Department of Plant Sciences, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
29
|
Choi SH, Mani M, Kim J, Cho WJ, Martin TFJ, Kim JH, Chu HS, Jeong WJ, Won YW, Lee BJ, Ahn B, Kim J, Jeon DY, Park JW. DRG2 is required for surface localization of PD-L1 and the efficacy of anti-PD-1 therapy. Cell Death Discov 2024; 10:260. [PMID: 38802348 PMCID: PMC11130180 DOI: 10.1038/s41420-024-02027-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/10/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024] Open
Abstract
More than half of tumor patients with high PD-L1 expression do not respond to anti-PD-1/PD-L1 therapy, and the underlying mechanisms are yet to be clarified. Here we show that developmentally regulated GTP-binding protein 2 (DRG2) is required for response of PD-L1-expressing tumors to anti-PD-1 therapy. DRG2 depletion enhanced IFN-γ signaling and increased the PD-L1 level in melanoma cells. However, it inhibited recycling of endosomal PD-L1 and reduced surface PD-L1 levels, which led to defects in interaction with PD-1. Anti-PD-1 did not expand effector-like T cells within DRG2-depleted tumors and failed to improve the survival of DRG2-depleted tumor-bearing mice. Cohort analysis revealed that patients bearing melanoma with low DRG2 protein levels were resistant to anti-PD-1 therapy. These findings identify DRG2 as a key regulator of recycling of endosomal PD-L1 and response to anti-PD-1 therapy and provide insights into how to increase the correlation between PD-L1 expression and response to anti-PD-1 therapy.
Collapse
Affiliation(s)
- Seong Hee Choi
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea
- RopheLBio, B102, Seoul Forest M Tower, Seoul, Korea
| | - Muralidharan Mani
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Jeonghwan Kim
- School of System Biomedical Science, Soongsil University, Seoul, Korea
| | - Wha Ja Cho
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea
| | - Thomas F J Martin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Jee Hyun Kim
- RopheLBio, B102, Seoul Forest M Tower, Seoul, Korea
| | - Hun Su Chu
- RopheLBio, B102, Seoul Forest M Tower, Seoul, Korea
| | | | - Young-Wook Won
- RopheLBio, B102, Seoul Forest M Tower, Seoul, Korea
- Department of Biomedical Engineering, University of North Texas, Denton, TX, USA
| | - Byung Ju Lee
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
| | - Byungyong Ahn
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
- Department of Food Science and Nutrition, University of Ulsan, Ulsan, Korea
| | - Junil Kim
- School of System Biomedical Science, Soongsil University, Seoul, Korea.
| | - Do Yong Jeon
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea.
| | - Jeong Woo Park
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea.
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea.
| |
Collapse
|
30
|
Saleh QW, Mohammadnejad A, Tepel M. FOXP3 splice variant expression in males and females in healthy populations and in kidney transplant recipients. Sci Rep 2024; 14:12112. [PMID: 38802392 PMCID: PMC11130272 DOI: 10.1038/s41598-024-62149-1] [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: 11/16/2023] [Accepted: 05/14/2024] [Indexed: 05/29/2024] Open
Abstract
The forkhead box P3 (FOXP3) transcript is essential for tolerance of alloantigens. Here, we describe the expression of FOXP3 mRNA variants in healthy females and males, and in kidney transplant recipients (KTR). We measured FOXP3 in peripheral blood mononuclear cells from healthy kidney donors (N = 101), and in blood from KTRs (N = 248) before and after transplantation. FOXP3 was measured with quantitative polymerase chain reaction, and differentiated between pre-mature mRNA FOXP3, Total mature FOXP3, FOXP3 in which exon two is spliced, and full length FOXP3. We found similar levels of FOXP3 in healthy female and male kidney donors. We confirmed this result in a publicly available cohort (N = 33) of healthy individuals (GSE97475). Homogenously, female and male KTR FOXP3 levels were similar pre-transplantation, one day post-transplantation and 29 days post-transplantation. This may suggest that kidney transplantation and related immunosuppressive treatments do not influence FOXP3 expression differently in females and males. Finally, fold difference analysis revealed that KTRs express lower levels of mature FOXP3 and higher levels of pre-mature FOXP3 mRNA pre-transplant compared to healthy individuals. This finding may suggest higher pre-mRNA synthesis, lower pre-mRNA degradation, lower spliceosome efficiency or higher degradation of mature FOXP3 mRNA in kidney transplant candidates.
Collapse
Affiliation(s)
- Qais W Saleh
- Department of Nephrology, Odense University Hospital, J. B. Winsløws Vej 4, 5000, Odense, Denmark
- Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, J.B. Winsløws Vej 21.3, 5000, Odense C, Denmark
| | - Afsaneh Mohammadnejad
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, J.B. Winsløws Vej 9 B, 5000, Odense C, Denmark
| | - Martin Tepel
- Department of Nephrology, Odense University Hospital, J. B. Winsløws Vej 4, 5000, Odense, Denmark.
- Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, J.B. Winsløws Vej 21.3, 5000, Odense C, Denmark.
| |
Collapse
|
31
|
Fischer F, Ermer MR, Howanski J, Yin Z, Bauer M, Wagner M, Fink B, Zenclussen AC, Schumacher A. Single and mixture effects of bisphenol A and benzophenone-3 on in vitro T helper cell differentiation. Chem Biol Interact 2024; 395:111011. [PMID: 38653352 DOI: 10.1016/j.cbi.2024.111011] [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: 01/23/2024] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
Immune homeostasis is key to guarantee that the immune system can elicit effector functions against pathogens and at the same time raise tolerance towards other antigens. A disturbance of this delicate balance may underlie or at least trigger pathologies. Endocrine disrupting chemicals (EDCs) are increasingly recognized as risk factors for immune dysregulation. However, the immunotoxic potential of specific EDCs and their mixtures is still poorly understood. Thus, we aimed to investigate the effect of bisphenol A (BPA) and benzophenone-3 (BP-3), alone and in combination, on in vitro differentiation of T helper (TH)17 cells and regulatory T (Treg) cells. Naïve T cells were isolated from mouse lymphoid tissues and differentiated into the respective TH population in the presence of 0.001-10 μM BP-3 and/or 0.01-100 μM BPA. Cell viability, proliferation and the expression of TH lineage specific transcription factors and cytokines was measured by flow cytometry and CBA/ELISA. Moreover, the transcription of hormone receptors as direct targets of EDCs was quantified by RT-PCR. We found that the highest BPA concentration adversely affected TH cell viability and proliferation. Moreover, the general differentiation potential of both TH populations was not altered in the presence of both EDCs. However, EDC exposure modulated the emergence of TH17 and Treg cell intermediate states. While BPA and BP-3 promoted the development of TH1-like TH17 cells under TH17-differentiating conditions, TH2-like Treg cells occurred under Treg polarization. Interestingly, differential effects could be observed in mixtures of the two tested compounds compared with the individual compounds. Notably, estrogen receptor β expression was decreased under TH17-differentiating conditions in the presence of BPA and BP-3 as mixture. In conclusion, our study provides solid evidence for both, the immune disruptive potential and the existence of cumulative effects of real nature EDC mixtures on T cell in vitro differentiation.
Collapse
Affiliation(s)
- Florence Fischer
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH - UFZ, Permoserstraße 15, 04318, Leipzig, Germany; Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, Leipzig University, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany
| | - Miriam Rebecca Ermer
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH - UFZ, Permoserstraße 15, 04318, Leipzig, Germany; Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, Leipzig University, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany
| | - Julia Howanski
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH - UFZ, Permoserstraße 15, 04318, Leipzig, Germany; Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, Leipzig University, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany
| | - Ziran Yin
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH - UFZ, Permoserstraße 15, 04318, Leipzig, Germany; Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, Leipzig University, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany
| | - Mario Bauer
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH - UFZ, Permoserstraße 15, 04318, Leipzig, Germany
| | - Marita Wagner
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH - UFZ, Permoserstraße 15, 04318, Leipzig, Germany
| | - Beate Fink
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH - UFZ, Permoserstraße 15, 04318, Leipzig, Germany
| | - Ana C Zenclussen
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH - UFZ, Permoserstraße 15, 04318, Leipzig, Germany; Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, Leipzig University, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany
| | - Anne Schumacher
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research GmbH - UFZ, Permoserstraße 15, 04318, Leipzig, Germany; Perinatal Immunology, Saxonian Incubator for Clinical Translation (SIKT), Medical Faculty, Leipzig University, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany.
| |
Collapse
|
32
|
Glass DR, Mayer-Blackwell K, Ramchurren N, Parks KR, Duran GE, Wright AK, Bastidas Torres AN, Islas L, Kim YH, Fling SP, Khodadoust MS, Newell EW. Multi-omic profiling reveals the endogenous and neoplastic responses to immunotherapies in cutaneous T cell lymphoma. Cell Rep Med 2024; 5:101527. [PMID: 38670099 PMCID: PMC11148639 DOI: 10.1016/j.xcrm.2024.101527] [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: 09/14/2023] [Revised: 02/17/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024]
Abstract
Cutaneous T cell lymphomas (CTCLs) are skin cancers with poor survival rates and limited treatments. While immunotherapies have shown some efficacy, the immunological consequences of administering immune-activating agents to CTCL patients have not been systematically characterized. We apply a suite of high-dimensional technologies to investigate the local, cellular, and systemic responses in CTCL patients receiving either mono- or combination anti-PD-1 plus interferon-gamma (IFN-γ) therapy. Neoplastic T cells display no evidence of activation after immunotherapy. IFN-γ induces muted endogenous immunological responses, while anti-PD-1 elicits broader changes, including increased abundance of CLA+CD39+ T cells. We develop an unbiased multi-omic profiling approach enabling discovery of immune modules stratifying patients. We identify an enrichment of activated regulatory CLA+CD39+ T cells in non-responders and activated cytotoxic CLA+CD39+ T cells in leukemic patients. Our results provide insights into the effects of immunotherapy in CTCL patients and a generalizable framework for multi-omic analysis of clinical trials.
Collapse
Affiliation(s)
- David R Glass
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA.
| | - Koshlan Mayer-Blackwell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Nirasha Ramchurren
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Cancer Immunotherapy Trials Network, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - K Rachael Parks
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - George E Duran
- Division of Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Anna K Wright
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Cancer Immunotherapy Trials Network, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | | | - Laura Islas
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Youn H Kim
- Division of Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Steven P Fling
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Cancer Immunotherapy Trials Network, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Michael S Khodadoust
- Division of Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Evan W Newell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA.
| |
Collapse
|
33
|
Zhdanov DD, Gladilina YA, Blinova VG, Abramova AA, Shishparenok AN, Eliseeva DD. Induction of FoxP3 Pre-mRNA Alternative Splicing to Enhance the Suppressive Activity of Regulatory T Cells from Amyotrophic Lateral Sclerosis Patients. Biomedicines 2024; 12:1022. [PMID: 38790984 PMCID: PMC11117958 DOI: 10.3390/biomedicines12051022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/15/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
Forkhead box protein 3 (FoxP3) is a key transcription factor responsible for the development, maturation, and function of regulatory T cells (Tregs). The FoxP3 pre-mRNA is subject to alternative splicing, resulting in the translation of multiple splice variants. We have shown that Tregs from patients with amyotrophic lateral sclerosis (ALS) have reduced expression of full-length (FL) FoxP3, while other truncated splice variants are expressed predominantly. A correlation was observed between the reduced number of Tregs in the peripheral blood of ALS patients, reduced total FoxP3 mRNA, and reduced mRNA of its FL splice variant. Induction of FL FoxP3 was achieved using splice-switching oligonucleotides capable of base pairing with FoxP3 pre-mRNA and selectively modulating the inclusion of exons 2 and 7 in the mature mRNA. Selective expression of FL FoxP3 resulted in the induction of CD127low, CD152, and Helios-positive cells, while the cell markers CD4 and CD25 were not altered. Such Tregs had an increased proliferative activity and a higher frequency of cell divisions per day. The increased suppressive activity of Tregs with the induced FL FoxP3 splice variant was associated with the increased synthesis of the pro-apoptotic granzymes A and B, and perforin, IL-10, and IL-35, which are responsible for contact-independent suppression, and with the increased ability to suppress telomerase in target cells. The upregulation of Treg suppressive and proliferative activity using splice-switching oligonucleotides to induce the predominant expression of the FoxP3 FL variant is a promising approach for regenerative cell therapy in Treg-associated diseases.
Collapse
Affiliation(s)
- Dmitry D. Zhdanov
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (Y.A.G.); (V.G.B.); (A.A.A.); (A.N.S.)
- Department of Biochemistry, Peoples’ Friendship University of Russia named after Patrice Lumumba (RUDN University), Miklukho-Maklaya St. 6, 117198 Moscow, Russia
| | - Yulia A. Gladilina
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (Y.A.G.); (V.G.B.); (A.A.A.); (A.N.S.)
| | - Varvara G. Blinova
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (Y.A.G.); (V.G.B.); (A.A.A.); (A.N.S.)
| | - Anna A. Abramova
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (Y.A.G.); (V.G.B.); (A.A.A.); (A.N.S.)
- Research Center of Neurology, Volokolamskoe Shosse, 80, 125367 Moscow, Russia;
| | - Anastasia N. Shishparenok
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (Y.A.G.); (V.G.B.); (A.A.A.); (A.N.S.)
| | - Daria D. Eliseeva
- Research Center of Neurology, Volokolamskoe Shosse, 80, 125367 Moscow, Russia;
| |
Collapse
|
34
|
Schafer S, Chen K, Ma L. Crosstalking with Dendritic Cells: A Path to Engineer Advanced T Cell Immunotherapy. FRONTIERS IN SYSTEMS BIOLOGY 2024; 4:1372995. [PMID: 38911455 PMCID: PMC11192543 DOI: 10.3389/fsysb.2024.1372995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Crosstalk between dendritic cells (DCs) and T cells plays a crucial role in modulating immune responses in natural and pathological conditions. DC-T cell crosstalk is achieved through contact-dependent (i.e., immunological synapse) and contact-independent mechanisms (i.e., cytokines). Activated DCs upregulate co-stimulatory signals and secrete proinflammatory cytokines to orchestrate T cell activation and differentiation. Conversely, activated T helper cells "license" DCs towards maturation, while regulatory T cells (Tregs) silence DCs to elicit tolerogenic immunity. Strategies to efficiently modulate the DC-T cell crosstalk can be harnessed to promote immune activation for cancer immunotherapy or immune tolerance for the treatment of autoimmune diseases. Here, we review the natural crosstalk mechanisms between DC and T cells. We highlight bioengineering approaches to modulate DC-T cell crosstalk, including conventional vaccines, synthetic vaccines, and DC-mimics, and key seminal studies leveraging these approaches to steer immune response for the treatment of cancer and autoimmune diseases.
Collapse
Affiliation(s)
- Sogand Schafer
- Center for Craniofacial Innovation, Children’s Hospital of Philadelphia Research Institute, Children’s Hospital of Philadelphia, PA 19104, USA
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Children’s Hospital of Philadelphia, PA 19104, USA
| | - Kaige Chen
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Leyuan Ma
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, US
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
35
|
Foyle KL, Robertson SA. Gamma delta (γδ) T cells in the female reproductive tract: active participants or indifferent bystanders in reproductive success? DISCOVERY IMMUNOLOGY 2024; 3:kyae004. [PMID: 38863792 PMCID: PMC11165432 DOI: 10.1093/discim/kyae004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 03/27/2024] [Accepted: 04/25/2024] [Indexed: 06/13/2024]
Abstract
The female reproductive tract accommodates and balances the unique immunological challenges of protection from sexually transmitted pathogens and tolerance of the fetus and placenta in pregnancy. Leukocytes in the female reproductive tract actively engage in extensive maternal adaptations that are imperative for embryo implantation, placental development, and fetal growth support. γδ T cells are abundant at many mucosal sites in the body, where they provide protection against pathogens and cancer, and have roles in tissue renewal and homeostasis. In this review, we summarize studies in humans and rodents showing that γδ T cells are prevalent in the female reproductive tract and fluctuate in response to hormone changes across the reproductive cycle. Emerging evidence points to a link between changes in their abundance and molecular repertoire in the uterus and pregnancy disorders including recurrent miscarriage and preterm birth. However, defining the precise functional role of female reproductive tract γδ T cells and understanding their physiological significance in reproduction and pregnancy have remained elusive. Here, we critically analyze whether reproductive tract γδ T cells could be active participants in reproductive events-or whether their principal function is immune defense, in which case they may compromise pregnancy success unless adequately regulated.
Collapse
Affiliation(s)
- Kerrie L Foyle
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Sarah A Robertson
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
36
|
Innuan P, Sirikul C, Anukul N, Rolin G, Dechsupa N, Kantapan J. Identifying transcriptomic profiles of iron-quercetin complex treated peripheral blood mononuclear cells from healthy volunteers and diabetic patients. Sci Rep 2024; 14:9441. [PMID: 38658734 PMCID: PMC11043337 DOI: 10.1038/s41598-024-60197-1] [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: 11/03/2023] [Accepted: 04/19/2024] [Indexed: 04/26/2024] Open
Abstract
Peripheral blood is an alternative source of stem/progenitor cells for regenerative medicine owing to its ease of retrieval and blood bank storage. Previous in vitro studies indicated that the conditioned medium derived from peripheral blood mononuclear cells (PBMCs) treated with the iron-quercetin complex (IronQ) contains potent angiogenesis and wound-healing properties. This study aims to unveil the intricate regulatory mechanisms governing the effects of IronQ on the transcriptome profiles of human PBMCs from healthy volunteers and those with diabetes mellitus (DM) using RNA sequencing analysis. Our findings revealed 3741 and 2204 differentially expressed genes (DEGs) when treating healthy and DM PBMCs with IronQ, respectively. Functional enrichment analyses underscored the biological processes shared by the DEGs in both conditions, including inflammatory responses, cell migration, cellular stress responses, and angiogenesis. A comprehensive exploration of these molecular alterations exposed a network of 20 hub genes essential in response to stimuli, cell migration, immune processes, and the mitogen-activated protein kinase (MAPK) pathway. The activation of these pathways enabled PBMCs to potentiate angiogenesis and tissue repair. Corroborating this, quantitative real-time polymerase chain reaction (qRT-PCR) and cell phenotyping confirmed the upregulation of candidate genes associated with anti-inflammatory, pro-angiogenesis, and tissue repair processes in IronQ-treated PBMCs. In summary, combining IronQ and PBMCs brings about substantial shifts in gene expression profiles and activates pathways that are crucial for tissue repair and immune response, which is promising for the enhancement of the therapeutic potential of PBMCs, especially in diabetic wound healing.
Collapse
Affiliation(s)
- Phattarawadee Innuan
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Chonticha Sirikul
- Division of Transfusion Science, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nampeung Anukul
- Division of Transfusion Science, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Gwenaël Rolin
- INSERM CIC-1431, CHU Besançon, 25000, Besançon, France
| | - Nathupakorn Dechsupa
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Jiraporn Kantapan
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
37
|
Koch KC, Jadon N, Thesmar I, Tew GN, Minter LM. Combating bone marrow failure with polymer materials. Front Immunol 2024; 15:1396486. [PMID: 38694497 PMCID: PMC11061490 DOI: 10.3389/fimmu.2024.1396486] [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: 03/05/2024] [Accepted: 04/03/2024] [Indexed: 05/04/2024] Open
Abstract
Bone marrow failure (BMF) has become one of the most studied autoimmune disorders, particularly due to its prevalence both as an inherited disease, but also as a result of chemotherapies. BMF is associated with severe symptoms such as bleeding episodes and susceptibility to infections, and often has underlying characteristics, such as anemia, thrombocytopenia, and neutropenia. The current treatment landscape for BMF requires stem cell transplantation or chemotherapies to induce immune suppression. However, there is limited donor cell availability or dose related toxicity associated with these treatments. Optimizing these treatments has become a necessity. Polymer-based materials have become increasingly popular, as current research efforts are focused on synthesizing novel cell matrices for stem cell expansion to solve limited donor cell availability, as well as applying polymer delivery vehicles to intracellularly deliver cargo that can aid in immunosuppression. Here, we discuss the importance and impact of polymer materials to enhance therapeutics in the context of BMF.
Collapse
Affiliation(s)
- Kayla C. Koch
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA, United States
| | - Nidhi Jadon
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| | - Iris Thesmar
- University of Massachusetts Amherst, Amherst, MA, United States
| | - Gregory N. Tew
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA, United States
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
- University of Massachusetts Amherst, Amherst, MA, United States
| | - Lisa M. Minter
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
- University of Massachusetts Amherst, Amherst, MA, United States
| |
Collapse
|
38
|
Saleh QW, Mohammadnejad A, Tepel M. FOXP3 full length splice variant is associated with kidney allograft tolerance. Front Immunol 2024; 15:1389105. [PMID: 38660296 PMCID: PMC11040551 DOI: 10.3389/fimmu.2024.1389105] [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: 02/20/2024] [Accepted: 03/29/2024] [Indexed: 04/26/2024] Open
Abstract
Background Progressive decline of allograft function leads to premature graft loss. Forkhead box P3 (FOXP3), a characteristic gene of T-regulatory cells, is known to be essential for auto-antigen tolerance. We assessed the hypothesis that low FOXP3 mRNA splice variant levels in peripheral blood cells early after transplantation are associated with progressive allograft injury. Methods Blood samples were prospectively collected from 333 incident kidney transplant recipients on the first and 29th postoperative day. We used quantitative polymerase chain reaction to determine transcripts of 3 isotypes of FOXP3 splice variants, including pre-mature FOXP3 and full length FOXP3 (FOXP3fl). We investigated the association between FOXP3 splice variant levels and the declines in estimated glomerular filtration rate (eGFR) of more than 5ml/min/1.73m2 within the first-year post-transplant using logistic regression. Results We observed lower FOXP3fl levels in recipients with declining eGFR (N = 132) than in recipients with stable eGFR (N = 201), (logarithmic value -4.13 [IQR -4.50 to -3.84] vs -4.00 [4.32 to -3.74], p=0.02). In ad hoc analysis pre-transplant FOXP3fl levels were similar in both groups. The association between FOXP3fl and declining eGFR was confirmed by multivariable analysis adjusted for potential confounding factors (Odds Ratio 0.51, 95% confidence interval 0.28 to 0.91: p=0.02). When stratifying FOXP3fl levels into quartiles, recipients with lower day1 FOXP3fl had the highest rate of declining eGFR (p=0.04). Conclusion Low FOXP3fl splice variant levels at the first postoperative day in kidney transplant recipients were associated with severe decline of eGFR, a well-known surrogate for hard endpoints.
Collapse
Affiliation(s)
- Qais W. Saleh
- Department of Nephrology, Odense University Hospital, Odense, Denmark
- Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Afsaneh Mohammadnejad
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Martin Tepel
- Department of Nephrology, Odense University Hospital, Odense, Denmark
- Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
39
|
Xiong Y, Ma X, He B, Zhi J, Liu X, Wang P, Zhou Z, Liu D. Multifaceted Effects of Subchronic Exposure to Chlorfenapyr in Mice: Implications from Serum Metabolomics, Hepatic Oxidative Stress, and Intestinal Homeostasis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:7423-7437. [PMID: 38502791 DOI: 10.1021/acs.jafc.3c09682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
As chlorfenapyr is a commonly used insecticide in agriculture, the health risks of subchronic exposure to chlorfenapyr remained unclear. This study aimed to extensively probe the health risks from subchronic exposure to chlorfenapyr at the NOAEL and 10-fold NOAEL dose in mice. Through pathological and biochemical examinations, the body metabolism, hepatic toxicity, and intestinal homeostasis were systematically assessed. After 12 weeks, a 10-fold NOAEL dose of chlorfenapyr resulted in weight reduction, increased daily food intake, and blood lipid abnormalities. Concurrently, this dosage induced hepatotoxicity and amplified oxidative stress in hepatocytes, a finding further supported in HepG2 cells. Moreover, chlorfenapyr resulted in intestinal inflammation, evidenced by increased inflammatory factors (IL-17a, IL-10, IL-1β, IL-6, IL-22), disrupted immune cells (RORγt, Foxp3), and compromised intestinal barriers (ZO-1 and occludin). By contrast, the NOAEL dose presented less toxicity in most evaluations. Serum metabolomic analyses unveiled widespread disruptions in pathways related to hepatotoxicity and intestinal inflammation, including NF-κB signaling, Th cell differentiation, and bile acid metabolism. Microbiomic analysis showed an increase in Lactobacillus, a decrease in Muribaculaceae, and diminished anti-inflammatory microbes, which further propelled the inflammatory response and leaded to intestinal inflammation. These findings revealed the molecular mechanisms underlying chlorfenapyr-induced hepatotoxicity and intestinal inflammation, highlighting the significant role of the gut microbiota.
Collapse
Affiliation(s)
- Yabing Xiong
- Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| | - Xiaoran Ma
- Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| | - Bingying He
- Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| | - Jianwen Zhi
- Department of Proctology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xueke Liu
- Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| | - Peng Wang
- Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| | - Zhiqiang Zhou
- Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| | - Donghui Liu
- Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| |
Collapse
|
40
|
Cardinez C, Hao Y, Kwong K, Davies AR, Downes MB, Roberts NA, Price JD, Hernandez RA, Lovell J, Chand R, Feng ZP, Enders A, Vinuesa CG, Miraghazadeh B, Cook MC. IKK2 controls the inflammatory potential of tissue-resident regulatory T cells in a murine gain of function model. Nat Commun 2024; 15:2345. [PMID: 38528069 PMCID: PMC10963799 DOI: 10.1038/s41467-024-45870-3] [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: 06/13/2023] [Accepted: 02/06/2024] [Indexed: 03/27/2024] Open
Abstract
Loss-of-function mutations have provided crucial insights into the immunoregulatory actions of Foxp3+ regulatory T cells (Tregs). By contrast, we know very little about the consequences of defects that amplify aspects of Treg function or differentiation. Here we show that mice heterozygous for an Ikbkb gain-of-function mutation develop psoriasis. Doubling the gene dose (IkbkbGoF/GoF) results in dactylitis, spondylitis, and characteristic nail changes, which are features of psoriatic arthritis. IkbkbGoF mice exhibit a selective expansion of Foxp3 + CD25+ Tregs of which a subset express IL-17. These modified Tregs are enriched in both inflamed tissues, blood and spleen, and their transfer is sufficient to induce disease without conventional T cells. Single-cell transcriptional and phenotyping analyses of isolated Tregs reveal expansion of non-lymphoid tissue (tissue-resident) Tregs expressing Th17-related genes, Helios, tissue-resident markers including CD103 and CD69, and a prominent NF-κB transcriptome. Thus, IKK2 regulates tissue-resident Treg differentiation, and overactivity drives dose-dependent skin and systemic inflammation.
Collapse
Affiliation(s)
- Chelisa Cardinez
- Centre for Personalised Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Translational Research Unit, The Canberra Hospital, Canberra, ACT, Australia
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Division of Genome Sciences and Cancer, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Yuwei Hao
- Centre for Personalised Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Translational Research Unit, The Canberra Hospital, Canberra, ACT, Australia
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Kristy Kwong
- Centre for Personalised Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Translational Research Unit, The Canberra Hospital, Canberra, ACT, Australia
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ainsley R Davies
- Centre for Personalised Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Translational Research Unit, The Canberra Hospital, Canberra, ACT, Australia
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Morgan B Downes
- Centre for Personalised Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Translational Research Unit, The Canberra Hospital, Canberra, ACT, Australia
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Nadia A Roberts
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Jason D Price
- Division of Genome Sciences and Cancer, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Raquel A Hernandez
- Centre for Personalised Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Translational Research Unit, The Canberra Hospital, Canberra, ACT, Australia
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Jessica Lovell
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Rochna Chand
- Centre for Personalised Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Translational Research Unit, The Canberra Hospital, Canberra, ACT, Australia
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Zhi-Ping Feng
- ANU Bioinformatics Consultancy, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Anselm Enders
- Centre for Personalised Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Carola G Vinuesa
- Centre for Personalised Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Francis Crick Institute, London, UK
| | - Bahar Miraghazadeh
- Centre for Personalised Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Translational Research Unit, The Canberra Hospital, Canberra, ACT, Australia
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Matthew C Cook
- Centre for Personalised Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia.
- Translational Research Unit, The Canberra Hospital, Canberra, ACT, Australia.
- Division of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia.
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
41
|
Zhang Y, Zhang Z, Li H, Chu C, Liang G, Fan N, Wei R, Zhang T, Li L, Wang B, Li X. Increased miR-6132 promotes deep vein thrombosis formation by downregulating FOXP3 expression. Front Cardiovasc Med 2024; 11:1356286. [PMID: 38572308 PMCID: PMC10987872 DOI: 10.3389/fcvm.2024.1356286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/08/2024] [Indexed: 04/05/2024] Open
Abstract
Background Deep vein thrombosis (DVT) is associated with aberrant gene expression that is a common peripheral vascular disease. Here, we aimed to elucidate that the epigenetic modification of forkhead box protein 3 (FOXP3) at the post-transcriptional level, which might be the key trigger leading to the down-regulation of FOXP3 expression in DVT. Methods In order to explore the relationship between microRNAs (miRNAs) and FOXP3, mRNA and microRNA microarray analysis were performed. Dual luciferase reporter assay was used to verify the upstream miRNAs of FOXP3. Quantitative real-time polymerase chain reaction, flow cytometry and Western blot were used to detect the relative expression of miR-6132 and FOXP3. Additionally, DVT models were established to investigate the role of miR-6132 by Murine Doppler Ultrasound and Hematoxylin-Eosin staining. Results Microarray and flow cytometry results showed that the FOXP3 expression was decreased while miR-6132 level was increased substantially in DVT, and there was significant negative correlation between miR-6132 and FOXP3. Moreover, we discovered that overexpressed miR-6132 reduced FOXP3 expression and aggravated DVT formation, while miR-6132 knockdown increased FOXP3 expression and alleviated DVT formation. Dual luciferase reporter assay validated the direct binding of miR-6132 to FOXP3. Conclusion Collectively, our data elucidate a new avenue through which up-regulated miR-6132 contributes to the formation and progression of DVT by inhibiting FOXP3 expression.
Collapse
Affiliation(s)
- Yunhong Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Zhen Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Haoyang Li
- International Business School, Tianjin Foreign Studies University, Tianjin, China
| | - Chu Chu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Gang Liang
- Department of Peripheral Vascular Disease, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Nannan Fan
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Ran Wei
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Tingting Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Lihua Li
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Bin Wang
- Department of Peripheral Vascular Disease, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Xia Li
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| |
Collapse
|
42
|
Cheng Y, Ferdousi F, Foronda BA, Linh TN, Ganbold M, Yada A, Arimura T, Isoda H. A comparative transcriptomics analysis reveals ethylene glycol derivatives of squalene ameliorate excessive lipogenesis and inflammatory response in 3T3-L1 preadipocytes. Heliyon 2024; 10:e26867. [PMID: 38463791 PMCID: PMC10923669 DOI: 10.1016/j.heliyon.2024.e26867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/27/2023] [Accepted: 02/21/2024] [Indexed: 03/12/2024] Open
Abstract
Squalene (SQ) is a natural compound with anti-inflammatory, anti-cancer, and anti-oxidant effects, but due to its low solubility, its biological properties have been greatly underestimated. This study aims to explore the differences in gene expression patterns of four newly synthesized amphipathic ethylene glycol (EG) derivatives of SQ by whole-genome transcriptomics analysis using DNA microarray to examine the mRNA expression profile of adipocytes differentiated from 3T3-L1 cells treated with SQ and its EG derivatives. Enrichment analyses of the transcriptional data showed that compared with SQ, its EG derivatives exerted different, in most cases desirable, biological responses. EG derivatives showed increased enrichment of mitochondrial functions, lipid and glucose metabolism, and inflammatory response. Mono-, di-, and tetra-SQ showed higher enrichment of the cellular component-ribosome. Histological staining showed EG derivatives prevented excessive lipid accumulation. Additionally, mitochondrial transcription factors showed upregulation in tetra-SQ-treated cells. Notably, EG derivatives showed better anti-inflammatory effects. Further, gene-disease association analysis predicted substantial improvement in the bioactivities of SQ derivatives in metabolic diseases. Cluster analyses revealed di- and tetra-SQ had more functional similarities than others, reflected in their scanning electron microscopy images; both di- and tetra-SQ self-organized into similar sizes and shapes of vesicles, subsequently improving their cation binding activities. Protein-protein interaction networks further revealed that cation binding activity might explain a major part, if not all, of the differences observed in functional analyses. Altogether, the addition of EG derivatives may improve the biological responses of SQ and thus may enhance its health-promoting potential.
Collapse
Affiliation(s)
- Yu Cheng
- Tsukuba Life Science Innovation Program (T-LSI), Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Japan
| | - Farhana Ferdousi
- Tsukuba Life Science Innovation Program (T-LSI), Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Japan
- Institute of Life and Environmental Sciences, University of Tsukuba, Japan
- Alliance of Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan
| | | | - Tran Ngoc Linh
- National Institute of Advanced Industrial Science and Technology (AIST)-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), University of Tsukuba, Tsukuba, Japan
| | - Munkhzul Ganbold
- National Institute of Advanced Industrial Science and Technology (AIST)-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), University of Tsukuba, Tsukuba, Japan
| | - Akira Yada
- National Institute of Advanced Industrial Science and Technology (AIST)-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), University of Tsukuba, Tsukuba, Japan
- Interdisciplinary Research Center for Catalytic Chemistry, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba Central 5, 1-1-1 Higashi, Tsukuba, 305-8565, Japan
| | - Takashi Arimura
- National Institute of Advanced Industrial Science and Technology (AIST)-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), University of Tsukuba, Tsukuba, Japan
| | - Hiroko Isoda
- Tsukuba Life Science Innovation Program (T-LSI), Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Japan
- Institute of Life and Environmental Sciences, University of Tsukuba, Japan
- Alliance of Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan
- National Institute of Advanced Industrial Science and Technology (AIST)-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
43
|
Callan A, Jha S, Valdez L, Baldado L, Tsin A. TGF-β Signaling Pathways in the Development of Diabetic Retinopathy. Int J Mol Sci 2024; 25:3052. [PMID: 38474297 PMCID: PMC10932130 DOI: 10.3390/ijms25053052] [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: 01/23/2024] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
Diabetic retinopathy (DR), a prevalent complication of diabetes mellitus affecting a significant portion of the global population, has long been viewed primarily as a microvascular disorder. However, emerging evidence suggests that it should be redefined as a neurovascular disease with multifaceted pathogenesis rooted in oxidative stress and advanced glycation end products. The transforming growth factor-β (TGF-β) signaling family has emerged as a major contributor to DR pathogenesis due to its pivotal role in retinal vascular homeostasis, endothelial cell barrier function, and pericyte differentiation. However, the precise roles of TGF-β signaling in DR remain incompletely understood, with conflicting reports on its impact in different stages of the disease. Additionally, the BMP subfamily within the TGF-β superfamily introduces further complexity, with BMPs exhibiting both pro- and anti-angiogenic properties. Furthermore, TGF-β signaling extends beyond the vascular realm, encompassing immune regulation, neuronal survival, and maintenance. The intricate interactions between TGF-β and reactive oxygen species (ROS), non-coding RNAs, and inflammatory mediators have been implicated in the pathogenesis of DR. This review delves into the complex web of signaling pathways orchestrated by the TGF-β superfamily and their involvement in DR. A comprehensive understanding of these pathways may hold the key to developing targeted therapies to halt or mitigate the progression of DR and its devastating consequences.
Collapse
Affiliation(s)
| | | | | | | | - Andrew Tsin
- School of Medicine, The University of Texas Rio Grande Valley, Edinburg, TX 78539, USA; (A.C.); (S.J.); (L.V.); (L.B.)
| |
Collapse
|
44
|
McCullough MJ, Tune MK, Cabrera JC, Torres-Castillo J, He M, Feng Y, Doerschuk CM, Dang H, Beltran AS, Hagan RS, Mock JR. Characterization of the MT-2 Treg-like cell line in the presence and absence of forkhead box P3 (FOXP3). Immunol Cell Biol 2024; 102:211-224. [PMID: 38288547 DOI: 10.1111/imcb.12725] [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: 03/22/2023] [Revised: 11/24/2023] [Accepted: 01/08/2024] [Indexed: 03/02/2024]
Abstract
CD4+ forkhead box P3 (FOXP3)+ regulatory T cells (Tregs) are essential in maintaining immune tolerance and suppressing excessive immune responses. Tregs also contribute to tissue repair processes distinct from their roles in immune suppression. For these reasons, Tregs are candidates for targeted therapies for inflammatory and autoimmune diseases, and in diseases where tissue damage occurs. MT-2 cells, an immortalized Treg-like cell line, offer a model to study Treg biology and their therapeutic potential. In the present study, we use clustered regularly interspaced palindromic repeats (CRISPR)-mediated knockdown of FOXP3 in MT-2 cells to understand the transcriptional and functional changes that occur when FOXP3 is lost and to compare MT-2 cells with primary human Tregs. We demonstrate that loss of FOXP3 affects the transcriptome of MT-2 cells and that FOXP3's potential downstream targets include a wide range of transcripts that participate in the cell cycle, promote growth and contribute to inflammatory processes, but do not wholly simulate previously reported human primary Treg transcriptional changes in the absence of FOXP3. We also demonstrate that FOXP3 regulates cell cycling and proliferation, expression of molecules crucial to Treg function and MT-2 cell-suppressive activities. Thus, MT-2 cells offer opportunities to address regulatory T-cell functions in vitro.
Collapse
Affiliation(s)
- Morgan J McCullough
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Miriya K Tune
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, USA
| | | | - Jose Torres-Castillo
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Minghong He
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yongqiang Feng
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Claire M Doerschuk
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Center for Airways Disease, University of North Carolina, Chapel Hill, NC, USA
| | - Hong Dang
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Adriana S Beltran
- Department of Genetics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Robert S Hagan
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Jason R Mock
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
45
|
Sarkar R, Shaaz M, Sehrawat S. Myeloid derived suppressor cells potentiate virus-specific memory CD8 + T cell response. Microbes Infect 2024; 26:105277. [PMID: 38103861 DOI: 10.1016/j.micinf.2023.105277] [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: 03/02/2023] [Revised: 11/03/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023]
Abstract
How therapeutically administered myeloid derived suppressor cells (MDSCs) modulate differentiation of virus-specific CD8+ T cell was investigated. In vitro generated MDSCs from bone marrow precursors inhibited the expansion of stimulated CD8+ T cells but the effector cells in the recipients of MDSCs showed preferential memory transition during Influenza A virus (IAV) or an α- (Herpes Simplex Virus) as well as a γ- (murine herpesvirus 68) herpesvirus infection. Memory CD8+ T cells thus generated constituted a heterogenous population with a large fraction showing effector memory (CD62LloCCR7-) phenotype. Such cells could be efficiently recalled in the rechallenged animals and controlled the secondary infection better. Memory potentiating effects of MDSCs occurred irrespective of the clonality of the responding CD8+ T cells as well as the nature of infecting viruses. Compared to the MDSCs recipients, effector cells of MDSCs recipients showed higher expression of molecules known to drive cellular survival (IL-7R, Bcl2) and memory formation (Tcf7, Id3, eomesodermin). Therapeutically administered MDSCs not only mitigated the tissue damaging response during a resolving IAV infection but also promoted the differentiation of functional memory CD8+ T cells. Therefore, MDSCs therapy could be useful in managing virus-induced immunopathological reactions without compromising immunological memory.
Collapse
Affiliation(s)
- Roman Sarkar
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar Knowledge City PO Manauli, Mohali 140306, Punjab, India
| | - Mohammad Shaaz
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar Knowledge City PO Manauli, Mohali 140306, Punjab, India
| | - Sharvan Sehrawat
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar Knowledge City PO Manauli, Mohali 140306, Punjab, India.
| |
Collapse
|
46
|
Petakh P, Kamyshna I, Oksenych V, Kamyshnyi O. Metformin Alters mRNA Expression of FOXP3, RORC, and TBX21 and Modulates Gut Microbiota in COVID-19 Patients with Type 2 Diabetes. Viruses 2024; 16:281. [PMID: 38400056 PMCID: PMC10893440 DOI: 10.3390/v16020281] [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: 12/12/2023] [Revised: 01/24/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
COVID-19 remains a significant global concern, particularly for individuals with type 2 diabetes who face an elevated risk of hospitalization and mortality. Metformin, a primary treatment for type 2 diabetes, demonstrates promising pleiotropic properties that may substantially mitigate disease severity and expedite recovery. Our study of the gut microbiota and the mRNA expression of pro-inflammatory and anti-inflammatory T-lymphocyte subpopulations showed that metformin increases bacterial diversity while modulating gene expression related to T-lymphocytes. This study found that people who did not take metformin had a downregulated expression of FOXP3 by 6.62-fold, upregulated expression of RORC by 29.0-fold, and upregulated TBX21 by 1.78-fold, compared to the control group. On the other hand, metformin patients showed a 1.96-fold upregulation in FOXP3 expression compared to the control group, along with a 1.84-fold downregulation in RORC expression and an 11.4-fold downregulation in TBX21 expression. Additionally, we found a correlation with gut microbiota (F/B ratio and alpha-diversity index) and pro-inflammatory biomarkers. This novel observation of metformin's impact on T-cells and gut microbiota opens new horizons for further exploration through clinical trials to validate and confirm our data. The potential of metformin to modulate immune responses and enhance gut microbiota diversity suggests a promising avenue for therapeutic interventions in individuals with type 2 diabetes facing an increased risk of severe outcomes from COVID-19.
Collapse
Affiliation(s)
- Pavlo Petakh
- Department of Biochemistry and Pharmacology, Uzhhorod National University, 88000 Uzhhorod, Ukraine
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| | - Iryna Kamyshna
- Department of Medical Rehabilitation, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine;
| | - Valentyn Oksenych
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine
| |
Collapse
|
47
|
Sun J, Ruiz Daniels R, Balic A, Andresen AMS, Bjørgen H, Dobie R, Henderson NC, Koppang EO, Martin SAM, Fosse JH, Taylor RS, Macqueen DJ. Cell atlas of the Atlantic salmon spleen reveals immune cell heterogeneity and cell-specific responses to bacterial infection. FISH & SHELLFISH IMMUNOLOGY 2024; 145:109358. [PMID: 38176627 DOI: 10.1016/j.fsi.2024.109358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/23/2023] [Accepted: 01/02/2024] [Indexed: 01/06/2024]
Abstract
The spleen is a conserved secondary lymphoid organ that emerged in parallel to adaptive immunity in early jawed vertebrates. Recent studies have applied single cell transcriptomics to reveal the cellular composition of spleen in several species, cataloguing diverse immune cell types and subpopulations. In this study, 51,119 spleen nuclei transcriptomes were comprehensively investigated in the commercially important teleost Atlantic salmon (Salmo salar L.), contrasting control animals with those challenged with the bacterial pathogen Aeromonas salmonicida. We identified clusters of nuclei representing the expected major cell types, namely T cells, B cells, natural killer-like cells, granulocytes, mononuclear phagocytes, endothelial cells, mesenchymal cells, erythrocytes and thrombocytes. We discovered heterogeneity within several immune lineages, providing evidence for resident macrophages and melanomacrophages, infiltrating monocytes, several candidate dendritic cell subpopulations, and B cells at distinct stages of differentiation, including plasma cells and an igt + subset. We provide evidence for twelve candidate T cell subsets, including cd4+ T helper and regulatory T cells, one cd8+ subset, three γδT subsets, and populations double negative for cd4 and cd8. The number of genes showing differential expression during the early stages of Aeromonas infection was highly variable across immune cell types, with the largest changes observed in macrophages and infiltrating monocytes, followed by resting mature B cells. Our analysis provides evidence for a local inflammatory response to infection alongside B cell maturation in the spleen, and upregulation of ccr9 genes in igt + B cells, T helper and cd8+ cells, and monocytes, consistent with the recruitment of immune cell populations to the gut to deal with Aeromonas infection. Overall, this study provides a new cell-resolved perspective of the immune actions of Atlantic salmon spleen, highlighting extensive heterogeneity hidden to bulk transcriptomics. We further provide a large catalogue of cell-specific marker genes that can be leveraged to further explore the function and structural organization of the salmonid immune system.
Collapse
Affiliation(s)
- Jianxuan Sun
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | - Rose Ruiz Daniels
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | - Adam Balic
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK; Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | | | - Håvard Bjørgen
- Unit of Anatomy, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Ross Dobie
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK; MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Erling Olaf Koppang
- Unit of Anatomy, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Samuel A M Martin
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | | | - Richard S Taylor
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK
| | - Daniel J Macqueen
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, UK.
| |
Collapse
|
48
|
Hao Y, Stuart T, Kowalski MH, Choudhary S, Hoffman P, Hartman A, Srivastava A, Molla G, Madad S, Fernandez-Granda C, Satija R. Dictionary learning for integrative, multimodal and scalable single-cell analysis. Nat Biotechnol 2024; 42:293-304. [PMID: 37231261 PMCID: PMC10928517 DOI: 10.1038/s41587-023-01767-y] [Citation(s) in RCA: 378] [Impact Index Per Article: 378.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 03/28/2023] [Indexed: 05/27/2023]
Abstract
Mapping single-cell sequencing profiles to comprehensive reference datasets provides a powerful alternative to unsupervised analysis. However, most reference datasets are constructed from single-cell RNA-sequencing data and cannot be used to annotate datasets that do not measure gene expression. Here we introduce 'bridge integration', a method to integrate single-cell datasets across modalities using a multiomic dataset as a molecular bridge. Each cell in the multiomic dataset constitutes an element in a 'dictionary', which is used to reconstruct unimodal datasets and transform them into a shared space. Our procedure accurately integrates transcriptomic data with independent single-cell measurements of chromatin accessibility, histone modifications, DNA methylation and protein levels. Moreover, we demonstrate how dictionary learning can be combined with sketching techniques to improve computational scalability and harmonize 8.6 million human immune cell profiles from sequencing and mass cytometry experiments. Our approach, implemented in version 5 of our Seurat toolkit ( http://www.satijalab.org/seurat ), broadens the utility of single-cell reference datasets and facilitates comparisons across diverse molecular modalities.
Collapse
Affiliation(s)
- Yuhan Hao
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Tim Stuart
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Madeline H Kowalski
- New York Genome Center, New York, NY, USA
- Institute for System Genetics, NYU Langone Medical Center, New York, NY, USA
| | - Saket Choudhary
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Paul Hoffman
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Austin Hartman
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Avi Srivastava
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | | | - Shaista Madad
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Carlos Fernandez-Granda
- Center for Data Science, New York University, New York, NY, USA
- Courant Institute of Mathematical Sciences, New York University, New York, NY, USA
| | - Rahul Satija
- Center for Genomics and Systems Biology, New York University, New York, NY, USA.
- New York Genome Center, New York, NY, USA.
| |
Collapse
|
49
|
Asri N, Nazemalhosseini Mojarad E, Taleghani MY, Houri H, Saeedi Niasar M, Rezaei-Tavirani M, Jahani-Sherafat S, Shahbazkhani A, Nikzamir A, Ehsani Ardakani MJ, Khodadost M, Rostami-Nejad M. Evaluating CD4 and Foxp3 mRNA Expression in Tissue Specimens of Celiac Disease and Colorectal Cancer Patients. Asian Pac J Cancer Prev 2024; 25:647-652. [PMID: 38415552 PMCID: PMC11077127 DOI: 10.31557/apjcp.2024.25.2.647] [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: 10/16/2023] [Accepted: 02/20/2024] [Indexed: 02/29/2024] Open
Abstract
OBJECTIVE Celiac disease (CD) and colorectal cancer (CRC) are distinct gastrointestinal conditions with a debated association. This study aimed to evaluate the mRNA expression of CD4 and Foxp3 in tissue specimens of CD and CRC patients. The findings can provide valuable insights into the complex connection between these different gastrointestinal conditions. METHODS Tissue samples from 100 CRC patients, 50 CD patients, and 50 healthy controls (HCs) were collected. RNA extraction, cDNA synthesis, and quantitative real-time PCR were performed. Statistical analysis was conducted using ANOVA and Pearson's correlation test. RESULT CD4 mRNA expression was significantly higher in CRC patients compared to CD patients and HCs (P<0.0001 for both). Foxp3 mRNA expression was significantly higher in CD patients compared to CRC patients and HCs (P<0.0001 for both). Clinicopathological characteristics did not correlate significantly with gene expression levels. CONCLUSION This study reveals differential expression patterns of CD4 and Foxp3 mRNA in CRC and CD patients. Upregulated CD4 mRNA suggests its potential role in promoting tumor growth, while increased Foxp3 mRNA expression may reflect an immunosuppressive mechanism in CD pathogenesis. These findings provide insights into the molecular and immunological aspects of CRC and CD, warranting further studies for potential therapeutic strategies.
Collapse
Affiliation(s)
- Nastaran Asri
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ehsan Nazemalhosseini Mojarad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Yaghoob Taleghani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hamidreza Houri
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mahsa Saeedi Niasar
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Somayeh Jahani-Sherafat
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Amirhossein Shahbazkhani
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abdolrahim Nikzamir
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Javad Ehsani Ardakani
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mahmood Khodadost
- Department of Traditional Medicine, School of Traditional Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Rostami-Nejad
- Celiac Disease and Gluten Related Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
50
|
Kim DW, Kim YC, Kovari BP, Martinez M, Miao R, Yu J, Mehta R, Strosberg J, Imanirad I, Kim RD. Biomarker Analysis from a Phase I/Ib Study of Regorafenib and Nivolumab in Mismatch Repair-Proficient Advanced Refractory Colorectal Cancer. Cancers (Basel) 2024; 16:556. [PMID: 38339307 PMCID: PMC10854756 DOI: 10.3390/cancers16030556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Previously, we reported the modest but durable anticancer activity of regorafenib/nivolumab in mismatch repair-proficient (pMMR) refractory colorectal cancer in our I/Ib study. Our finding suggests the necessity of biomarkers for better selection of patients. Baseline clinical and pathological characteristics, blood and tumor samples from the patients in the trial were collected and evaluated to discover potential biomarkers. The obtained samples were assessed for immunohistochemistry, ELISA and RNA sequencing. Their correlations with clinical outcome were analyzed. A high albumin level was significantly associated with improved progression-free survival (PFS), overall survival (OS) and disease control. Non-liver metastatic disease showed prolonged PFS and OS. Low regulatory T-cell (Treg) infiltration correlated with prolonged PFS. Low MIP-1β was associated with durable response and improved OS significantly. Upregulation of 23 genes, including CAPN9, NAPSA and ROS1, was observed in the durable disease control group, and upregulation of 10 genes, including MRPS18A, MAIP1 and CMTR2, was associated with a statistically significant improvement of PFS. This study suggests that pretreatment albumin, MIP-1β, non-liver metastatic disease and Treg infiltration may be potential predictive biomarkers of regorafenib/nivolumab in pMMR colorectal cancer. Further studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Dae Won Kim
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (D.W.K.); (M.M.); (R.M.); (J.Y.); (R.M.); (J.S.); (I.I.)
| | - Young-Chul Kim
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Bence P. Kovari
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Maria Martinez
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (D.W.K.); (M.M.); (R.M.); (J.Y.); (R.M.); (J.S.); (I.I.)
| | - Ruoyu Miao
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (D.W.K.); (M.M.); (R.M.); (J.Y.); (R.M.); (J.S.); (I.I.)
| | - James Yu
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (D.W.K.); (M.M.); (R.M.); (J.Y.); (R.M.); (J.S.); (I.I.)
| | - Rutika Mehta
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (D.W.K.); (M.M.); (R.M.); (J.Y.); (R.M.); (J.S.); (I.I.)
| | - Jonathan Strosberg
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (D.W.K.); (M.M.); (R.M.); (J.Y.); (R.M.); (J.S.); (I.I.)
| | - Iman Imanirad
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (D.W.K.); (M.M.); (R.M.); (J.Y.); (R.M.); (J.S.); (I.I.)
| | - Richard D. Kim
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (D.W.K.); (M.M.); (R.M.); (J.Y.); (R.M.); (J.S.); (I.I.)
| |
Collapse
|