1
|
Franco-Fuquen P, Figueroa-Aguirre J, Martínez DA, Moreno-Cortes EF, Garcia-Robledo JE, Vargas-Cely F, Castro-Martínez DA, Almaini M, Castro JE. Cellular therapies in rheumatic and musculoskeletal diseases. J Transl Autoimmun 2025; 10:100264. [PMID: 39931050 PMCID: PMC11808717 DOI: 10.1016/j.jtauto.2024.100264] [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: 10/29/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 02/13/2025] Open
Abstract
A substantial proportion of patients diagnosed with rheumatologic and musculoskeletal diseases (RMDs) exhibit resistance to conventional therapies or experience recurrent symptoms. These diseases, which include autoimmune disorders such as multiple sclerosis, rheumatoid arthritis, and systemic lupus erythematosus, are marked by the presence of autoreactive B cells that play a critical role in their pathogenesis. The persistence of these autoreactive B cells within lymphatic organs and inflamed tissues impairs the effectiveness of B-cell-depleting monoclonal antibodies like rituximab. A promising therapeutic approach involves using T cells genetically engineered to express chimeric antigen receptors (CARs) that target specific antigens. This strategy has demonstrated efficacy in treating B-cell malignancies by achieving long-term depletion of malignant and normal B cells. Preliminary data from patients with RMDs, particularly those with lupus erythematosus and dermatomyositis, suggest that CAR T-cells targeting CD19 can induce rapid and sustained depletion of circulating B cells, leading to complete clinical and serological responses in cases that were previously unresponsive to conventional therapies. This review will provide an overview of the current state of preclinical and clinical studies on the use of CAR T-cells and other cellular therapies for RMDs. Additionally, it will explore potential future applications of these innovative treatment modalities for managing patients with refractory and recurrent manifestations of these diseases.
Collapse
Affiliation(s)
- Pedro Franco-Fuquen
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Juana Figueroa-Aguirre
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - David A. Martínez
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Eider F. Moreno-Cortes
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Juan E. Garcia-Robledo
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Fabio Vargas-Cely
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | | | - Mustafa Almaini
- Rheumatology, Allergy & Clinical Immunology Division, Mafraq Hospital, United Arab Emirates
| | - Januario E. Castro
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
2
|
Yan C, Wang G. Advances in research on flavonoids in tumor immunotherapy (Review). Mol Med Rep 2025; 31:150. [PMID: 40211703 PMCID: PMC11995692 DOI: 10.3892/mmr.2025.13515] [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: 09/03/2024] [Accepted: 02/25/2025] [Indexed: 04/16/2025] Open
Abstract
Cancer immunotherapy is an approach used in anti‑tumor treatment; however, its efficacy is limited to specific tumor types that are inherently sensitive to immune system modulation. Expanding the scope of indications and enhancing the efficacy of cancer immunotherapy are key goals for continued advancement. Flavonoids modulate the tumor‑immunosuppressive microenvironment. Integrating flavonoids with immunotherapeutic modalities, including cancer vaccines, immune checkpoint inhibitors and adoptive immune‑cell therapy, has potential in terms of augmenting the therapeutic efficacy of immunotherapy. The present review aimed to summarize flavonoids that enhance cancer immunotherapy, focusing on their underlying mechanisms and the application of nanotechnology to overcome inherent limitations such as poor solubility, low bioavailability, rapid metabolism, and instability under physiological conditions, thereby highlighting the potential of flavonoids in advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Chaoguang Yan
- Department of Oncology, Weifang Chinese Medicine Hospital, Weifang, Shandong 261000 P.R. China
| | - Guangchun Wang
- Department of Oncology, Weifang Chinese Medicine Hospital, Weifang, Shandong 261000 P.R. China
| |
Collapse
|
3
|
Bellocchi C, Favalli EG, Maioli G, Agape E, Rossato M, Paini M, Severino A, Vigone B, Biggioggero M, Trombetta E, Caporali R, Beretta L. Whole-Blood RNA Sequencing Profiling of Patients With Rheumatoid Arthritis Treated With Tofacitinib. ACR Open Rheumatol 2025. [PMID: 40388487 DOI: 10.1002/acr2.11761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 09/13/2024] [Accepted: 09/19/2024] [Indexed: 05/21/2025] Open
Abstract
OBJECTIVE Patients with rheumatoid arthritis (RA) often fail to respond to therapies, including JAK inhibitors (JAKi), and treatment allocation is made via a trial-and-error strategy. A comprehensive analysis of responses to JAKi, including tofacitinib, by RNA sequencing (RNAseq) would allow the discovery of transcriptomic markers with a two-fold meaning: (1) an improved knowledge about the mechanisms of response to treatment (inference modeling) and (2) the definition of features that may be useful in treatment optimization and assignment (predictive modeling). METHODS Thirty-three patients with active RA were treated with a tofacitinib dose of 5 mg twice a day for 24 weeks and evaluated for EULAR Disease Activity Score in 28 joints using the C-reactive protein level response. Whole-blood RNA was collected before and after treatment to perform RNAseq transcriptome analysis. Linear models were used to determine differentially expressed genes (DEGs) (1) at baseline according to clinical responses and (2) in the pre-post comparison after tofacitinib treatment and in relation to EULAR responses. The capability of DEGs to predict a successful treatment was tested via machine learning modeling after extensive internal validation. RESULTS Of 26 patients who completed the study (per-protocol analysis), 15 (57.7%) achieved good responses, and 7 (26.9%) and 4 (15.3%) had moderate and no responses, respectively. Overall, 273 baseline genes were significantly associated with the attainment of good responses, contributing to several pathways linked to the immune system or RA pathogenesis (eg, citrullination processes and the negative regulation of natural killer function). The expression of several molecules was reverted by tofacitinib when good responses were reached, including AKT3, GK5, KLF12, FCRL3, BIRC3, TSPOAP1, and P2RY10. Finally, we isolated 14 markers that singularly were capable of predicting the attainment of good responses, including, NKG2D, CD226, CLEC2D, and CD52. CONCLUSION Whole-blood transcriptome analysis of patients with RA treated with tofacitinib identified genes whose expression may be relevant in prognostication and understanding the mechanisms of responses to therapy.
Collapse
Affiliation(s)
- Chiara Bellocchi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico and University of Milan, Milan, Italy
| | | | - Gabriella Maioli
- University of Milan and ASST PiniCTO - Presidio Gaetano Pini, Milan, Italy
| | | | | | | | - Adriana Severino
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Barbara Vigone
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Elena Trombetta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Roberto Caporali
- University of Milan and ASST PiniCTO - Presidio Gaetano Pini, Milan, Italy
| | - Lorenzo Beretta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
4
|
He Y, Feng J, Shi W, Ren Y, Liu Y, Kang H, Tian J, Jie Y. Correlation among ocular surface changes and systemic hematologic indexes and disease activity in primary Sjögren's syndrome: a cross-sectional study. BMC Ophthalmol 2025; 25:270. [PMID: 40329232 PMCID: PMC12054185 DOI: 10.1186/s12886-025-04050-7] [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: 01/09/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND To explore the relationship among ocular surface changes, systemic hematologic indexes, and disease activity in primary Sjögren's syndrome patients. METHODS Thirty-three primary Sjögren's syndrome patients and 36 healthy controls were recruited in this cross-sectional study. All participants underwent complete ocular surface testing, including dry eye symptoms and signs, tear multi-cytokine analysis, and conjunctival impression cytology (CIC). Multiple systemic hematologic indexes and disease activity were also evaluated, including autoantibodies, immune cells, the EULAR Sjögren's Syndrome Patient Reported Index (ESSPRI), and the EULAR Sjögren's Syndrome Disease Activity Index (ESSDAI). RESULTS Primary Sjögren's syndrome patients exhibited significant dry eye, severe conjunctivochalasis, decreased goblet cell density, and severe squamous epithelial on the ocular surface. Interferon-inducible T cell alpha chemoattractant (I-TAC), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin (IL)-1β, IL-5, IL-8, IL-10, IL-13, IL-21, C-C motif chemokine ligand (CCL)4, interferon-gamma (IFN-γ), CCL20, and tumor necrosis factor-gamma (TNF-α) in the tear fluid of pSS patients changed significantly. Correlation analysis showed that anti-SSA was relevant to ocular surface disease index (OSDI) score, tear break-up time (TBUT), and meibomian gland secretion (MGS). CD8+ T cell percentages were relevant to TBUT and corneal fluorescein staining score (CFS). IL-8, IL-13, CCL4, and TNF-α were correlated with RF-IgA. IL-1β, CCL4, and TNF-α were correlated with CD8+ T cell counts. IL-5 and CCL20 were correlated with the ratio of helper T cells and suppressor T cells. Tear I-TAC, IL-8, CCL20, and TNF-α were significantly correlated with the ESSDAI of different domains. CONCLUSIONS Our results revealed that the ocular surface changes in pSS patients were significantly correlated with systemic hematologic indexes and disease activity.
Collapse
Affiliation(s)
- Yan He
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmic and Visual Science Key Laboratory, Beijing, China
| | - Jianing Feng
- Xi'an People's Hospital (Xi'an Fourth Hospital), Shaanxi Eye Hospital, Northwest University Affiliated People's Hospital, Xi'an, Shaanxi, China
| | - Wen Shi
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Yuerong Ren
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Yingyi Liu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmic and Visual Science Key Laboratory, Beijing, China
| | - Huanmin Kang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jing Tian
- Department of Rheumatism and Immunology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Ying Jie
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmic and Visual Science Key Laboratory, Beijing, China.
| |
Collapse
|
5
|
Aghaee F, Abedinpour M, Anvari S, Saberi A, Fallah A, Bakhshi A. Natural killer cells in multiple sclerosis: foe or friends? Front Cell Neurosci 2025; 19:1500770. [PMID: 40255388 PMCID: PMC12006147 DOI: 10.3389/fncel.2025.1500770] [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: 09/23/2024] [Accepted: 03/20/2025] [Indexed: 04/22/2025] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated disorder involving the central nervous system (CNS), in which demyelination is caused. The initiation and progression of MS is thought to depend largely on CD4+ T lymphocytes, yet new data has emphasized the involvement of the innate immune system in the MS disease responses. Generally, several types of immune cells play a part, with natural killer (NK) cells being essential. Different subsets of natural killer cells function differently within the course of an autoimmune disease, such as MS. There are mainly two types of natural killers in humans: immature CD56 bright CD16- and mature CD56 dim CD16+ natural killers, together with their respective subtypes. Factors from natural killers expand the T cell population and control the process by which native CD4+ T cells differentiate into Th1 or Th2 lymphocytes, which affect autoimmune responses. Natural killer subsets CD56 bright and CD56 dim may have differing roles in MS development. The impact of these NK cell subsets is influenced by factors such as Granzymes, genetics, infections, TLR, and HSP. We reviewed and evaluated the relationship between natural killer cells and MS.
Collapse
Affiliation(s)
- Fatemeh Aghaee
- Member Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammadreza Abedinpour
- Member Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Saeid Anvari
- Department of Neurology, Neurosciences Research Center, Poursina Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Alia Saberi
- Department of Internal Medicine, Regenerative Medicine Research Center, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Amir Fallah
- Member Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Arash Bakhshi
- Member Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
6
|
Duan X, Lv X, Wang X, Zhang Y, Hu Y, Li H, Zhou Y, Jing Y. Impact of immune cell metabolism on membranous nephropathy and prospective therapy. Commun Biol 2025; 8:405. [PMID: 40065158 PMCID: PMC11893770 DOI: 10.1038/s42003-025-07816-3] [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: 08/26/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Membranous nephropathy (MN) is a primary glomerular disease commonly causing adult nephrotic syndrome. Characterized by thickened glomerular capillary walls due to immune complex deposition, MN is a complex autoimmune disorder. Its pathogenesis involves immune deposit formation, complement activation, and a heightened risk of renal failure. Central to MN is immune system dysfunction, particularly the dysregulation of B and T cell responses. B cells contribute to renal injury through the production of autoantibodies, particularly IgG targeting the phospholipase A2 receptor (PLA2R) on podocytes, while T cells modulate immune responses that influence disease progression. Metabolic reprogramming alters lymphocyte survival, differentiation, proliferation, and function, potentially triggering autoimmune processes. Although the link between immune cell metabolism and MN remains underexplored, this review highlights recent advances in understanding immune metabolism and its role in MN. These insights may provide novel biomarkers and therapeutic strategies for MN treatment.
Collapse
Affiliation(s)
- Xuemei Duan
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Xin Lv
- Department of Nephrology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Xiaocui Wang
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yunfei Zhang
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Ying Hu
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Haonan Li
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yongnian Zhou
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| | - Yukai Jing
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| |
Collapse
|
7
|
Guo R, Mi L, Gao J, Yang Y, Zhao M, He X, Ji Y, Hu Y, Gao Y, Xu K. Natural killer cells are decreased in systemic sclerosis and have diagnostic value for pulmonary arterial hypertension incorporation. Sci Rep 2025; 15:5178. [PMID: 39939388 PMCID: PMC11821854 DOI: 10.1038/s41598-025-89238-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/04/2025] [Indexed: 02/14/2025] Open
Abstract
The aim of this study was to investigate lymphocyte subsets, especially natural killer (NK) cells, in patients with systemic sclerosis (SSc) and evaluate the diagnostic value of NK cells in secondary pulmonary arterial hypertension (PAH). A total of 115 SSc patients and 100 age- and sex-matched health controls (HCs) were enrolled in this study. Flow cytometry was employed to quantify NK cells, while the association between NK cells and disease activity as well as PAH was investigated to further elucidate its diagnostic potential. The absolute count of NK (CD3-CD56+) cells significantly decreased in SSc patients. There was a negative correlation between the mRSS score and the injury index. The levels of cytokine exhibited significant elevation among SSc patients. Conversely, SSc-PAH patients demonstrated significantly elevated levels of CRP, UA, and BNP. Additionally, there was a significant reduction in the absolute level of NK cells. ROC curve analysis revealed that the optimal cut-off point for NK cells was 185 cells/µL, while for BNP it was 70.50 pg/mL and for UA it was 323.00 µmol/L. Our study revealed a significant inverse correlation between peripheral blood NK cell levels and the incidence of complicated PAH in patients with SSc.
Collapse
Affiliation(s)
- Ronghong Guo
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- Department of Rheumatology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Liangyu Mi
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- Department of Rheumatology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Jinfang Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- Department of Rheumatology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yanli Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- Department of Rheumatology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Miaomiao Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- Department of Rheumatology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Xiaoyao He
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- Department of Rheumatology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yuli Ji
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- Department of Rheumatology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yuting Hu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- Department of Rheumatology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yanan Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
- Department of Rheumatology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Ke Xu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
- Department of Rheumatology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| |
Collapse
|
8
|
Wiendl H, Barkhof F, Montalban X, Achiron A, Derfuss T, Chan A, Hodgkinson S, Prat A, Leocani L, Schmierer K, Sellebjerg F, Vermersch P, Jin H, Chudecka A, Kloetgen A, Lin D, Gardner L, De Stefano N. Blood biomarker dynamics in people with relapsing multiple sclerosis treated with cladribine tablets: results of the 2-year MAGNIFY-MS study. Front Immunol 2025; 16:1512189. [PMID: 39963134 PMCID: PMC11830603 DOI: 10.3389/fimmu.2025.1512189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/08/2025] [Indexed: 02/20/2025] Open
Abstract
Background and objectives Cladribine tablets (CladT) represent an effective immune reconstitution therapy, administered in short treatment courses over two consecutive years. To better understand the amplitude of immune changes, we performed a comprehensive analysis during the 2-year study period for the entire MAGNIFY-MS population (N=270). In addition to lymphocyte kinetics, we studied intracellular cytokines serum proteins, and their associations with clinical outcomes. To put these changes into perspective, we analyzed transcriptional changes in T and B cells and associated biological pathways before and after each treatment course with CladT. Methods Immunophenotyping and transcriptomics were performed at regular visits with major differences reported between baseline (BL) and after each yearly treatment course. Assessments included: lymphocyte dynamics, RNA sequencing (B and T cells), intracellular cytokines, serum proteins (immunoglobulins [IgG and IgM], and serum neurofilament light chain [sNfL]). Clinical measures included: MRI activity, annualized relapse rate (ARR), 6-month confirmed disability progression (6mCDP), timed 25-foot walk (T25FW), and 9-hole peg test (9HPT). Results All B, T and NK cells were reduced at month (M)3 after CladT administration, except regulatory B cells which increased above BL from M3 to M24. Naïve and transitional B cells recovered at M6; all other B and T cell subsets remained below BL levels. Reductions in all NK cell subtypes were observed at M3, CD16lowCD56bright and NKp46 cells reconstituted at M6 and M12 respectively. Changes in genes and pathways associated with innate and adaptive immune response were observed after CladT treatment, along with reductions in pro-inflammatory cytokine-producing B and T cells and increases in anti-inflammatory cytokine-producing T cells. IgG and IgM levels remained above the lower limits of normal in most participants. sNfL levels decreased, remaining reduced by M24. Significant reductions in the annualized combined unique active lesion count occurred from M2 onwards. ARR was 0.11 (95% confidence interval: 0.09,0.15), with 83% participants free of qualifying relapses. Over 90% of participants were free of 6mCDP, around 87% had no confirmed progression on T25FW and 9HPT. No significant correlations were seen between clinical parameters and lymphocyte dynamics to M6. The safety profile was consistent with previous reports. Discussion Deep longitudinal immunophenotyping, analysis of transcriptional changes, reduction in cells expressing pro-inflammatory cytokines, along with the marker of neuroaxonal damage provide novel and innovative evidence of CladT rebalancing the immune system towards a more homeostatic and less pathogenic state. Clinical Trial Registration https://clinicaltrials.gov/study/, identifier NCT03364036.
Collapse
Affiliation(s)
- Heinz Wiendl
- Department of Neurology, Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, London, United Kingdom
| | - Xavier Montalban
- Department of Neurology-Neuroimmunology, Centre d’Esclerosi Múltiple de Catalunya (Cemcat), Hospital Universitario Vall d’Hebron, Barcelona, Spain
| | - Anat Achiron
- Multiple Sclerosis Center, Sheba Academic Medical Center, Ramat Gan, Israel
- Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Tobias Derfuss
- Department of Neurology, University Hospital Basel, Basel, Switzerland
| | - Andrew Chan
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Suzanne Hodgkinson
- Ingham Institute for Applied Medical Research, University of New South Wales Medicine and Liverpool Hospital, Sydney, NSW, Australia
| | - Alexandre Prat
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Letizia Leocani
- Department of Neurology, University Vita-Salute San Raffaele, Milan, Italy
- Experimental Neurophysiology Unit, Scientific Institute IRCCS San Raffaele, Milan, Italy
- Department of Neurorehabilitation Science, Casa di Cura Igea, Milan, Italy
| | - Klaus Schmierer
- The Blizard Institute, Centre for Neuroscience, Surgery and Trauma, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Clinical Board Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS, Trust, London, United Kingdom
| | - Finn Sellebjerg
- Danish MS Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Patrick Vermersch
- Univ. Lille, Inserm U1172 LilNCog, CHU Lille, FHU Precise, Lille, France
| | - Hulin Jin
- Clinical Measurement Sciences, Merck Healthcare KGaA, Darmstadt, Germany
| | - Anita Chudecka
- Clinical Research Services, Cytel Inc., Geneva, Switzerland
| | - Andreas Kloetgen
- Clinical Measurement Sciences, Merck Healthcare KGaA, Darmstadt, Germany
| | - Dongdong Lin
- Clinical Measurement Sciences, EMD Serono Research & Development Institute, Inc., an affiliate of Merck KGaA, Billerica, MA, United States
| | - Lidia Gardner
- Neurology & Immunology Medical Unit, EMD Serono Research & Development Institute, Inc., an affiliate of Merck KGaA, Billerica, MA, United States
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| |
Collapse
|
9
|
Gallo D, Piantanida E, Bombelli R, Lepanto S, Bruno A, Gallazzi M, Bilato G, Borgese M, Baci D, Mortara L, Tanda ML. Natural Killer Cells in Graves' Disease: Increased Frequency but Impaired Degranulation Ability Compared to Healthy Controls. Int J Mol Sci 2025; 26:977. [PMID: 39940745 PMCID: PMC11816991 DOI: 10.3390/ijms26030977] [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: 12/24/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Graves' disease (GD) is an autoimmune disorder, driven by the appearance of circulating autoantibodies (Ab) against the thyroid stimulating hormone (TSH) receptor, thus causing hyperthyroidism. While antithyroid drugs, the only available treatment for GD, carry a significant risk of relapse, advances in immunology could pave the way for more effective therapies. Natural killer (NK) cells, divided into cytotoxic CD56dim and cytokine-secreting CD56bright subsets, regulate immune responses through cytokine production and cell lysis and may play a role in the pathogenesis of GD. To investigate their involvement, we conducted flow cytometry on peripheral blood samples from 131 GD patients at various stages (disease onset, on antithyroid drugs, and in remission) and 97 age- and sex-matched healthy controls (HC). We analyzed NK cell subsets, activating (CD16, CD69, NKG2D, NKp30) and inhibitory receptors (CD161, NKG2A), degranulation (CD107a), and intracellular cytokines expression (interferon γ, tumor necrosis factor α). Statistical comparisons were made between GD patients and HC and across disease stages. GD patients had a higher frequency of total NK cells (p < 0.028) and CD56bright NK cells (p < 0.01) but a lower frequency of CD56dim NK cells (p = 0.005) compared to HC. NK cells in GD patients expressed activating receptors more frequently, except for NKG2D, but had decreased cytokine expression and degranulation ability. At GD onset, patients had higher frequencies of total NK cells, CD56bright NK cells, and NK cells expressing activating receptors compared to patients receiving ATD treatment and those in remission. CD161+ NK cells were lower at GD onset and returned to levels of HC following treatment. Correlation analysis revealed that free thyroxine (FT4) levels were inversely correlated with CD107a+ NK cells (p < 0.05) and positively correlated with CD69+ NK cells (p < 0.01). These findings suggest that hyperthyroidism impairs NK cell degranulation, with the increased frequency of NK cells potentially compensating for their reduced function. This dysfunction may contribute to the unregulated immune response in GD, highlighting NK cells as a potential target for novel therapeutic strategies.
Collapse
Affiliation(s)
- Daniela Gallo
- Endocrine Unit, Department of Medicine and Surgery, University of Insubria, ASST Dei Sette Laghi, 21100 Varese, Italy; (E.P.); (S.L.); (M.L.T.)
| | - Eliana Piantanida
- Endocrine Unit, Department of Medicine and Surgery, University of Insubria, ASST Dei Sette Laghi, 21100 Varese, Italy; (E.P.); (S.L.); (M.L.T.)
| | - Raffaella Bombelli
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (R.B.); (A.B.); (G.B.); (D.B.)
| | - Silvia Lepanto
- Endocrine Unit, Department of Medicine and Surgery, University of Insubria, ASST Dei Sette Laghi, 21100 Varese, Italy; (E.P.); (S.L.); (M.L.T.)
| | - Antonino Bruno
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (R.B.); (A.B.); (G.B.); (D.B.)
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico MultiMedica, 20138 Milan, Italy;
| | - Matteo Gallazzi
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico MultiMedica, 20138 Milan, Italy;
| | - Giorgia Bilato
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (R.B.); (A.B.); (G.B.); (D.B.)
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico MultiMedica, 20138 Milan, Italy;
| | - Marina Borgese
- Department of Medicine and Technological Innovation, University of Insubria, 21100 Varese, Italy;
| | - Denisa Baci
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (R.B.); (A.B.); (G.B.); (D.B.)
- Laboratory of Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, 20097 Milan, Italy
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (R.B.); (A.B.); (G.B.); (D.B.)
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico MultiMedica, 20138 Milan, Italy;
| | - Maria Laura Tanda
- Endocrine Unit, Department of Medicine and Surgery, University of Insubria, ASST Dei Sette Laghi, 21100 Varese, Italy; (E.P.); (S.L.); (M.L.T.)
| |
Collapse
|
10
|
Yang M, Chen Y, Feng C, Zhang M, Wang H, Zheng Y, Li X. Single-cell RNA sequencing uncovers molecular mechanisms of intravenous immunoglobulin plus methylprednisolone in Kawasaki disease: attenuated monocyte-driven inflammation and improved NK cell cytotoxicity. Front Immunol 2024; 15:1455925. [PMID: 39524437 PMCID: PMC11543420 DOI: 10.3389/fimmu.2024.1455925] [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: 06/27/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Intravenous immunoglobulin (IVIG) plus methylprednisolone as initial intensive therapy or additional therapy in Kawasaki disease (KD) has been used in clinical practice. However, its molecular and cellular mechanism is unclear. Methods We performed single-cell analysis on 14 peripheral blood mononuclear cell (PBMC) samples obtained from 7 KD patients who received either IVIG monotherapy or IVIG plus methylprednisolone therapy. This encompassed 4 samples from KD patients collected before and after IVIG treatment, as well as 3 samples from KD patients before and after IVIG plus methylprednisolone therapy. Results Both IVIG monotherapy and IVIG plus methylprednisolone therapy can increase lymphocyte counts (e.g. CD4+T, CD8+T, and gdT cells) to address lymphopenia. They can also decrease monocyte counts and repress the expression of S100A12, NLRP3, and genes associated with immune-cell migration in monocytes. IVIG combined with methylprednisolone downregulates more monocyte-driven inflammatory pathways than IVIG alone. Additionally, this combination uniquely enhances NK cell cytotoxicity by modulating receptor homeostasis, while significantly upregulating interferon-related genes in CD4+ T cells, CD8+ T cells, and B cells, particularly type I interferons. Conclusion The combination of IVIG with methylprednisolone attenuated monocyte-driven inflammation and improved NK cell cytotoxicity which might provide clues for pediatricians to consider treatment options for children with KD. Whether the monocyte-driven hyperinflammatory state and NK cell function can be indicators for the clinical choice of IVIG with methylprednisolone therapy in KD needs further investigation.
Collapse
Affiliation(s)
- Minna Yang
- Department of Cardiovascular Medicine, Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, China
| | - Yeshi Chen
- Department of Cardiovascular Medicine, Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, China
| | - Chenhui Feng
- Department of Cardiovascular Medicine, Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, China
| | - Mingming Zhang
- Department of Cardiovascular Medicine, Children’s Hospital Capital Institute of Pediatrics, Beijing, China
| | - Hongmao Wang
- Department of Cardiovascular Medicine, Children’s Hospital Capital Institute of Pediatrics, Beijing, China
| | - Yang Zheng
- Department of Cardiovascular Medicine, Peking Union Medical College Graduate School, Beijing, China
| | - Xiaohui Li
- Department of Cardiovascular Medicine, Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, China
- Department of Cardiovascular Medicine, Children’s Hospital Capital Institute of Pediatrics, Beijing, China
- Department of Cardiovascular Medicine, Peking Union Medical College Graduate School, Beijing, China
| |
Collapse
|
11
|
Tevlek A. Diagnostic use of circulating cells and sub-cellular bio-particles. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 192:19-36. [PMID: 39159788 DOI: 10.1016/j.pbiomolbio.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/22/2024] [Accepted: 08/11/2024] [Indexed: 08/21/2024]
Abstract
In the bloodstream or other physiological fluids, "circulating cells and sub-cellular bio-particles" include many microscopic biological elements such as circulating tumor cells (CTCs), cell-free DNA (cfDNA), exosomes, microRNAs, platelets, immune cells, and proteins are the most well-known and investigated. These structures are crucial biomarkers in healthcare and medical research for the early detection of cancer and other disorders, enabling treatment to commence before the onset of clinical symptoms and enhancing the efficacy of treatments. As the size of these biomarkers to be detected decreases and their numbers in body fluids diminishes, the detection materials, ranging from visual inspection to advanced microscopy techniques, begin to become smaller, more sensitive, faster, and more effective, thanks to developing nanotechnology. This review first defines the circulating cells and subcellular bio-particles with their biological, physical, and mechanical properties and second focuses on their diagnostic importance, including their most recent applications as biomarkers, the biosensors that are utilized to detect them, the present obstacles that must be surmounted, and prospective developments in the domain. As technology advances and biomolecular pathways are deepens, diagnostic tests will become more sensitive, specific, and thorough. Finally, integrating recent advances in the diagnostic use of circulating cells and bioparticles into clinical practice is promising for precision medicine and patient outcomes.
Collapse
Affiliation(s)
- Atakan Tevlek
- Department of Medical Biology, Faculty of Medicine, Atilim University, Ankara, 06836, Turkey.
| |
Collapse
|
12
|
Gambino CM, Agnello L, Vidali M, Lo Sasso B, Mansueto P, Seidita A, Giuliano A, Scazzone C, Massa D, Masucci A, Tamburello M, Vassallo R, Ciaccio AM, Candore G, Carroccio A, Ciaccio M. The role of Killer immunoglobulin-like receptors (KIRs) in the genetic susceptibility to non-celiac wheat sensitivity (NCWS). Clin Chem Lab Med 2024; 62:1814-1823. [PMID: 38639193 DOI: 10.1515/cclm-2024-0034] [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: 01/10/2024] [Accepted: 04/07/2024] [Indexed: 04/20/2024]
Abstract
OBJECTIVES Non-celiac wheat sensitivity (NCWS) is an emerging clinical condition characterized by gastrointestinal and extraintestinal symptoms following the ingestion of gluten-containing foods in patients without celiac disease (CD) or wheat allergy. Despite the great interest for NCWS, the genetic risk factors still need to be fully clarified. In this study, we first assessed the possible contribution of KIR genes and KIR haplotypes on the genetic predisposition to NCWS. METHODS Fifty patients with NCWS, 50 patients with CD, and 50 healthy controls (HC) were included in this study. KIR genes and KIR genotyping were investigated in all subjects by polymerase chain reaction with the sequence oligonucleotide probe (PCR-SSOP) method using Luminex technology. RESULTS We found a statistically different distribution of some KIR genes among NCWS, CD, and HC. Specifically, NCWS showed a decreased frequency of KIR2DL1, -2DL3, -2DL5, -2DS2, -2DS3, -2DS4, -2DS5, and -3DS1 genes, and an increased frequency of -3DL1 gene respect to both CD and HC. No difference was detected in the KIR haplotype expression. At the multivariate analysis, KIR2DL5, -2DS4, and -2DS5 were independent predictors of NCWS. CONCLUSIONS Our findings suggest a role of KIR genes in NCWS susceptibility, with KIR2DL5, -2DS4, and -2DS5 having a protective effect. Further large-scale multicentric studies are required to validate these preliminary findings.
Collapse
Affiliation(s)
- Caterina Maria Gambino
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, 18998 Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, University of Palermo , Palermo, Italy
- Department of Laboratory Medicine, University Hospital "P. Giaccone", Palermo, Italy
| | - Luisa Agnello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, 18998 Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, University of Palermo , Palermo, Italy
| | - Matteo Vidali
- Clinical Pathology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Bruna Lo Sasso
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, 18998 Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, University of Palermo , Palermo, Italy
- Department of Laboratory Medicine, University Hospital "P. Giaccone", Palermo, Italy
| | - Pasquale Mansueto
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine, and Medical Specialties (PROMISE), Unit of Internal Medicine, 18998 University of Palermo , Palermo, Italy
| | - Aurelio Seidita
- Unit of Internal Medicine, "V. Cervello" Hospital, Ospedali Riuniti "Villa Sofia-Cervello", Palermo, Italy
| | - Alessandra Giuliano
- Unit of Internal Medicine, "V. Cervello" Hospital, Ospedali Riuniti "Villa Sofia-Cervello", Palermo, Italy
| | - Concetta Scazzone
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, 18998 Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, University of Palermo , Palermo, Italy
| | - Davide Massa
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, 18998 Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, University of Palermo , Palermo, Italy
| | - Anna Masucci
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, 18998 Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, University of Palermo , Palermo, Italy
| | - Martina Tamburello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, 18998 Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, University of Palermo , Palermo, Italy
| | - Roberta Vassallo
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, 18998 Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, University of Palermo , Palermo, Italy
| | - Anna Maria Ciaccio
- Department of Health Promotion, Maternal and Child Health, Internal Medicine, and Specialty Excellence "G. D'Alessandro" (PROMISE), Internal Medicine and Stroke Care Ward, University of Palermo, Palermo, Italy
| | - Giuseppina Candore
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, 18998 Laboratory of Immunopathology and Immunosenescence, University of Palermo , Palermo, Italy
| | - Antonio Carroccio
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine, and Medical Specialties (PROMISE), Unit of Internal Medicine, 18998 University of Palermo , Palermo, Italy
- Unit of Internal Medicine, "V. Cervello" Hospital, Ospedali Riuniti "Villa Sofia-Cervello", Palermo, Italy
| | - Marcello Ciaccio
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, 18998 Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Clinical Laboratory Medicine, University of Palermo , Palermo, Italy
- Department of Laboratory Medicine, University Hospital "P. Giaccone", Palermo, Italy
| |
Collapse
|
13
|
Wang N, Wang G, Feng X, Yang T. Identification of Immune Gene Signature Associated with T Cells and Natural Killer Cells in Type 1 Diabetes. Diabetes Metab Syndr Obes 2024; 17:2983-2996. [PMID: 39139741 PMCID: PMC11321349 DOI: 10.2147/dmso.s470738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/23/2024] [Indexed: 08/15/2024] Open
Abstract
Purpose This study aimed to investigate the abnormal infiltration of immune cells in type 1 diabetes mellitus (T1D) and elucidate their regulatory mechanisms. Methods Public T1D-related gene expression data were obtained from the Gene Expression Omnibus database.The GSE123658 dataset analyzed whole blood RNA-seq data from type 1 diabetic patients and healthy volunteers. The GSE110914 dataset analyzed neutrophils purified from peripheral blood of patients with symptomatic and pre-symptomatic type 1 diabetes (T1D), at risk of T1D, and healthy controls. Immune cell infiltration analysis was performed to identify abnormally infiltrating immune cells. Differentially expressed immune genes (DEIGs) in T1D samples were identified, followed by the construction of an immune gene signature (IGS) using a protein-protein interaction (PPI) network and Least absolute shrinkage and selection operator Cox regression analyses (LASSO Cox regression analyses). The regulatory mechanisms underlying IGS were explored using gene set enrichment analysis. Furthermore, expression validation, diagnostic efficacy evaluation, and upstream miRNA prediction of hub signature genes were performed. We verified the miRNA expression of the key gene colony stimulating factor 1 (CSF1) and microRNA-326 (miR-326) by reverse transcription-quantitative PCR (RT‒qPCR). Results The proportion of infiltrating T and natural killer (NK) cells differed between the T1D and control samples, and 207 immune genes (IGs) related to these immune cells were extracted. After differential expression, PPI, and LASSO Cox regression analyses, four signature DEIGs were identified for IGS construction: notch receptor 1 (NOTCH1), Janus kinase 3 (JAK3), tumor necrosis factor receptor superfamily member 4(TNFRSF4), and CSF1. Key pathways such as the Toll-like receptor signaling pathway were significantly activated in the high-risk group. Moreover, the upregulation of CSF1 in T1D samples was confirmed using a validation dataset, and CSF1 showed high diagnostic efficacy for T1D. Furthermore, CSF1 was targeted by miR-326.We used validated key genes in T1D patients, several of which were confirmed by RT‒qPCR. Conclusion In conclusion, the identified key IGs may play an important role in T1D. CSF1 can be developed as a novel diagnostic biomarker for T1D.
Collapse
Affiliation(s)
- Na Wang
- Department of Endocrinology, Lianyungang Clinical College of Nanjing Medical University, Lianyungang City, Jiangsu Province, 222000, People’s Republic of China
- Department of Endocrinology, Jinzhou Medical University(The First People’s Hospital of Lianyungang), Lianyungang City, Jiangsu Province, 222000, People’s Republic of China
| | - Guofeng Wang
- Department of Endocrinology, Jinzhou Medical University(The First People’s Hospital of Lianyungang), Lianyungang City, Jiangsu Province, 222000, People’s Republic of China
| | - Xiuli Feng
- Department of Endocrinology, Jinzhou Medical University(The First People’s Hospital of Lianyungang), Lianyungang City, Jiangsu Province, 222000, People’s Republic of China
| | - Teng Yang
- Department of Endocrinology, Jinzhou Medical University(The First People’s Hospital of Lianyungang), Lianyungang City, Jiangsu Province, 222000, People’s Republic of China
| |
Collapse
|
14
|
Wang D, Dou L, Sui L, Xue Y, Xu S. Natural killer cells in cancer immunotherapy. MedComm (Beijing) 2024; 5:e626. [PMID: 38882209 PMCID: PMC11179524 DOI: 10.1002/mco2.626] [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: 12/21/2023] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 06/18/2024] Open
Abstract
Natural killer (NK) cells, as innate lymphocytes, possess cytotoxic capabilities and engage target cells through a repertoire of activating and inhibitory receptors. Particularly, natural killer group 2, member D (NKG2D) receptor on NK cells recognizes stress-induced ligands-the MHC class I chain-related molecules A and B (MICA/B) presented on tumor cells and is key to trigger the cytolytic response of NK cells. However, tumors have developed sophisticated strategies to evade NK cell surveillance, which lead to failure of tumor immunotherapy. In this paper, we summarized these immune escaping strategies, including the downregulation of ligands for activating receptors, upregulation of ligands for inhibitory receptors, secretion of immunosuppressive compounds, and the development of apoptosis resistance. Then, we focus on recent advancements in NK cell immune therapies, which include engaging activating NK cell receptors, upregulating NKG2D ligand MICA/B expression, blocking inhibitory NK cell receptors, adoptive NK cell therapy, chimeric antigen receptor (CAR)-engineered NK cells (CAR-NK), and NKG2D CAR-T cells, especially several vaccines targeting MICA/B. This review will inspire the research in NK cell biology in tumor and provide significant hope for improving cancer treatment outcomes by harnessing the potent cytotoxic activity of NK cells.
Collapse
Affiliation(s)
- DanRu Wang
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - LingYun Dou
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - LiHao Sui
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - Yiquan Xue
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
| | - Sheng Xu
- National Key Lab of Immunity and Inflammation and Institute of Immunology Naval Medical University Shanghai China
- Shanghai Institute of Stem Cell Research and Clinical Translation Dongfang Hospital Shanghai China
| |
Collapse
|
15
|
Santiago-Lamelas L, Castro-Santos P, Carracedo Á, Olloquequi J, Díaz-Peña R. Unveiling the Significance of HLA and KIR Diversity in Underrepresented Populations. Biomedicines 2024; 12:1333. [PMID: 38927540 PMCID: PMC11202227 DOI: 10.3390/biomedicines12061333] [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: 04/29/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Human leukocyte antigen (HLA) molecules and their relationships with natural killer (NK) cells, specifically through their interaction with killer-cell immunoglobulin-like receptors (KIRs), exhibit robust associations with the outcomes of diverse diseases. Moreover, genetic variations in HLA and KIR immune system genes offer limitless depths of complexity. In recent years, a surge of high-powered genome-wide association studies (GWASs) utilizing single nucleotide polymorphism (SNP) arrays has occurred, significantly advancing our understanding of disease pathogenesis. Additionally, advances in HLA reference panels have enabled higher resolution and more reliable imputation, allowing for finer-grained evaluation of the association between sequence variations and disease risk. However, it is essential to note that the majority of these GWASs have focused primarily on populations of Caucasian and Asian origins, neglecting underrepresented populations in Latin America and Africa. This omission not only leads to disparities in health care access but also restricts our knowledge of novel genetic variants involved in disease pathogenesis within these overlooked populations. Since the KIR and HLA haplotypes prevalent in each population are clearly modelled by the specific environment, the aim of this review is to encourage studies investigating HLA/KIR involvement in infection and autoimmune diseases, reproduction, and transplantation in underrepresented populations.
Collapse
Affiliation(s)
- Lucía Santiago-Lamelas
- Fundación Pública Galega de Medicina Xenómica, SERGAS, Grupo de Medicina Xenomica-USC, Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain; (L.S.-L.); (P.C.-S.); (Á.C.)
| | - Patricia Castro-Santos
- Fundación Pública Galega de Medicina Xenómica, SERGAS, Grupo de Medicina Xenomica-USC, Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain; (L.S.-L.); (P.C.-S.); (Á.C.)
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3460000, Chile
| | - Ángel Carracedo
- Fundación Pública Galega de Medicina Xenómica, SERGAS, Grupo de Medicina Xenomica-USC, Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain; (L.S.-L.); (P.C.-S.); (Á.C.)
- Grupo de Medicina Xenómica, CIMUS, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jordi Olloquequi
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3460000, Chile
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Roberto Díaz-Peña
- Fundación Pública Galega de Medicina Xenómica, SERGAS, Grupo de Medicina Xenomica-USC, Instituto de Investigación Sanitaria de Santiago (IDIS), 15706 Santiago de Compostela, Spain; (L.S.-L.); (P.C.-S.); (Á.C.)
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3460000, Chile
| |
Collapse
|
16
|
Sun HW, Zhang X, Shen CC. The shared circulating diagnostic biomarkers and molecular mechanisms of systemic lupus erythematosus and inflammatory bowel disease. Front Immunol 2024; 15:1354348. [PMID: 38774864 PMCID: PMC11106441 DOI: 10.3389/fimmu.2024.1354348] [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: 12/12/2023] [Accepted: 04/22/2024] [Indexed: 05/24/2024] Open
Abstract
Background Systemic lupus erythematosus (SLE) is a multi-organ chronic autoimmune disease. Inflammatory bowel disease (IBD) is a common chronic inflammatory disease of the gastrointestinal tract. Previous studies have shown that SLE and IBD share common pathogenic pathways and genetic susceptibility, but the specific pathogenic mechanisms remain unclear. Methods The datasets of SLE and IBD were downloaded from the Gene Expression Omnibus (GEO). Differentially expressed genes (DEGs) were identified using the Limma package. Weighted gene coexpression network analysis (WGCNA) was used to determine co-expression modules related to SLE and IBD. Pathway enrichment was performed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis for co-driver genes. Using the Least AbsoluteShrinkage and Selection Operator (Lasso) regressionand Support Vector Machine-Recursive Feature Elimination (SVM-RFE), common diagnostic markers for both diseases were further evaluated. Then, we utilizedthe CIBERSORT method to assess the abundance of immune cell infiltration. Finally,we used the single-cell analysis to obtain the location of common diagnostic markers. Results 71 common driver genes were identified in the SLE and IBD cohorts based on the DEGs and module genes. KEGG and GO enrichment results showed that these genes were closely associated with positive regulation of programmed cell death and inflammatory responses. By using LASSO regression and SVM, five hub genes (KLRF1, GZMK, KLRB1, CD40LG, and IL-7R) were ultimately determined as common diagnostic markers for SLE and IBD. ROC curve analysis also showed good diagnostic performance. The outcomes of immune cell infiltration demonstrated that SLE and IBD shared almost identical immune infiltration patterns. Furthermore, the majority of the hub genes were commonly expressed in NK cells by single-cell analysis. Conclusion This study demonstrates that SLE and IBD share common diagnostic markers and pathogenic pathways. In addition, SLE and IBD show similar immune cellinfiltration microenvironments which provides newperspectives for future treatment.
Collapse
Affiliation(s)
- Hao-Wen Sun
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Xin Zhang
- Department of Dermatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Cong-Cong Shen
- Department of Dermatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| |
Collapse
|
17
|
Huang W, Jin T, Zheng W, Yin Q, Yan Q, Pan H, Xu C. Identifying the genetic association between systemic lupus erythematosus and the risk of autoimmune liver diseases. J Autoimmun 2024; 145:103188. [PMID: 38458076 DOI: 10.1016/j.jaut.2024.103188] [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: 12/13/2023] [Revised: 02/14/2024] [Accepted: 02/18/2024] [Indexed: 03/10/2024]
Abstract
BACKGROUND Previous studies on the relationship between systemic lupus erythematosus (SLE) and autoimmune liver diseases (AILDs) are inconclusive. Therefore, we employed Mendelian randomization (MR) to explore the causal associations between SLE and AILDs. METHODS A two-sample MR analysis was performed using summary-level statistics sourced from genome-wide association study (GWAS) datasets. Inverse-variance weighting (IVW), MR‒Egger, and weighted median (WM) were further supported by several sensitivity analyses. RESULTS We detected causal genetic associations between SLE and primary biliary cholangitis (PBC) (odds ratio (OR) = 1.31, 95% CI = 1.15-1.51, P < 0.01; adjusted OR = 1.63, 95% CI = 1.39-1.90, P < 0.01) and between SLE and primary sclerosing cholangitis (PSC) (OR = 1.09, 95% CI = 1.01-1.08, P = 0.03; adjusted OR = 1.10, 95% CI = 1.00-1.21, P = 0.04). No causal association was found between SLE and autoimmune hepatitis. CONCLUSIONS We are the first to use MR analysis to explore the causal relationships between SLE and various AILDs, revealing an increased risk of PBC and PSC in individuals with SLE.
Collapse
MESH Headings
- Humans
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/epidemiology
- Genome-Wide Association Study
- Genetic Predisposition to Disease
- Mendelian Randomization Analysis
- Polymorphism, Single Nucleotide
- Hepatitis, Autoimmune/genetics
- Hepatitis, Autoimmune/epidemiology
- Liver Cirrhosis, Biliary/genetics
- Liver Cirrhosis, Biliary/epidemiology
- Liver Cirrhosis, Biliary/etiology
- Cholangitis, Sclerosing/genetics
- Cholangitis, Sclerosing/epidemiology
- Autoimmune Diseases/genetics
- Autoimmune Diseases/epidemiology
- Autoimmune Diseases/etiology
- Odds Ratio
- Risk Factors
- Liver Diseases/genetics
- Liver Diseases/epidemiology
- Liver Diseases/etiology
Collapse
Affiliation(s)
- Wei Huang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China; Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Tianyu Jin
- Department of Rehabilitation Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Wei Zheng
- Center for General Practice Medicine, Department of Infectious Diseases, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China.
| | - Qiaoqiao Yin
- Center for General Practice Medicine, Department of Infectious Diseases, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China.
| | - Qiqi Yan
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Hongying Pan
- Center for General Practice Medicine, Department of Infectious Diseases, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China.
| | - Chengan Xu
- Center for General Practice Medicine, Department of Infectious Diseases, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China.
| |
Collapse
|
18
|
Kamyan D, Hassane M, Alnaqbi A, Souid AK, Rasbi ZA, Tahrawi AA, Shamsi MA. Ozanimod-mediated remission in experimental autoimmune encephalomyelitis is associated with enhanced activity of CNS CD27 low/- NK cell subset. Front Immunol 2024; 15:1230735. [PMID: 38533505 PMCID: PMC10963535 DOI: 10.3389/fimmu.2024.1230735] [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: 06/01/2023] [Accepted: 01/30/2024] [Indexed: 03/28/2024] Open
Abstract
Background Ozanimod (RPC1063) is an immunomodulator that has been recently approved by the FDA (2020) for the treatment of relapsing-remitting multiple sclerosis (RRMS). It is a selective agonist of the sphingosine-1-phophate receptors 1 and 5, expressed on naïve and central memory T and B cells, as well as natural killer (NK) cells, and is involved in lymphocyte trafficking. Oral administration of ozanimod was reported to result in rapid and reversible reduction in circulating lymphocytes in multiple sclerosis (MS) patients, however, only minimal effect on NK cells was observed. In this study, we sought to investigate the effect of ozanimod on NK cells and assess whether they play any role in ozanimod-induced remission in experimental autoimmune encephalomyelitis (EAE), the animal model of MS. Methods Active EAE induction was done in C57BL/6 female mice, followed by daily oral treatment with ozanimod (0.6mg/kg) starting at disease onset (score 1). Flow cytometry of blood and CNS was performed 24 hours after the last oral dose of ozanimod treatment in diseased mice. Histological analysis of lumbar spinal cord was performed for evaluating the level of inflammation and demyelination. Depletion of peripheral NK cells was done using anti-NK1.1 mouse antibody (mAb) at day 5 post-EAE induction. Results Ozanimod was effective in reducing the clinical severity of EAE and reducing the percentage of autoreactive CD4+ and CD8+ T cells along with significant inhibition of lymphocyte infiltration into the spinal cord, accompanied by reversed demyelination. Furthermore, ozanimod treatment resulted in a significant increase in the frequency of total NK cells in the blood and CNS along with upregulation of the activating receptor NKG2D on CD27low/- NK cell subset in the CNS. The effectiveness of ozanimod treatment in inhibiting the progression of the disease was reduced when NK cells were depleted using anti-NK1.1 mAb. Conclusion The current study demonstrated that ozanimod treatment significantly improved clinical symptoms in EAE mice. Ozanimod and anti-NK1.1 mAb appear to function in opposition to one another. Collectively, our data suggest that ozanimod-mediated remission is associated with an increased percentage of total NK cells and CD27low/- NK cells expressing the activating receptor, NKG2D in the CNS.
Collapse
Affiliation(s)
- Doua Kamyan
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates (UAE) University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Maya Hassane
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates (UAE) University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Alanood Alnaqbi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates (UAE) University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Abdul-Kader Souid
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates (UAE) University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Zakeya Al Rasbi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates (UAE) University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Abeer Al Tahrawi
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates (UAE) University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Mariam Al Shamsi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates (UAE) University, Al Ain, Abu Dhabi, United Arab Emirates
| |
Collapse
|
19
|
Padilla CM, Valenzi E, Tabib T, Nazari B, Sembrat J, Rojas M, Fuschiotti P, Lafyatis R. Increased CD8+ tissue resident memory T cells, regulatory T cells and activated natural killer cells in systemic sclerosis lungs. Rheumatology (Oxford) 2024; 63:837-845. [PMID: 37310903 PMCID: PMC10907815 DOI: 10.1093/rheumatology/kead273] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 05/10/2023] [Accepted: 05/15/2023] [Indexed: 06/15/2023] Open
Abstract
OBJECTIVE Multiple observations indicate a role for lymphocytes in driving autoimmunity in SSc. While T and NK cells have been studied in SSc whole blood and bronchoalveolar lavage fluid, their role remains unclear, partly because no studies have analysed these cell types in SSc-interstitial lung disease (ILD) lung tissue. This research aimed to identify and analyse the lymphoid subpopulations in SSc-ILD lung explants. METHODS Lymphoid populations from 13 SSc-ILD and 6 healthy control (HC) lung explants were analysed using Seurat following single-cell RNA sequencing. Lymphoid clusters were identified by their differential gene expression. Absolute cell numbers and cell proportions in each cluster were compared between cohorts. Additional analyses were performed using pathway analysis, pseudotime and cell ligand-receptor interactions. RESULTS Activated CD16+ NK cells, CD8+ tissue resident memory T cells and Treg cells were proportionately higher in SSc-ILD compared with HC lungs. Activated CD16+ NK cells in SSc-ILD showed upregulated granzyme B, IFN-γ and CD226. Amphiregulin, highly upregulated by NK cells, was predicted to interact with epidermal growth factor receptor on several bronchial epithelial cell populations. Shifts in CD8+ T cell populations indicated a transition from resting to effector to tissue resident phenotypes in SSc-ILD. CONCLUSIONS SSc-ILD lungs show activated lymphoid populations. Activated cytotoxic NK cells suggest they may kill alveolar epithelial cells, while their expression of amphiregulin suggests they may also induce bronchial epithelial cell hyperplasia. CD8+ T cells in SSc-ILD appear to transition from resting to the tissue resident memory phenotype.
Collapse
Affiliation(s)
- Cristina M Padilla
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eleanor Valenzi
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tracy Tabib
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Banafsheh Nazari
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - John Sembrat
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mauricio Rojas
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Ohio State University College of Medicine, Columbus, OH, USA
| | - Patrizia Fuschiotti
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Robert Lafyatis
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
20
|
Saadh MJ, Rasulova I, Khalil M, Farahim F, Sârbu I, Ciongradi CI, Omar TM, Alhili A, Jawad MJ, Hani T, Ali T, Lakshmaiya N. Natural killer cell-mediated immune surveillance in cancer: Role of tumor microenvironment. Pathol Res Pract 2024; 254:155120. [PMID: 38280274 DOI: 10.1016/j.prp.2024.155120] [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: 11/28/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 01/29/2024]
Abstract
In the immunological surveillance against cancer, natural killer (NK) cells are essential effectors that help eradicate altered cells. The complex interactions that occur between NK cells and the tumor microenvironment (TME) are thoroughly examined in this review. The review examines how cytokine stimulation affects NK cell activation, focusing on the dynamic modulation of NK cell function within the TME. It looks at NK cell-related biomarkers such as PD-1/PD-L1, methylation HOXA9 (Homeobox A9), Stroma AReactive Invasion Front Areas (SARIFA), and NKG2A/HLA-E, providing critical information about prognosis and treatment outcomes. The changing landscape of immunotherapies-including checkpoint inhibitors, CAR-NK cells, and cytokine-based interventions-is examined in the context of enhancing NK cell activity. The review highlights the potential pathways for precision medicine going forward, focusing on customized immunotherapies based on unique biomarker profiles and investigating combination medicines to produce more robust anti-tumor responses.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan; Applied Science Research Center. Applied Science Private University, Amman, Jordan
| | - Irodakhon Rasulova
- MD, PhD, Senior Researcher, School of Humanities, Natural & Social Sciences, New Uzbekistan University, 54 Mustaqillik Ave., Tashkent, 100007, Uzbekistan; Department of Public Health, Samarkand State Medical University, Amir Temur street 18, Samarkand, Uzbekistan
| | | | - Farha Farahim
- Department of Nursing, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Ioan Sârbu
- 2nd Department of Surgery-Pediatric Surgery and Orthopedics, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania.
| | - Carmen Iulia Ciongradi
- 2nd Department of Surgery-Pediatric Surgery and Orthopedics, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania.
| | - Thabit Moath Omar
- Department of Medical Laboratory Technics, Al-Noor University College, Nineveh, Iraq
| | - Ahmed Alhili
- Medical Technical College, Al-Farahidi University, Iraq
| | | | - Thamer Hani
- Dentistry Department, Al-Turath University College, Baghdad, Iraq
| | - Talat Ali
- Department of Basic Medical Sciences, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Natrayan Lakshmaiya
- Department of Mechanical Engineering, Saveetha School of Engineering, SIMATS, Chennai, Tamil Nadu, India
| |
Collapse
|
21
|
Jucá PM, de Almeida Duque É, Covre LHH, Mariano KAA, Munhoz CD. Microglia and Systemic Immunity. ADVANCES IN NEUROBIOLOGY 2024; 37:287-302. [PMID: 39207698 DOI: 10.1007/978-3-031-55529-9_16] [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: 09/04/2024]
Abstract
Microglia are specialized immune cells that reside in the central nervous system (CNS) and play a crucial role in maintaining the homeostasis of the brain microenvironment. While traditionally regarded as a part of the innate immune system, recent research has highlighted their role in adaptive immunity. The CNS is no longer considered an immune-privileged organ, and increasing evidence suggests bidirectional communication between the immune system and the CNS. Microglia are sensitive to systemic immune signals and can respond to systemic inflammation by producing various inflammatory cytokines and chemokines. This response is mediated by activating pattern recognition receptors (PRRs), which recognize pathogen- and danger-associated molecular patterns in the systemic circulation. The microglial response to systemic inflammation has been implicated in several neurological conditions, including depression, anxiety, and cognitive impairment. Understanding the complex interplay between microglia and systemic immunity is crucial for developing therapeutic interventions to modulate immune responses in the CNS.
Collapse
Affiliation(s)
- Paloma Marinho Jucá
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, São Paulo, Brazil
| | - Érica de Almeida Duque
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, São Paulo, Brazil
| | - Luiza Helena Halas Covre
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, São Paulo, Brazil
| | | | - Carolina Demarchi Munhoz
- Department of Pharmacology, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, São Paulo, Brazil.
| |
Collapse
|
22
|
Wang L, Mao L, Xiao W, Chen P. Natural killer cells immunosenescence and the impact of lifestyle management. Biochem Biophys Res Commun 2023; 689:149216. [PMID: 37976836 DOI: 10.1016/j.bbrc.2023.149216] [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: 09/03/2023] [Revised: 10/28/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
Natural killer cells (NKs) are lymphocytes of the innate immune system that quickly respond to viruses, infections, and tumors during their short cell life cycle. However, it was recently found that NKs undergo quantitative, distributional, structural, and functional phenotypic changes during aging that suppress immune responses, which is known as immunosenescence. The aging host environment, cytokine regulation, cytomegalovirus status, and hypothalamic‒pituitary‒adrenal axis have significant effects on NK function. Different lifestyle management interventions modulate the number and cytotoxic activity of NKs, which are essential for rebuilding the immune barrier against pathogens in elderly individuals. Based on recent studies, we review the phenotypic changes of and potential threats of NKs during aging and explore the underlying mechanisms. By summarizing the effects of lifestyle management on NKs and their application prospects, we aim to provide evidence for enhancing immune system function against immune diseases in elderly individuals.
Collapse
Affiliation(s)
- Lian Wang
- The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, 200438, China; Shanghai Key Lab of Human Performance, Shanghai University of Sport, Shanghai, 200438, China.
| | - Liwei Mao
- The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, 200438, China; Shanghai Key Lab of Human Performance, Shanghai University of Sport, Shanghai, 200438, China.
| | - Weihua Xiao
- The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, 200438, China; Shanghai Key Lab of Human Performance, Shanghai University of Sport, Shanghai, 200438, China.
| | - Peijie Chen
- The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, 200438, China; Shanghai Key Lab of Human Performance, Shanghai University of Sport, Shanghai, 200438, China.
| |
Collapse
|
23
|
Connors J, Cusimano G, Mege N, Woloszczuk K, Konopka E, Bell M, Joyner D, Marcy J, Tardif V, Kutzler MA, Muir R, Haddad EK. Using the power of innate immunoprofiling to understand vaccine design, infection, and immunity. Hum Vaccin Immunother 2023; 19:2267295. [PMID: 37885158 PMCID: PMC10760375 DOI: 10.1080/21645515.2023.2267295] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/03/2023] [Indexed: 10/28/2023] Open
Abstract
In the field of immunology, a systems biology approach is crucial to understanding the immune response to infection and vaccination considering the complex interplay between genetic, epigenetic, and environmental factors. Significant progress has been made in understanding the innate immune response, including cell players and critical signaling pathways, but many questions remain unanswered, including how the innate immune response dictates host/pathogen responses and responses to vaccines. To complicate things further, it is becoming increasingly clear that the innate immune response is not a linear pathway but is formed from complex networks and interactions. To further our understanding of the crosstalk and complexities, systems-level analyses and expanded experimental technologies are now needed. In this review, we discuss the most recent immunoprofiling techniques and discuss systems approaches to studying the global innate immune landscape which will inform on the development of personalized medicine and innovative vaccine strategies.
Collapse
Affiliation(s)
- Jennifer Connors
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Gina Cusimano
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Nathan Mege
- Tower Health, Reading Hospital, West Reading, PA, USA
| | - Kyra Woloszczuk
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Emily Konopka
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Matthew Bell
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - David Joyner
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Molecular and Cellular Biology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Jennifer Marcy
- Department of Molecular and Cellular Biology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Virginie Tardif
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Michele A. Kutzler
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Roshell Muir
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Family, Community, and Preventative Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Elias K. Haddad
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA
- Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
24
|
Sánchez‐Cerrillo I, Calzada‐Fraile D, Triguero‐Martínez A, Calvet‐Mirabent M, Popova O, Delgado‐Arévalo C, Valdivia‐Mazeyra M, Ramírez‐Huesca M, de Luis EV, Benguría A, Aceña‐Gonzalo T, Moreno‐Vellisca R, de Llano MA, de la Fuente H, Tsukalov I, Delgado‐Wicke P, Fernández‐Ruiz E, Roy‐Vallejo E, Tejedor‐Lázaro R, Ramiro A, Iborra S, Sánchez‐Madrid F, Dopazo A, Álvaro IG, Castañeda S, Martin‐Gayo E. MICa/b-dependent activation of natural killer cells by CD64 + inflammatory type 2 dendritic cells contributes to autoimmunity. EMBO J 2023; 42:e113714. [PMID: 37916875 PMCID: PMC10690448 DOI: 10.15252/embj.2023113714] [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: 02/08/2023] [Revised: 09/27/2023] [Accepted: 10/05/2023] [Indexed: 11/03/2023] Open
Abstract
Primary Sjögren's syndrome (pSS) is an inflammatory autoimmune disorder largely mediated by type I and II interferon (IFN). The potential contribution of innate immune cells, such as natural killer (NK) cells and dendritic cells (DC), to the pSS pathology remains understudied. Here, we identified an enriched CD16+ CD56hi NK cell subset associated with higher cytotoxic function, as well as elevated proportions of inflammatory CD64+ conventional dendritic cell (cDC2) subtype that expresses increased levels of MICa/b, the ligand for the activating receptor NKG2D, in pSS individuals. Circulating cDC2 from pSS patients efficiently induced activation of cytotoxic NK cells ex vivo and were found in proximity to CD56+ NK cells in salivary glands (SG) from pSS patients. Interestingly, transcriptional activation of IFN signatures associated with the RIG-I/DDX60 pathway, IFN I receptor, and its target genes regulate the expression of NKG2D ligands on cDC2 from pSS patients. Finally, increased proportions of CD64hi RAE-1+ cDC2 and NKG2D+ CD11b+ CD27+ NK cells were present in vivo in the SG after poly I:C injection. Our study provides novel insight into the contribution and interplay of NK and cDC2 in pSS pathology and identifies new potential therapy targets.
Collapse
Affiliation(s)
- Ildefonso Sánchez‐Cerrillo
- Immunology UnitHospital Universitario La Princesa, Medicine Department, Universidad Autónoma de Madrid, Instituto Investigación Sanitaria‐Princesa IIS‐IPMadridSpain
| | - Diego Calzada‐Fraile
- Immunology UnitHospital Universitario La Princesa, Medicine Department, Universidad Autónoma de Madrid, Instituto Investigación Sanitaria‐Princesa IIS‐IPMadridSpain
- Vascular Pathophysiology DepartmentCentro Nacional de Investigaciones CardiovascularesMadridSpain
| | - Ana Triguero‐Martínez
- Rheumatology UnitHospital Universitario La Princesa, Instituto de Investigación Sanitaria‐Princesa IIS‐IPMadridSpain
| | - Marta Calvet‐Mirabent
- Immunology UnitHospital Universitario La Princesa, Medicine Department, Universidad Autónoma de Madrid, Instituto Investigación Sanitaria‐Princesa IIS‐IPMadridSpain
| | - Olga Popova
- Immunology UnitHospital Universitario La Princesa, Medicine Department, Universidad Autónoma de Madrid, Instituto Investigación Sanitaria‐Princesa IIS‐IPMadridSpain
| | - Cristina Delgado‐Arévalo
- Immunology UnitHospital Universitario La Princesa, Medicine Department, Universidad Autónoma de Madrid, Instituto Investigación Sanitaria‐Princesa IIS‐IPMadridSpain
| | | | - Marta Ramírez‐Huesca
- Vascular Pathophysiology DepartmentCentro Nacional de Investigaciones CardiovascularesMadridSpain
| | | | - Alberto Benguría
- Genomic UnitCentro Nacional de Investigaciones CardiovascularesMadridSpain
| | - Teresa Aceña‐Gonzalo
- Immunology UnitHospital Universitario La Princesa, Medicine Department, Universidad Autónoma de Madrid, Instituto Investigación Sanitaria‐Princesa IIS‐IPMadridSpain
| | | | | | - Hortensia de la Fuente
- Immunology UnitHospital Universitario La Princesa, Medicine Department, Universidad Autónoma de Madrid, Instituto Investigación Sanitaria‐Princesa IIS‐IPMadridSpain
- CIBER Cardiovascular, Instituto de Salud Carlos IIIMadridSpain
| | - Ilya Tsukalov
- Immunology UnitHospital Universitario La Princesa, Medicine Department, Universidad Autónoma de Madrid, Instituto Investigación Sanitaria‐Princesa IIS‐IPMadridSpain
| | - Pablo Delgado‐Wicke
- Immunology UnitHospital Universitario La Princesa, Medicine Department, Universidad Autónoma de Madrid, Instituto Investigación Sanitaria‐Princesa IIS‐IPMadridSpain
| | - Elena Fernández‐Ruiz
- Immunology UnitHospital Universitario La Princesa, Medicine Department, Universidad Autónoma de Madrid, Instituto Investigación Sanitaria‐Princesa IIS‐IPMadridSpain
| | - Emilia Roy‐Vallejo
- Rheumatology UnitHospital Universitario La Princesa, Instituto de Investigación Sanitaria‐Princesa IIS‐IPMadridSpain
| | - Reyes Tejedor‐Lázaro
- Immunology UnitHospital Universitario La Princesa, Medicine Department, Universidad Autónoma de Madrid, Instituto Investigación Sanitaria‐Princesa IIS‐IPMadridSpain
| | - Almudena Ramiro
- Vascular Pathophysiology DepartmentCentro Nacional de Investigaciones CardiovascularesMadridSpain
| | - Salvador Iborra
- Vascular Pathophysiology DepartmentCentro Nacional de Investigaciones CardiovascularesMadridSpain
| | - Francisco Sánchez‐Madrid
- Immunology UnitHospital Universitario La Princesa, Medicine Department, Universidad Autónoma de Madrid, Instituto Investigación Sanitaria‐Princesa IIS‐IPMadridSpain
- Vascular Pathophysiology DepartmentCentro Nacional de Investigaciones CardiovascularesMadridSpain
- CIBER Cardiovascular, Instituto de Salud Carlos IIIMadridSpain
| | - Ana Dopazo
- Vascular Pathophysiology DepartmentCentro Nacional de Investigaciones CardiovascularesMadridSpain
- Genomic UnitCentro Nacional de Investigaciones CardiovascularesMadridSpain
- CIBER Cardiovascular, Instituto de Salud Carlos IIIMadridSpain
| | - Isidoro González Álvaro
- Rheumatology UnitHospital Universitario La Princesa, Instituto de Investigación Sanitaria‐Princesa IIS‐IPMadridSpain
| | - Santos Castañeda
- Rheumatology UnitHospital Universitario La Princesa, Instituto de Investigación Sanitaria‐Princesa IIS‐IPMadridSpain
- Cátedra UAM‐Roche, EPID‐Future, Department of MedicineUniversidad Autónoma de Madrid (UAM)MadridSpain
| | - Enrique Martin‐Gayo
- Immunology UnitHospital Universitario La Princesa, Medicine Department, Universidad Autónoma de Madrid, Instituto Investigación Sanitaria‐Princesa IIS‐IPMadridSpain
- CIBER Enfermedades Infecciosas (CIBERINFECC), Instituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
25
|
Shabrish S, Chandrakasan S, Madkaikar M. Editorial: NK cell defects: diagnosis and treatment. Front Immunol 2023; 14:1323793. [PMID: 38022628 PMCID: PMC10644773 DOI: 10.3389/fimmu.2023.1323793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Affiliation(s)
- Snehal Shabrish
- Translational Research Laboratory, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Shanmuganathan Chandrakasan
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, United States
| | - Manisha Madkaikar
- Department of Pediatric Immunology & Leukocyte Biology, Indian Council of Medical Research - National Institute of Immunohaematology, King Edward Memorial (KEM) Hospital, Mumbai, Maharashtra, India
| |
Collapse
|
26
|
Hours CM, Gil S, Gressens P. Molecular and Cellular Insights: A Focus on Glycans and the HNK1 Epitope in Autism Spectrum Disorder. Int J Mol Sci 2023; 24:15139. [PMID: 37894820 PMCID: PMC10606426 DOI: 10.3390/ijms242015139] [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: 08/16/2023] [Revised: 10/08/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is a synaptic disorder with a GABA/glutamate imbalance in the perineuronal nets and structural abnormalities such as increased dendritic spines and decreased long distance connections. Specific pregnancy disorders significantly increase the risk for an ASD phenotype such as preeclampsia, preterm birth, hypoxia phenomena, and spontaneous miscarriages. They are associated with defects in the glycosylation-immune placental processes implicated in neurogenesis. Some glycans epitopes expressed in the placenta, and specifically in the extra-villous trophoblast also have predominant functions in dendritic process and synapse function. Among these, the most important are CD57 or HNK1, CD22, CD24, CD33 and CD45. They modulate the innate immune cells at the maternal-fetal interface and they promote foeto-maternal tolerance. There are many glycan-based pathways of immunosuppression. N-glycosylation pathway dysregulation has been found to be associated with autoimmune-like phenotypes and maternal-autoantibody-related (MAR) autism have been found to be associated with central, systemic and peripheric autoimmune processes. Essential molecular pathways associated with the glycan-epitopes expression have been found to be specifically dysregulated in ASD, notably the Slit/Robo, Wnt, and mTOR/RAGE signaling pathways. These modifications have important effects on major transcriptional pathways with important genetic expression consequences. These modifications lead to defects in neuronal progenitors and in the nervous system's implementation specifically, with further molecular defects in the GABA/glutamate system. Glycosylation placental processes are crucial effectors for proper maternofetal immunity and endocrine/paracrine pathways formation. Glycans/ galectins expression regulate immunity and neurulation processes with a direct link with gene expression. These need to be clearly elucidated in ASD pathophysiology.
Collapse
Affiliation(s)
- Camille M Hours
- INSERM 1141, NeuroDiderot, Neuroprotection of the Developing Brain, Université Paris Cité, 75019 Paris, France
- Service de Psychiatrie de l'Enfant et de l'Adolescent, APHP, Hôpital Robert Debré, 75019 Paris, France
| | - Sophie Gil
- INSERM 1144, Therapeutics in Neuropsychopharmacology, Université Paris Cité, 75019 Paris, France
| | - Pierre Gressens
- INSERM 1141, NeuroDiderot, Neuroprotection of the Developing Brain, Université Paris Cité, 75019 Paris, France
- Neurologie Pédiatrique, APHP, Hôpital Robert Debré, 75019 Paris, France
| |
Collapse
|
27
|
Wang E, Zhou R, Li T, Hua Y, Zhou K, Li Y, Luo S, An Q. The Molecular Role of Immune Cells in Dilated Cardiomyopathy. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1246. [PMID: 37512058 PMCID: PMC10385992 DOI: 10.3390/medicina59071246] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023]
Abstract
Dilated cardiomyopathy (DCM) is a rare and severe condition characterized by chamber dilation and impaired contraction of the left ventricle. It constitutes a fundamental etiology for profound heart failure and abrupt cardiac demise, rendering it a prominent clinical indication for heart transplantation (HTx) among both adult and pediatric populations. DCM arises from various etiologies, including genetic variants, epigenetic disorders, infectious insults, autoimmune diseases, and cardiac conduction abnormalities. The maintenance of cardiac function involves two distinct types of immune cells: resident immune cells and recruited immune cells. Resident immune cells play a crucial role in establishing a harmonious microenvironment within the cardiac tissue. Nevertheless, in response to injury, cardiomyocytes initiate a cytokine cascade that attracts peripheral immune cells, thus perturbing this intricate equilibrium and actively participating in the initiation and pathological remodeling of dilated cardiomyopathy (DCM), particularly during the progression of myocardial fibrosis. Additionally, immune cells assume a pivotal role in orchestrating the inflammatory processes, which are intimately linked to the prognosis of DCM. Consequently, understanding the molecular role of various immune cells and their regulation mechanisms would provide an emerging era for managing DCM. In this review, we provide a summary of the most recent advancements in our understanding of the molecular mechanisms of immune cells in DCM. Additionally, we evaluate the effectiveness and limitations of immunotherapy approaches for the treatment of DCM, with the aim of optimizing future immunotherapeutic strategies for this condition.
Collapse
Affiliation(s)
- Enping Wang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Ruofan Zhou
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Tiange Li
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yimin Hua
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Kaiyu Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yifei Li
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shuhua Luo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Qi An
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
28
|
Khameneh HJ, Fonta N, Zenobi A, Niogret C, Ventura P, Guerra C, Kwee I, Rinaldi A, Pecoraro M, Geiger R, Cavalli A, Bertoni F, Vivier E, Trumpp A, Guarda G. Myc controls NK cell development, IL-15-driven expansion, and translational machinery. Life Sci Alliance 2023; 6:e202302069. [PMID: 37105715 PMCID: PMC10140547 DOI: 10.26508/lsa.202302069] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 04/19/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
MYC is a pleiotropic transcription factor involved in cancer, cell proliferation, and metabolism. Its regulation and function in NK cells, which are innate cytotoxic lymphocytes important to control viral infections and cancer, remain poorly defined. Here, we show that mice deficient for Myc in NK cells presented a severe reduction in these lymphocytes. Myc was required for NK cell development and expansion in response to the key cytokine IL-15, which induced Myc through transcriptional and posttranslational mechanisms. Mechanistically, Myc ablation in vivo largely impacted NK cells' ribosomagenesis, reducing their translation and expansion capacities. Similar results were obtained by inhibiting MYC in human NK cells. Impairing translation by pharmacological intervention phenocopied the consequences of deleting or blocking MYC in vitro. Notably, mice lacking Myc in NK cells exhibited defective anticancer immunity, which reflected their decreased numbers of mature NK cells exerting suboptimal cytotoxic functions. These results indicate that MYC is a central node in NK cells, connecting IL-15 to translational fitness, expansion, and anticancer immunity.
Collapse
Affiliation(s)
- Hanif J Khameneh
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Nicolas Fonta
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Alessandro Zenobi
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Charlène Niogret
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Pedro Ventura
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Concetta Guerra
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Ivo Kwee
- BigOmics Analytics SA, Lugano, Switzerland
| | - Andrea Rinaldi
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute of Oncology Research, Bellinzona, Switzerland
| | - Matteo Pecoraro
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Roger Geiger
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute of Oncology Research, Bellinzona, Switzerland
| | - Andrea Cavalli
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Francesco Bertoni
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute of Oncology Research, Bellinzona, Switzerland
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Eric Vivier
- Aix-Marseille Université, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- Innate Pharma Research Laboratories, Marseille, France
- APHM, Hôpital de la Timone, Marseille-Immunopôle, Marseille, France
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, DKFZ, Heidelberg, Germany
- HI-STEM: The Heidelberg Institute for Stem Cell Technology and Experimental Medicine gGmbH, Heidelberg, Germany
| | - Greta Guarda
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| |
Collapse
|
29
|
Li YM, Li YX, Hu XZ, Li DY, An L, Yuan ZY, Liu ZL, Du KM, Zheng ZZ. Exploration of KIR genes and hematological-related diseases in Chinese Han population. Sci Rep 2023; 13:9773. [PMID: 37328612 PMCID: PMC10276034 DOI: 10.1038/s41598-023-36882-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 06/12/2023] [Indexed: 06/18/2023] Open
Abstract
The function of natural killer (NK) cells has previously been implicated in hematopoietic-related diseases. Killer immunoglobulin-like receptors (KIR) play an important role in NK cells after hematopoietic stem cell transplantation. To explore the immunogenetic predisposition of hematological-related diseases, herein, a multi-center retrospective study in China was conducted, analyzing and comparing 2519 patients with hematopathy (mainly, acute lymphoblastic leukemia, acute myeloid leukemia, aplastic anemia, and myelodysplastic syndrome) to 18,108 individuals without known pathology. Genotyping was performed by polymerase chain reaction with specific sequence primers (PCR-SSP). As a result, we discovered four genes including KIR2DL5 (OR: 0.74, 95% CI 0.59-0.93; Pc = 0.0405), 2DS1 (OR: 0.74, 95% CI 0.59-0.93; Pc = 0.0405), 2DS3 (OR: 0.58, 95% CI 0.41-0.81; Pc = 0.0180), and 3DS1 (OR: 0.74, 95% CI 0.58-0.94; Pc = 0.0405) to be protective factors that significantly reduce the risk of aplastic anemia. Our findings offer new approaches to immunotherapy for hematological-related diseases. As these therapies mature, they are promising to be used alone or in combination with current treatments to help to make blood disorders a manageable disease.
Collapse
Affiliation(s)
- Ye-Mo Li
- Shanghai Tissuebank Biotechnology Co, Ltd, Shanghai, China
| | - Yu-Xia Li
- Shanghai Tissuebank Biotechnology Co, Ltd, Shanghai, China
| | - Xiao-Zhuang Hu
- Shanghai Tissuebank Biotechnology Co, Ltd, Shanghai, China
| | - Dai-Yang Li
- Shanghai Tissuebank Biotechnology Co, Ltd, Shanghai, China
| | - Lin An
- Shanghai Tissuebank Biotechnology Co, Ltd, Shanghai, China
| | - Zhi-Yang Yuan
- Shanghai Tissuebank Biotechnology Co, Ltd, Shanghai, China
| | | | - Ke-Ming Du
- Shanghai Tissuebank Biotechnology Co, Ltd, Shanghai, China
| | | |
Collapse
|
30
|
Hojjatipour T, Maali A, Azad M. Natural killer cell epigenetic reprogramming in tumors and potential for cancer immunotherapy. Epigenomics 2023; 15:249-266. [PMID: 37125432 DOI: 10.2217/epi-2022-0454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Natural killer (NK) cells are critical members of the innate lymphoid cell population and have a pivotal role in cancer eradication. NK cell maturation, development and function are tightly regulated by epigenetic modifications, which can also be recruited for cancer propagation and immune escape. NK cells have the potential to be activated against tumors through several epigenetic regulators. Given that epigenetic changes are inducible and reversible, focusing on aberrant epigenetic regulations recruited by tumor cells provides a tremendous opportunity for cancer treatment. This review presents a comprehensive picture of NK cell normal epigenetic regulation and cancer-driven epigenetic modifications. From our perspective, a better understanding of epigenetic regulators that can edit and revise NK cells' activity is a promising avenue for NK cell-based therapy in cancer management.
Collapse
Affiliation(s)
- Tahereh Hojjatipour
- Department of Hematology & Blood Transfusion, Students Research Center, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Department of Medical Biotechnology, School of Paramedicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mehdi Azad
- Department of Medical Laboratory Sciences, School of Paramedicine, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
31
|
Wu WK, Stier MT, Stokes JW, Ukita R, Patel YJ, Cortelli M, Landstreet SR, Talackine JR, Cardwell NL, Simonds EM, Mentz M, Lowe C, Benson C, Demarest CT, Alexopoulos SP, Shaver CM, Bacchetta M. Immune characterization of a xenogeneic human lung cross-circulation support system. SCIENCE ADVANCES 2023; 9:eade7647. [PMID: 37000867 PMCID: PMC10065447 DOI: 10.1126/sciadv.ade7647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Improved approaches to expanding the pool of donor lungs suitable for transplantation are critically needed for the growing population with end-stage lung disease. Cross-circulation (XC) of whole blood between swine and explanted human lungs has previously been reported to enable the extracorporeal recovery of donor lungs that declined for transplantation due to acute, reversible injuries. However, immunologic interactions of this xenogeneic platform have not been characterized, thus limiting potential translational applications. Using flow cytometry and immunohistochemistry, we demonstrate that porcine immune cell and immunoglobulin infiltration occurs in this xenogeneic XC system, in the context of calcineurin-based immunosuppression and complement depletion. Despite this, xenogeneic XC supported the viability, tissue integrity, and physiologic improvement of human donor lungs over 24 hours of xeno-support. These findings provide targets for future immunomodulatory strategies to minimize immunologic interactions on this organ support biotechnology.
Collapse
Affiliation(s)
- Wei K. Wu
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Surgery, Division of Hepatobiliary Surgery and Liver Transplantation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthew T. Stier
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John W. Stokes
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rei Ukita
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yatrik J. Patel
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael Cortelli
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stuart R. Landstreet
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jennifer R. Talackine
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nancy L. Cardwell
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Elizabeth M. Simonds
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Meredith Mentz
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cindy Lowe
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Clayne Benson
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Caitlin T. Demarest
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sophoclis P. Alexopoulos
- Department of Surgery, Division of Hepatobiliary Surgery and Liver Transplantation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ciara M. Shaver
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Corresponding author. (M.B.); (C.M.S.)
| | - Matthew Bacchetta
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Corresponding author. (M.B.); (C.M.S.)
| |
Collapse
|
32
|
Lima CAD, Jammal MP, Etchebehere RM, Murta EFC, Nomelini RS. Immunostaining of stromal CD56 cells in ovarian malignancies. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2023; 69:e20220992. [PMID: 37194901 DOI: 10.1590/1806-9282.20220992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/23/2023] [Indexed: 05/18/2023]
Abstract
OBJECTIVES The aim of this study was to evaluate CD56 immunostaining in the stroma of benign and malignant ovarian epithelial neoplasms and associate the CD56 immunostaining with prognostic factors and survival in ovarian cancer. METHODS Patients with ovarian epithelial neoplasia (n=77) were studied with a prospective cohort. The CD56 immunostaining was evaluated in the peritumoral stroma. Two groups were evaluated: benign ovarian neoplasms (n=40) and malignant ovarian neoplasms (n=37). Data were recorded for histological type and grade, International Federation of Gynecology and Obstetrics staging, molecular subtype, and lymph node metastases. Fisher's exact test and Kaplan-Meier survival curves were used, with a significance level of ≤0.05. RESULTS We found greater CD56 stromal immunostaining in malignant neoplasms when compared to the group of benign neoplasms (p=0.00001). There was no significant difference in relation to the prognostic factors and survival. CONCLUSION Malignant ovarian neoplasms showed higher stromal CD56 immunostaining. As the prognostic value of natural killer in ovarian cancer is controversial, knowing the specific function of each cell present both in the tumor tissue and systemically may help guide successful immunotherapies in the near future.
Collapse
Affiliation(s)
- Cid Almeida de Lima
- Universidade Federal do Triângulo Mineiro, Department of Gynecology and Obstetrics - Uberaba (MG), Brazil
| | - Millena Prata Jammal
- Universidade Federal do Triângulo Mineiro, Department of Gynecology and Obstetrics - Uberaba (MG), Brazil
| | | | | | - Rosekeila Simões Nomelini
- Universidade Federal do Triângulo Mineiro, Department of Gynecology and Obstetrics - Uberaba (MG), Brazil
| |
Collapse
|
33
|
Sieniawska J, Krzewska A, Skowronek A, Wrobel W, Tomczyk Z, Pach E, Rosolowska I, Wilczynska B, Beń-Skowronek I. Lower percentages of natural killer cells in children with type 1 diabetes and their siblings. Pediatr Endocrinol Diabetes Metab 2023; 29:214-224. [PMID: 38282490 PMCID: PMC10826694 DOI: 10.5114/pedm.2023.132029] [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: 07/10/2023] [Accepted: 09/04/2023] [Indexed: 01/30/2024]
Abstract
INTRODUCTION One of the most common children's endocrine and autoimmune diseases in the world is type 1 diabetes mellitus (T1DM). The incidence of type 1 diabetes is constantly increasing, and according to current estimates, the number of children with T1DM in the world has exceeded 542,000. There are 3 main components emphasized in the pathogenesis: genetic and environmental factors, and the patient's immune system. Many publications have confirmed the role of natural killer cells (NK) in the pathogenesis of type 1 diabetes and other autoimmune diseases. AIM The aim of the study was to evaluate the population of NK cells and pancreatic β cell autoantibodies in a group of children with T1DM and their healthy siblings in comparison with children from families with no history of autoimmune diseases. MATERIAL AND METHODS The research included 76 children with T1DM, 101 children from the sibling group, and 30 children from the control group. Peripheral blood was analysed on a FACSCalibur flow cytometer (Becton Dickinson) to evaluate the NK cell population. The results were presented as the percentage of NK cells among lymphocytes. Statistical analysis was performed using STATIS-TICA 10 PL software. RESULTS The mean percentage of NK cells in children with T1D (10.59 ±5.37) and in the sibling group (11.93 ±5.62) was statistically reduced in comparison to the control group (14.89 ±7.78) in sequence (Student's t -test: t = -3.24; df = 103; p = 0.002) (Stu-dent's t -test: t = -2.30; df = 128; p = 0.02). There was no statistically significant difference in the percentage of NK cells be-tween the group of children with T1DM and their siblings (Student's t -test: t = -1.59; df = 173; p = 0.11). In the group of sib-lings, the younger the child, the lower the reported percentage of NK cells. This relationship was statistically significant (test for the Pearson correlation coefficient t = 3.41; p = 0.0009; r = 0.33). In the group of children with type 1 diabetes, a similar relationship was not found. The concentration of anti-IA2 and anti-Znt8 antibodies was statistically significantly higher in the sibling group compared to the control group (anti-IA2 p = 0.0000001; anti-ZnT8 p = 0.00001), and the concentration of anti-GAD antibodies was comparable in both groups. In the group of children with type 1 diabetes, a positive correlation was demonstrated between the reduced percentage of NK cells and the coexistence of anti-GAD and anti-ZnT8 antibodies (Mann-Whitney U test Z = -2.02; p = 0.04). There was no similar relationship in the group of siblings. CONCLUSIONS The reduced percentage of NK cells in children with T1DM and in their siblings compared to the control group suggests the role of NK cells in the pathogenesis of T1DM. Genetic predisposition and dysfunction of NK cells probably underlie the pathogenesis of T1DM.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Iwona Beń-Skowronek
- Department of Paediatric Endocrinology and Diabetology with Laboratory of Endocrinology and Metabolism, Medical University of Lublin, Poland
| |
Collapse
|
34
|
Agarbati S, Benfaremo D, Viola N, Paolini C, Svegliati Baroni S, Funaro A, Moroncini G, Malavasi F, Gabrielli A. Increased expression of the ectoenzyme CD38 in peripheral blood plasmablasts and plasma cells of patients with systemic sclerosis. Front Immunol 2022; 13:1072462. [PMID: 36618427 PMCID: PMC9811259 DOI: 10.3389/fimmu.2022.1072462] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Objective CD38 is a type II glycoprotein highly expressed on plasmablasts and on short- and long-lived plasma cells, but weakly expressed by lymphoid, myeloid, and non-hematopoietic cells. CD38 is a target for therapies aimed at depleting antibody-producing plasma cells. Systemic sclerosis (SSc) is an immune-mediated disease with a well-documented pathogenic role of B cells. We therefore analyzed CD38 expression in different subsets of peripheral blood mononuclear cells (PBMCs) from a cohort of SSc patients. Methods Cell surface expression of CD38 was evaluated on PBMCs from SSc patients using eight-color flow cytometry analysis performed with a FacsCanto II (BD). Healthy individuals were used as controls (HC). Results Forty-six SSc patients (mean age 56, range 23-79 years; 38 females and 8 males), and thirty-two age- and sex-matched HC were studied. Twenty-eight patients had the limited cutaneous form and eighteen the diffuse cutaneous form of SSc. The mean disease duration was 7 years. Fourteen patients were on immunosuppressive therapy (14 MMF, 5 RTX). The total percentages of T, B and NK cells were not different between SSc and HC. Compared to HC, SSc patients had higher levels of CD3+CD38+ T cells (p<0.05), higher percentage (p<0.001) of CD3+CD4+CD25+FOXP3+ regulatory T cells, lower percentage (p<0.05) of CD3+CD56+ NK T cells. Moreover, SSc patients had higher levels of CD24highCD19+CD38high regulatory B cells than HC (p<0.01), while the amount of CD24+CD19+CD38+CD27+ memory B cells was lower (p<0.001). Finally, the percentages of circulating CD38highCD27+ plasmablasts and CD138+CD38high plasma cells were both higher in the SSc group than in HC (p<0.001). We did not observe any correlations between these immunophenotypes and disease subsets or duration, and ongoing immunosuppressive treatment. Conclusions The increased expression of CD38 in peripheral blood plasmablasts and plasma cells of SSc patients may suggest this ectoenzyme as a candidate therapeutic target, under the hypothesis that depletion of these cells may beneficially downregulate the chronic immune response in SSc patients. Validation of this data in multicenter cohorts shall be obtained prior to clinical trials with existing anti-CD38 drugs.
Collapse
Affiliation(s)
- S. Agarbati
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - D. Benfaremo
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy,Clinica Medica, Department of Internal Medicine, Azienda Ospedaliero Universitaria delle Marche, Ancona, Italy
| | - N. Viola
- Immunologia Clinica, Department of Internal Medicine, Azienda Ospedaliero Universitaria delle Marche, Ancona, Italy
| | - C. Paolini
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - S. Svegliati Baroni
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - A. Funaro
- Department of Medical Sciences, University of Turin, Torino, Italy
| | - G. Moroncini
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy,Clinica Medica, Department of Internal Medicine, Azienda Ospedaliero Universitaria delle Marche, Ancona, Italy,*Correspondence: G. Moroncini,
| | - F. Malavasi
- Department of Medical Sciences, University of Turin, Torino, Italy,Fondazione Ricerca Molinette, Torino, Italy
| | - A. Gabrielli
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| |
Collapse
|
35
|
Fang D, Chen B, Lescoat A, Khanna D, Mu R. Immune cell dysregulation as a mediator of fibrosis in systemic sclerosis. Nat Rev Rheumatol 2022; 18:683-693. [DOI: 10.1038/s41584-022-00864-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2022] [Indexed: 11/11/2022]
|
36
|
An Q, Zhao J, Zhu X, Yang B, Wu Z, Su Y, Zhang L, Xu K, Ma D. Exploiting the role of T cells in the pathogenesis of Sjögren's syndrome for therapeutic treatment. Front Immunol 2022; 13:995895. [PMID: 36389806 PMCID: PMC9650646 DOI: 10.3389/fimmu.2022.995895] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 10/17/2022] [Indexed: 08/19/2023] Open
Abstract
Sjögrens syndrome (SS) is caused by autoantibodies that attack proprioceptive salivary and lacrimal gland tissues. Damage to the glands leads to dry mouth and eyes and affects multiple systems and organs. In severe cases, SS is life-threatening because it can lead to interstitial lung disease, renal insufficiency, and lymphoma. Histological examination of the labial minor salivary glands of patients with SS reveals focal lymphocyte aggregation of T and B cells. More studies have been conducted on the role of B cells in the pathogenesis of SS, whereas the role of T cells has only recently attracted the attention of researchers. This review focusses on the role of various populations of T cells in the pathogenesis of SS and the progress made in research to therapeutically targeting T cells for the treatment of patients with SS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Dan Ma
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
37
|
Studying the Interactions of U24 from HHV-6 in Order to Further Elucidate Its Potential Role in MS. Viruses 2022; 14:v14112384. [PMID: 36366483 PMCID: PMC9696605 DOI: 10.3390/v14112384] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 01/31/2023] Open
Abstract
A number of studies have suggested that human herpesvirus 6A (HHV-6A) may play a role in multiple sclerosis (MS). Three possible hypotheses have been investigated: (1) U24 from HHV-6A (U24-6A) mimics myelin basic protein (MBP) through analogous phosphorylation and interaction with Fyn-SH3; (2) U24-6A affects endocytic recycling by binding human neural precursor cell (NPC) expressed developmentally down-regulated protein 4-like WW3* domain (hNedd4L-WW3*); and (3) MS patients who express Killer Cell Immunoglobulin Like Receptor 2DL2 (KIR2DL2) on natural killer (NK) cells are more susceptible to HHV-6 infection. In this contribution, we examined the validity of these propositions by investigating the interactions of U24 from HHV-6B (U24-6B), a variant less commonly linked to MS, with Fyn-SH3 and hNedd4L-WW3* using heteronuclear single quantum coherence (HSQC) nuclear magnetic resonance (NMR) titrations and isothermal titration calorimetry (ITC). In addition, the importance of phosphorylation and the specific role of U24 in NK cell activation in MS patients were examined. Overall, the findings allowed us to shed light into the models linking HHV-6 to MS and the involvement of U24.
Collapse
|
38
|
Sato M, Arakaki R, Tawara H, Nagao R, Tanaka H, Tamura K, Kawahito Y, Otsuka K, Ushio A, Tsunematsu T, Ishimaru N. Disturbed natural killer cell homeostasis in the salivary gland enhances autoimmune pathology via IFN-γ in a mouse model of primary Sjögren's syndrome. Front Med (Lausanne) 2022; 9:1036787. [PMID: 36388880 PMCID: PMC9643684 DOI: 10.3389/fmed.2022.1036787] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/12/2022] [Indexed: 07/22/2023] Open
Abstract
OBJECTIVE Innate lymphoid cells (ILCs), including natural killer (NK) cells, ILC1, ILC2, lymphoid tissue-inducer (LTi) cells, and ILC3 cell, play a key role in various immune responses. Primary Sjögren's syndrome (pSS) is an autoimmune disease characterized by chronic inflammation of exocrine glands, such as the lacrimal and salivary glands (SGs). The role of NK cells among ILCs in the pathogenesis of pSS is still unclear. In this study, the characteristics and subsets of NK cells in the salivary gland (SG) tissue were analyzed using a murine model of pSS. METHODS Multiple phenotypes and cytotoxic signature of the SG NK cells in control and pSS model mice were evaluated by flow cytometric analysis. Intracellular expression of interferon-γ (IFN-γ) among T cells and NK cells from the SG tissues was compared by in vitro experiments. In addition, pathological analysis was performed using anti-asialo-GM1 (ASGM1) antibody (Ab)-injected pSS model mice. RESULTS The number of conventional NK (cNK) cells in the SG of pSS model mice significantly increased compared with that in control mice at 6 weeks of age. The production level of IFN-γ was significantly higher in SG NK cells than in SG T cells. The depletion of NK cells by ASGM1 Ab altered the ratio of tissue resident NK (rNK) cells to cNK cells, which inhibited the injury to SG cells with the recovery of saliva secretion in pSS model mice. CONCLUSION The results indicate that SG cNK cells may enhance the autoreactive response in the target organ by upregulating of IFN-γ, whereas SG rNK cells protect target cells against T cell cytotoxicity. Therefore, the activation process and multiple functions of NK cells in the target organ could be helpful to develop potential markers for determining autoimmune disease activity and target molecules for incurable immune disorders.
Collapse
|
39
|
Wang X, Xiong H, Ning Z. Implications of NKG2A in immunity and immune-mediated diseases. Front Immunol 2022; 13:960852. [PMID: 36032104 PMCID: PMC9399941 DOI: 10.3389/fimmu.2022.960852] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 07/21/2022] [Indexed: 11/22/2022] Open
Abstract
In recent studies, NKG2A is revealed to be a key immune checkpoint for both natural killer (NK) cells and CD8+ T cells. It form heterodimer receptors with CD94, and targets the peptide-presenting human leukocyte antigen-E (HLA-E) molecules. Upon crosslinking, NKG2A/CD94 delivers inhibitory signals for NK cells and CD8+ T cells, while blocking NKG2A can effectively unleash functions of these cytotoxic lymphocytes. The interaction between NKG2A and HLA-E contributes to tumor immune escape, and NKG2A-mediated mechanisms are currently being exploited to develop potential antitumor therapeutic strategies. In addition, growing evidence shows that NKG2A also plays important roles in other immune-related diseases including viral infections, autoimmune diseases, inflammatory diseases, parasite infections and transplant rejection. Therefore, the current work focuses on describing the effect of NKG2A on immune regulation and exploring its potential role in immune-mediated disorders.
Collapse
Affiliation(s)
- Xiaotong Wang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, China
- *Correspondence: Zhaochen Ning, ; Huabao Xiong,
| | - Zhaochen Ning
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, China
- *Correspondence: Zhaochen Ning, ; Huabao Xiong,
| |
Collapse
|
40
|
Beliën J, Goris A, Matthys P. Natural Killer Cells in Multiple Sclerosis: Entering the Stage. Front Immunol 2022; 13:869447. [PMID: 35464427 PMCID: PMC9019710 DOI: 10.3389/fimmu.2022.869447] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/14/2022] [Indexed: 11/14/2022] Open
Abstract
Studies investigating the immunopathology of multiple sclerosis (MS) have largely focused on adaptive T and B lymphocytes. However, in recent years there has been an increased interest in the contribution of innate immune cells, amongst which the natural killer (NK) cells. Apart from their canonical role of controlling viral infections, cell stress and malignancies, NK cells are increasingly being recognized for their modulating effect on the adaptive immune system, both in health and autoimmune disease. From different lines of research there is now evidence that NK cells contribute to MS immunopathology. In this review, we provide an overview of studies that have investigated the role of NK cells in the pathogenesis of MS by use of the experimental autoimmune encephalomyelitis (EAE) animal model, MS genetics or through ex vivo and in vitro work into the immunology of MS patients. With the advent of modern hypothesis-free technologies such as single-cell transcriptomics, we are exposing an unexpected NK cell heterogeneity, increasingly blurring the boundaries between adaptive and innate immunity. We conclude that unravelling this heterogeneity, as well as the mechanistic link between innate and adaptive immune cell functions will lay the foundation for the use of NK cells as prognostic tools and therapeutic targets in MS and a myriad of other currently uncurable autoimmune disorders.
Collapse
Affiliation(s)
- Jarne Beliën
- Department of Neurosciences, Laboratory for Neuroimmunology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - An Goris
- Department of Neurosciences, Laboratory for Neuroimmunology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Patrick Matthys
- Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
41
|
Muscari I, Fierabracci A, Adorisio S, Moretti M, Cannarile L, Thi Minh Hong V, Ayroldi E, Delfino DV. Glucocorticoids and natural killer cells: A suppressive relationship. Biochem Pharmacol 2022; 198:114930. [PMID: 35149054 DOI: 10.1016/j.bcp.2022.114930] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 11/02/2022]
Abstract
Glucocorticoids exert their pharmacological actions by mimicking and amplifying the function of the endogenous glucocorticoid system's canonical physiological stress response. They affect the immune system at the levels of inflammation and adaptive and innate immunity. These effects are the basis for therapeutic use of glucocorticoids. Innate immunity is the body's first line of defense against disease conditions. It is relatively nonspecific and, among its mediators, natural killer (NK) cells link innate and acquired immunity. NK cell numbers are altered in patients with auto immune diseases, and research suggests that interactions between glucocorticoids and natural killer cells are critical for successful glucocorticoid therapy. The aim of this review is to summarize these interactions while highlighting the latest and most important developments in this field. Production and release in the blood of endogenous glucocorticoids are strictly regulated by the hypothalamus-pituitary adrenal axis. A self-regulatory mechanism prevents excessive plasma levels of these hormones. However, exogenous stimuli such as stress, inflammation, infections, cancer, and autoimmune disease can trigger the hypothalamus-pituitary-adrenal axis response and lead to excessive systemic release of glucocorticoids. Thus, stress stimuli, such as sleep deprivation, intense exercise, depression, viral infections, and cancer, can result in release of glucocorticoids and associated immunosuppressant effects. Among these effects are decreases in the numbers and activities of NK cells in inflammatory and autoimmune diseases (e.g., giant cell arteritis, polymyalgia rheumatica, and familial hypogammaglobulinemia).
Collapse
Affiliation(s)
- Isabella Muscari
- Section of Onco-hematology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Alessandra Fierabracci
- Infectivology and Clinical Trials Research Department, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Sabrina Adorisio
- Foligno Nursing School, Department of Medicine and Surgery, University of Perugia, Foligno, PG, Italy
| | - Marina Moretti
- Section of Onco-hematology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lorenza Cannarile
- Section of Pharmacology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Emira Ayroldi
- Section of Pharmacology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Domenico V Delfino
- Foligno Nursing School, Department of Medicine and Surgery, University of Perugia, Foligno, PG, Italy; Section of Pharmacology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| |
Collapse
|
42
|
Yang X, Li M, Qin X, Tan S, Du L, Ma C, Li M. Photophosphatidylserine Guides Natural Killer Cell Photoimmunotherapy via Tim-3. J Am Chem Soc 2022; 144:3863-3874. [PMID: 35226805 DOI: 10.1021/jacs.1c11498] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Natural killer (NK) cells, in addition to their cytotoxicity function, harbor prominent cytokine production capabilities and contribute to regulating autoimmune responses. T-cell immunoglobulin and mucin domain containing protein-3 (Tim-3) is one of the inhibitory receptors on NK cells and a promising immune checkpoint target. We recently found that phosphatidylserine (PS) binding to Tim-3 can suppress NK cell activation. Therefore, based on the therapeutic potential of Tim-3 in NK-cell-mediated diseases, we developed a photoswitchable ligand of Tim-3, termed photophosphatidylserine (phoPS), that mimics the effects of PS. Upon 365 or 455 nm light irradiation, the isomer of phoPS cyclically conversed the cis/trans configuration, resulting in an active/inactive Tim-3 ligand, thus modulating the function of NK cells in vitro and in vivo. We also demonstrated that reversible phoPS enabled optical control of acute hepatitis. Together, phoPS may be an appealing tool for autoimmune diseases and cytokine storms in the future.
Collapse
Affiliation(s)
- Xingye Yang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Mengzhen Li
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiaojun Qin
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Siyu Tan
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Lupei Du
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Minyong Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Helmholtz International Lab, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China
| |
Collapse
|
43
|
Dubis J, Niepiekło-Miniewska W, Jędruchniewicz N, Sobczyński M, Witkiewicz W, Zapotoczny N, Kuśnierczyk P. Associations of Genes for Killer Cell Immunoglobulin-like Receptors and Their Human Leukocyte Antigen-A/B/C Ligands with Abdominal Aortic Aneurysm. Cells 2021; 10:cells10123357. [PMID: 34943866 PMCID: PMC8699266 DOI: 10.3390/cells10123357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/20/2021] [Accepted: 11/25/2021] [Indexed: 11/28/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is an immune-mediated disease with a genetic component. The multifactorial pathophysiology is not clear and there is still no pharmacotherapy to slow the growth of aneurysms. The signal integration of cell-surface KIRs (killer cell immunoglobulin-like receptors) with HLA (ligands, human leukocyte class I antigen molecules) modulates the activity of natural killer immune cells. The genetic diversity of the KIR/HLA system is associated with the risk of immune disorders. This study was a multivariate analysis of the association between genetic variants of KIRs, HLA ligands, clinical data and AAA formation. Genotyping was performed by single polymerase chain reaction with sequence-specific primers using commercial assays. Patients with HLA-A-Bw4 have a larger aneurysm by an average of 4 mm (p = 0.008). We observed a relationship between aneurysm diameter and BMI in patients with AAA and co-existing CAD; its shape was determined by the presence of HLA-A-Bw4. There was also a nearly 10% difference in KIR3DL1 allele frequency between the study and control groups. High expression of the cell surface receptor KIR3DL1 may protect, to some extent, against AAA. The presence of HLA-A-Bw4 may affect the rate of aneurysm growth and represents a potential regional pathogenetic risk of autoimmune injury to the aneurysmal aorta.
Collapse
Affiliation(s)
- Joanna Dubis
- Research and Development Centre, Regional Specialist Hospital, 51-124 Wroclaw, Poland;
- Correspondence: (J.D.); (P.K.)
| | - Wanda Niepiekło-Miniewska
- Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland;
| | | | - Maciej Sobczyński
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland;
| | - Wojciech Witkiewicz
- Department of Vascular Surgery, Regional Specialist Hospital in Wroclaw, 51-124 Wrocław, Poland; (W.W.); (N.Z.)
| | - Norbert Zapotoczny
- Department of Vascular Surgery, Regional Specialist Hospital in Wroclaw, 51-124 Wrocław, Poland; (W.W.); (N.Z.)
| | - Piotr Kuśnierczyk
- Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland;
- Correspondence: (J.D.); (P.K.)
| |
Collapse
|
44
|
Reevaluation of NOD/SCID Mice as NK Cell-Deficient Models. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8851986. [PMID: 34805408 PMCID: PMC8598338 DOI: 10.1155/2021/8851986] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 07/21/2021] [Accepted: 10/21/2021] [Indexed: 12/02/2022]
Abstract
Objective Natural killer (NK) cell-deficient mice are useful models in biomedical research. NOD/SCID mice have been used as a model of this type in research. However, the actual status of NK cells in NOD/SCID mice and CB17/SCID mice in comparison with that in BALB/c mice has not been sufficiently evaluated. Methods Splenocytes from naïve or poly(I:C)-treated mice were isolated for phenotyping and analysis of cytotoxicity-related molecules and inhibitory receptors; for cytotoxicity assay, purified NK cells were also used. Results The proportion of splenic NK cells did not differ significantly between NOD/SCID and CB17/SCID mice. The perforin levels in NK cells were similar between the poly(I:C)-treated CB17/SCID and NOD/SCID mice, while the granzyme B and NKG2A/C/E levels in NK cells from NOD/SCID mice were significantly lower than those from CB17/SCID mice. Moreover, the NKG2D and Ly49A levels in NK cells from NOD/SCID mice were higher than those from CB17/SCID. The splenocytes from CB17/SCID mice showed higher cytotoxicity than those from NOD/SCID mice, while the cytotoxicity of purified NK cells basically did not differ between the two strains. After in vitro stimulation with cytokines, the splenocytes from CB17/SCID mice showed higher IFN-γ production than those from NOD/SCID mice; however, NK cells did not. Conclusion There was no significant difference in the proportion of splenic NK cells between CB17/SCID and NOD/SCID mice, and the function of NK cells was only partially compromised in NOD/SCID mice. Caution should be taken when considering the use of NOD/SCID mice as an NK-deficient model.
Collapse
|
45
|
The Role of miR-23b in Cancer and Autoimmune Disease. JOURNAL OF ONCOLOGY 2021; 2021:6473038. [PMID: 34777498 PMCID: PMC8580694 DOI: 10.1155/2021/6473038] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022]
Abstract
Short-stranded miRNAs are single-stranded RNA molecules involved in the regulation of gene expression. miRNAs are involved in a variety of cellular physiological processes, including cell proliferation, differentiation, and apoptosis. miR-23b have been identified to act both as oncogenes and as tumor suppressors. In addition, miR-23b is related to inflammation resistance to various autoimmune diseases and restrained inflammatory cell migration. The characterization of the specific alterations in the patterns of miR-23b expression in cancer and autoimmune disease has great potential for identifying biomarkers for early disease diagnosis, as well as for potential therapeutic intervention in various diseases. In this review, we summarize the ever-expanding role of miR-23b and its target genes in different models and offer insight into how this multifunctional miRNA modulates tumor cell proliferation and apoptosis or inflammatory cell activation, differentiation, and migration.
Collapse
|
46
|
Erken E, Senel ME, Dinkci S, Goruroglu Ozturk O, Altunoren O, Gungor O, Erken E. Killer cell immunoglobulin-like receptor (KIR) gene distribution and allograft kidney function. Int J Clin Pract 2021; 75:e14790. [PMID: 34480825 DOI: 10.1111/ijcp.14790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/27/2021] [Accepted: 09/02/2021] [Indexed: 01/06/2023] Open
Affiliation(s)
- Ertugrul Erken
- Department of Nephrology, Sutcu Imam University, Kahramanmaras, Turkey
| | - Mahmut Egemen Senel
- Department of Internal Medicine, Sutcu Imam University, Kahramanmaras, Turkey
| | - Suzan Dinkci
- Department of Rheumatology/Immunology, Cukurova University, Adana, Turkey
| | - Ozlem Goruroglu Ozturk
- Faculty of Medicine, Balcali Hospital, Central Laboratory, Cukurova University, Adana, Turkey
| | - Orcun Altunoren
- Department of Nephrology, Sutcu Imam University, Kahramanmaras, Turkey
| | - Ozkan Gungor
- Department of Nephrology, Sutcu Imam University, Kahramanmaras, Turkey
| | - Eren Erken
- Department of Rheumatology/Immunology, Cukurova University, Adana, Turkey
| |
Collapse
|
47
|
Contribution of Antigen-Processing Machinery Genetic Polymorphisms to Atopic Dermatitis. Life (Basel) 2021; 11:life11040333. [PMID: 33920176 PMCID: PMC8070454 DOI: 10.3390/life11040333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 01/12/2023] Open
Abstract
Atopic dermatitis (AD) is a chronic and recurrent inflammatory dermatosis. We recently described an association of the C allele of the single nucleotide polymorphism (SNP) rs26618 in the ERAP1 gene and a synergism of ERAP1 and ERAP2 effects on AD risk. Here, we examined whether polymorphisms of other antigen-presenting machinery genes encoding immunoproteasome components LMP2 and LMP7 and peptide transporter components TAP1 and TAP2 may also affect susceptibility to AD or its outcome. We found that the LMP7 rs2071543*T allele decreased disease risk by about 1.5-fold (odds ratio 0.66, 95% confidence interval 0.44–0.99). On the other hand, the LMP2 rs1351383*C allele reduced the mean age at diagnosis from 23 to 15 years (p < 0.001). Similarly, the TAP1 rs1135216*C allele decreased the mean age at diagnosis from almost 20 to 14 years (p = 0.033). The results are discussed in light of other reports on the role of these polymorphisms in human disease.
Collapse
|