1
|
Shi W, Hu J, Wang H, Zhong H, Zhang W, Wang J, Shao H, Shen H, Bo H, Tao C, Wu F. miR-143-3p Promotes T SCM Differentiation and Inhibits Progressive T Cell Differentiation via Inhibiting ABL2 and PAG1. Genes (Basel) 2025; 16:466. [PMID: 40282426 PMCID: PMC12027245 DOI: 10.3390/genes16040466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Adoptive cell therapy (ACT), including CAR-T and TCR-T therapies, shows promise for cancer treatment, depending on infused T cell expansion, persistence and activity. We previously characterized four T-cell subsets (TN, TSCM, TCM and TEM) and their miRNA profiles. OBJECTIVES This study investigates miR-143-3p's role in T cell differentiation. METHODS Using qPCR, we analyzed miR-143-3p expression. Target genes were validated by dual-luciferase assays. Functional assays assessed differentiation markers, proliferation, apoptosis and cytokine secretion. RESULTS miR-143-3p was upregulated in early-differentiated TSCM but downregulated during progression. We confirmed ABL2 and PAG1 as direct targets suppressed by miR-143-3p. Overexpression increased early markers (LEF1, CCR7 and CD62L) while decreasing late markers (EOMES, KLRG1 and CD45RO). It also enhanced proliferation, reduced apoptosis and suppressed cytokine secretion. CONCLUSIONS miR-143-3p promotes TSCM differentiation and inhibits progressive differentiation by targeting ABL2/PAG1, suggesting new ACT optimization strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Fenglin Wu
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China; (W.S.); (J.H.); (H.W.); (H.Z.); (W.Z.); (J.W.); (H.S.); (H.S.); (H.B.); (C.T.)
| |
Collapse
|
2
|
Liu S, Pu P, Xiang Q, Pu X. Exploration of common molecular mechanisms of psoriatic arthritis and aging based on integrated bioinformatics and single-cell RNA-seq analysis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167730. [PMID: 39965531 DOI: 10.1016/j.bbadis.2025.167730] [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: 09/09/2024] [Revised: 01/13/2025] [Accepted: 02/13/2025] [Indexed: 02/20/2025]
Abstract
OBJECTIVE This study investigated the key genes shared between Psoriatic arthritis (PSA) and aging. METHODS By integrating and analyzing single-cell RNA sequencing data from the synovial fluid of PsA patients, peripheral blood of senescent patients, and the normal population, the subpopulation of cells that were jointly upregulated in both was obtained as the core cellular subpopulation. We analyzed the proposed chronology of this core cellular subpopulations and the function of cellular communication, screened the differentially expressed genes in the core cellular subpopulations compared with other categories, analyzed the causal relationship between the differentially expressed genes and PsA by Mendelian randomization and analyzed the enriched pathways of key genes. RESULTS T cell subsets were represented in a significant proportion of both PsA and senescent patients, in which CD8-CM was expressed up-regulated in both PsA and senescent populations, and a total of 98 differentially expressed genes were obtained, and a Mendelian randomization study revealed that TGFBR3, PPP3CC, and APOBEC3G were causally associated with PsA. Colocalization analysis was performed to identify co-localized association signals in the PsA GWAS results and expression quantitative trait Loci (eQTL) dataset of key genes, and metabolic pathways that were predominantly enriched for key genes were analyzed by Kyoto Encyclopedia of Genes and Genomes (KEGG). CONCLUSIONS In this study, we found that CD8-CM expression was up-regulated in PsA and senescent populations, and identified key genes for PsA and senescence: TGFBR3, PPP3CC and APOBEC3G. This provides new insights into the pathogenesis and combined treatment of PsA and aging.
Collapse
Affiliation(s)
- Shuang Liu
- Yu-Yue Pathology Scientific Research Center, China.
| | - Peng Pu
- Yu-Yue Pathology Scientific Research Center, China.
| | - Qing Xiang
- Yu-Yue Pathology Scientific Research Center, China.
| | - Xiangling Pu
- Yu-Yue Pathology Scientific Research Center, China.
| |
Collapse
|
3
|
Nakamura T, Nakamura HM, Iwasaki Y, Enomoto-Iwamoto M, Nakashima N, Fukumoto S, Pacifici M, Iwamoto M, Wakamori M. Molecular mechanism for transcriptional regulation of the parathyroid hormone gene by Epiprofin. FEBS J 2025. [PMID: 40164571 DOI: 10.1111/febs.70085] [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: 10/05/2024] [Revised: 01/29/2025] [Accepted: 03/21/2025] [Indexed: 04/02/2025]
Abstract
Epiprofin (Epfn), an Sp/KLF family transcription factor that regulates cell proliferation and determines cell fates, is essential for normal skin, hair follicle, and tooth development. We found that Epfn was expressed in parathyroid glands, and Epfn-knockout mice displayed elevated serum parathyroid hormone (PTH) concentrations, decreased bone volume, and intracranial ectopic calcification. To investigate the role of Epfn in the regulation of PTH expression, parathyroid gland explant and parathyroid cell line culture methods were used. Epfn expression was found to be upregulated in response to an increase in extracellular calcium concentration, whereas PTH expression was downregulated, thus demonstrating an inverse correlation. Forced expression of Epfn inhibited PTH gene expression and PTH promoter reporter activity in parathyroid cells. In addition, with a high extracellular calcium concentration, Epfn silencing in cultured parathyroid glands failed to block PTH gene expression. ChIP-qPCR analysis also revealed Epfn binding in the proximal region of the PTH promoter, which was accelerated in the presence of a high concentration of calcium ions. The results from our in vitro and ex vivo analyses suggest that Epfn is a newly identified negative regulator of PTH transcription by regulating the proximal PTH promoter. Furthermore, the expression of Epfn was significantly reduced in parathyroid adenomas of primary hyperparathyroidism patients. The identification of Epfn as a potential therapeutic target for the control of PTH production in hyperparathyroidism patients opens new avenues for targeted treatment approaches.
Collapse
Affiliation(s)
- Takashi Nakamura
- Division of Molecular Pharmacology & Cell Biophysics, Department of Disease Management Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Hannah M Nakamura
- Division of Nephrology and Endocrinology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yasumasa Iwasaki
- Department of Endocrinology, Metabolism and Nephrology, Kochi Medical School, Kochi University, Japan
| | - Motomi Enomoto-Iwamoto
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Noriaki Nakashima
- Department of Breast Cancer and Endocrine Surgery, Tohoku University Hospital, Sendai, Japan
| | - Satoshi Fukumoto
- Division of Pediatric Dentistry, Kyushu University Graduate School of Dentistry, Fukuoka, Japan
| | - Maurizio Pacifici
- Division of Orthopedic Surgery, Department of Surgery, Children's Hospital of Philadelphia, PA, USA
| | - Masahiro Iwamoto
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Minoru Wakamori
- Division of Molecular Pharmacology & Cell Biophysics, Department of Disease Management Dentistry, Tohoku University Graduate School of Dentistry, Sendai, Japan
| |
Collapse
|
4
|
Jin J, Zhang R, Li J, Gao F, Liao Z, Yu Y, Wang Y, Bucci D, Xiao M, Ma R, Ma Q, Gao S, Lio J, Novais F, Huang SCC, Zhu J, Ghoneim H, Wen H, Li Z, Sun N, Xin G. The NAE1-mediated neddylation operates as an essential post-translational modification checkpoint for effector CD8 + T cells. Proc Natl Acad Sci U S A 2025; 122:e2424061122. [PMID: 40030035 PMCID: PMC11912420 DOI: 10.1073/pnas.2424061122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/02/2025] [Indexed: 03/19/2025] Open
Abstract
Optimal activation of CD8+ T cells is crucial for immunity-mediated destruction of cancer, requiring a substantial amount of proteins involved in metabolism, proliferation, and effector function. Despite extensive studies emphasizing the role of transcriptional regulation in this process, paired transcriptomic and proteomic analyses reveal that the RNA profile is poorly correlated with protein levels. This discrepancy underscores the importance of post-translational modifications (PTMs) in controlling protein abundance during activation. However, the impact of PTMs on the CD8+ T cell protein dynamic remains underexplored. We identify that neddylation, a recently discovered PTM, is activated in response to T cell receptor (TCR) stimulation and enriched in effector CD8+ T cells from colon cancer patients. Mechanistically, we found the rate-limiting enzyme of neddylation, neural precursor cell expressed developmentally down-regulated protein 8 activating enzyme E1 (NAE1), is induced by the NFATc1, a critical transcription factor downstream of TCR signaling. Our observation revealed that genetic ablation of NAE1 significantly disturbed the proteomic landscape related to activation and mitochondrial function. As a result, CD8+ T cells lacking NAE1 exhibited severely compromised activation, proliferation, and survival, which was accompanied by impaired mitochondrial function. Consistently, deletion of NAE1 in CD8+ T cells abolished their antitumor function and promoted tumor progression. By contrast, the overexpression of NAE1 significantly improved the function of tumor-infiltrating CD8+ T cells. Overall, we uncovered neddylation, a previously underappreciated PTM, as a proteomic checkpoint for CD8+ T cell activation. Enforced expression of NAE1 offers promising therapeutic potential for boosting the antitumor CD8+ T cell responses.
Collapse
Affiliation(s)
- Jiacheng Jin
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Ruohan Zhang
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH43210
| | - Jianying Li
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Fengxia Gao
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Zhiwei Liao
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Yanbao Yu
- Department of Chemistry and Biochemistry, Mass Spectrometry Facility, University of Delaware, Newark, DE19716
| | - Yi Wang
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
| | - Donna Bucci
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Min Xiao
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Ruilin Ma
- Department of Chemistry, New York University, New York, NY10003
| | - Qin Ma
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH43210
| | - Shuaixin Gao
- Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, OH43210
| | - Jerry Lio
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Fernanda Novais
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Stanley Ching-Cheng Huang
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Jiangjiang Zhu
- Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, OH43210
| | - Hazem Ghoneim
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Haitao Wen
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| | - Zihai Li
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
| | - Nuo Sun
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH43210
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
| | - Gang Xin
- Pelotonia Institute for Immuno-oncology, Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, College of Medicine, The Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, OH43210
| |
Collapse
|
5
|
Imamichi T, Yang J, Chen Q, Goswami S, Marquez M, Kariyawasam U, Sharma HN, Wiscovitch-Russo R, Li X, Aioi A, Adelsberger JW, Chang W, Higgins J, Sui H. Interleukin-27-polarized HIV-resistant M2 macrophages are a novel subtype of macrophages that express distinct antiviral gene profiles in individual cells: implication for the antiviral effect via different mechanisms in the individual cell-dependent manner. Front Immunol 2025; 16:1550699. [PMID: 40129989 PMCID: PMC11931227 DOI: 10.3389/fimmu.2025.1550699] [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: 12/23/2024] [Accepted: 02/17/2025] [Indexed: 03/26/2025] Open
Abstract
Introduction Interleukin (IL)-27 is an anti-viral cytokine. IL-27-treated monocyte-derived macrophages (27-Mac) suppressed HIV replication. Macrophages are generally divided into two subtypes, M1 and M2 macrophages. M2 macrophages can be polarized into M2a, M2b, M2c, and M2d by various stimuli. IL-6 and adenosine induce M2d macrophages. Since IL-27 is a member of the IL-6 family of cytokines, 27-Mac was considered M2d macrophages. In the current study, we compared biological function and gene expression profiles between 27-Mac and M2d subtypes. Methods Monocytes derived from health donors were differentiated to M2 using macrophage colony-stimulating factor. Then, the resulting M2 was polarized into different subtypes using IL-27, IL-6, or BAY60-658 (an adenosine analog). HIV replication was monitored using a p24 antigen capture assay, and the production of reactive oxygen species (ROS) was determined using a Hydrogen Peroxide Assay. Phagocytosis assay was run using GFP-labeled opsonized E. coli. Cytokine production was detected by the IsoPlexis system, and the gene expression profiles were analyzed using single-cell RNA sequencing (scRNA-seq). Results and Discussion 27-Mac and BAY60-658-polarized M2d (BAY-M2d) resisted HIV infection, but IL-6-polarized M2d (6-M2d) lacked the anti-viral effect. Although phagocytosis activity was comparable among the three macrophages, only 27-Mac, but neither 6-M2d nor BAY-M2d, enhanced the generation of ROS. The cytokine-producing profile of 27-Mac did not resemble that of the two subtypes. The scRNA-seq revealed that 27-Mac exhibited a different clustering pattern compared to other M2ds, and each 27-Mac expressed a distinct combination of anti-viral genes. Furthermore, 27-Mac did not express the biomarkers of M2a, M2b, and M2c. However, it significantly expressed CD38 (p<0.01) and secreted CXCL9 (p<0.001), which are biomarkers of M1. Conclusions These data suggest that 27-Mac may be classified as either an M1-like subtype or a novel subset of M2, which resists HIV infection mediated by a different mechanism in individual cells using different anti-viral gene products. Our results provide a new insight into the function of IL-27 and macrophages.
Collapse
Affiliation(s)
- Tomozumi Imamichi
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Jun Yang
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Qian Chen
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Suranjana Goswami
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Mayra Marquez
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Udeshika Kariyawasam
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Homa Nath Sharma
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Rosana Wiscovitch-Russo
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Xuan Li
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Akihiro Aioi
- Laboratory of Basic Research, Septem-Soken, Osaka, Japan
| | - Joseph W. Adelsberger
- AIDS Monitoring Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Weizhong Chang
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Jeanette Higgins
- AIDS Monitoring Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Hongyan Sui
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| |
Collapse
|
6
|
Bei Y, Si X, Ma W, Qi P, Ye Y. The Role of NFAT5 in Immune Response and Antioxidant Defense in the Thick-Shelled Mussel ( Mytilus coruscus). Animals (Basel) 2025; 15:726. [PMID: 40076009 PMCID: PMC11898675 DOI: 10.3390/ani15050726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/18/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Nuclear Factor of Activated T Cells 5 (NFAT5) is a transcription factor that plays a pivotal role in immune regulation. While its functions have been extensively studied in mammalian immune systems, its role in marine invertebrates, particularly in bivalves, remains largely unexplored. This study provides the first characterization of the NFAT5 gene in the thick-shelled mussel (Mytilus coruscus), investigating its evolutionary characteristics and immunological functions. Using direct RNA sequencing, McNFAT5 was comprehensively analyzed, revealing its critical involvement in the innate immune response of M. coruscus to Vibrio alginolyticus challenge. Differential expression patterns of McNFAT5 were observed across various tissues with the highest expression detected in hemolymphs. The knockdown of McNFAT5 using small interfering RNA (siRNA) led to a significant reduction in the activities of superoxide dismutase (SOD), Na+/K+-ATPase, and antioxidant enzymes compared to levels observed post-infection. These findings highlight the central role of McNFAT5 in modulating antioxidant defense mechanisms. In conclusion, McNFAT5 is a key regulatory factor in the innate immune system of M. coruscus, providing valuable insights into the immune adaptive mechanisms and evolutionary mechanisms of bivalve immunity. This study contributes to a deeper understanding of the immune regulatory networks in marine invertebrates.
Collapse
Affiliation(s)
- Yijiang Bei
- Zhejiang Fisheries Technical Extension Center, Hangzhou No. 181, Jingchang Road, Wuchang Street, Yuhang District, Hangzhou 310012, China; (Y.B.); (W.M.)
| | - Xirui Si
- National Engineering Research Center for Marine Aquaculture, Zhejiang Ocean University, Zhoushan 316022, China;
| | - Wenjun Ma
- Zhejiang Fisheries Technical Extension Center, Hangzhou No. 181, Jingchang Road, Wuchang Street, Yuhang District, Hangzhou 310012, China; (Y.B.); (W.M.)
| | - Pengzhi Qi
- National Engineering Research Center for Marine Aquaculture, Zhejiang Ocean University, Zhoushan 316022, China;
| | - Yingying Ye
- National Engineering Research Center for Marine Aquaculture, Zhejiang Ocean University, Zhoushan 316022, China;
| |
Collapse
|
7
|
Lee I, Deng S, Ning Y. Optimal variable clustering for high-dimensional matrix valued data. INFORMATION AND INFERENCE : A JOURNAL OF THE IMA 2025; 14:iaaf001. [PMID: 40084241 PMCID: PMC11899537 DOI: 10.1093/imaiai/iaaf001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 10/07/2024] [Accepted: 01/24/2025] [Indexed: 03/16/2025]
Abstract
Matrix valued data has become increasingly prevalent in many applications. Most of the existing clustering methods for this type of data are tailored to the mean model and do not account for the dependence structure of the features, which can be very informative, especially in high-dimensional settings or when mean information is not available. To extract the information from the dependence structure for clustering, we propose a new latent variable model for the features arranged in matrix form, with some unknown membership matrices representing the clusters for the rows and columns. Under this model, we further propose a class of hierarchical clustering algorithms using the difference of a weighted covariance matrix as the dissimilarity measure. Theoretically, we show that under mild conditions, our algorithm attains clustering consistency in the high-dimensional setting. While this consistency result holds for our algorithm with a broad class of weighted covariance matrices, the conditions for this result depend on the choice of the weight. To investigate how the weight affects the theoretical performance of our algorithm, we establish the minimax lower bound for clustering under our latent variable model in terms of some cluster separation metric. Given these results, we identify the optimal weight in the sense that using this weight guarantees our algorithm to be minimax rate-optimal. The practical implementation of our algorithm with the optimal weight is also discussed. Simulation studies show that our algorithm performs better than existing methods in terms of the adjusted Rand index (ARI). The method is applied to a genomic dataset and yields meaningful interpretations.
Collapse
Affiliation(s)
- Inbeom Lee
- Booth School of Business, University of Chicago, 5807 S. Woodlawn Ave., Chicago, IL 60637, USA
| | - Siyi Deng
- Amazon, 425 106th Ave NE, Bellevue, WA 98004, USA
| | - Yang Ning
- Department of Statistics and Data Science, Cornell University, 1198 Comstock Hall, 129 Garden Ave., Ithaca, NY 14853, USA
| |
Collapse
|
8
|
Li H, Cao Y, Zhao G, Wang G, Huang G, Wang L, Ding Z, Tang PMK, Li C. ORAI2 is Important for the Development of Early-Stage Postirradiation Fibrosis in Salivary Glands. Int J Radiat Oncol Biol Phys 2025; 121:798-810. [PMID: 39384103 DOI: 10.1016/j.ijrobp.2024.09.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 09/10/2024] [Accepted: 09/20/2024] [Indexed: 10/11/2024]
Abstract
PURPOSE Although postirradiation hyposalivation significantly impairs patient quality of life, the underlying mechanisms driving radiation-induced salivary gland fibrosis and hyposalivation remain poorly understood. This study aims to explore the role of calcium-mediated signaling pathways in radiation-induced salivary gland fibrosis. METHODS AND MATERIALS Primary human submandibular gland (SG) cells and C57BL/6J female mouse SGs were exposed to irradiation to model fibrosis development. Following 15 Gy irradiation exposure, RNA sequencing and bioinformatic analysis were conducted on mouse SGs. The effects of store-operated calcium entry (SOCE) inhibition using SKF96365 and YM58483 on fibrosis markers were assessed in vitro and in vivo. Additionally, the involvement of ORAI2 protein and the newly identified JNK/NFAT1/transforming growth factor β1 (TGF-β1) signaling axis in SG fibrosis was explored. RESULTS We identified that the calcium release-activated calcium modulator ORAI2 was important in promoting early-stage postirradiation fibrosis in SGs. Calcium channel signaling was activated in both human patients and irradiated C57BL/6J female mice SGs. Inhibition of SOCE signaling effectively blocked fibrosis in an ORAI2-dependent manner 30 days after irradiation. Our mechanistic studies revealed a novel ORAI2/JNK/NFAT1 axis within the SOCE pathway critical in driving TGF-β1-mediated fibrogenesis. Encouragingly, pharmacologic inhibition of NFAT1 significantly mitigated radiation-induced SG fibrosis and restored saliva flow to 84.61% of normal levels in treated mice 30 days after irradiation, without detectable side effects. CONCLUSIONS Our findings highlight the significance of the ORAI2-mediated calcium signaling pathway, specifically via the ORAI2/JNK/NFAT1 axis, in promoting TGF-β1 expression and contributing to the development of early-stage salivary gland fibrosis following irradiation exposure. Targeting the ORAI2/JNK/NFAT1 axis emerges as a promising therapeutic strategy to alleviate radiation-induced hyposalivation and fibrosis, potentially improving the quality of life for patients undergoing radiation therapy.
Collapse
Affiliation(s)
- Honglin Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yubin Cao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Guile Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Guanru Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Guangzhao Huang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lei Wang
- Department of Dentistry, The Second People's Hospital of Tibet Autonomous Region, Lhasa, China
| | - Zhangfan Ding
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong.
| | - Chunjie Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
9
|
Krings KS, Ritchie A, Schmitt L, Hatzfeld J, Totzke G, Lenz T, Mendiburo MJ, Stork B, Teusch N, Proksch P, Stühler K, Müller L, Wesselborg S. The Polybrominated Diphenyl Ether Bromoxib Disrupts Nuclear Import and Export by Affecting Nucleoporins of the Nuclear Pore Complex. Mar Drugs 2025; 23:108. [PMID: 40137294 PMCID: PMC11943847 DOI: 10.3390/md23030108] [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: 02/03/2025] [Revised: 02/24/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) are natural products with potent antimicrobial and antineoplastic activity. We have previously shown that the polybrominated diphenyl ether bromoxib (4,5,6-tribromo-2-(2',4'-dibromophenoxy) phenol), isolated from the marine sponge Dysidea species, exhibits a strong cytotoxic potential in leukemia and lymphoma cells by targeting mitochondrial metabolism. Here, using a mass spectrometric thermal proteome profiling (TPP) approach, we observed that bromoxib induces a rapid reduction in the levels of 19 nucleoporins (NUPs) that are part of the nuclear pore complex (NPC). This apparently affected the functionality of the NPC, as evidenced by the bromoxib-mediated inhibition of the nuclear translocation and subsequent gene reporter activity of transcription factors such as nuclear factor of activated T cells (NFAT) and nuclear factor κB (NF-κB). In addition, bromoxib inhibited the nuclear export of the mRNA of the human immunodeficiency virus transactivator of transcription (HIV-Tat) and the subsequent import of the HIV-Tat protein into the nucleus as determined by the decrease in Tat-dependent gene reporter luciferase activity. Inhibition of nuclear mRNA-export also affected expression of the short-lived anti-apoptotic Bcl-2 protein Mcl-1, which has been shown to induce apoptosis. Thus, its ability to target both mitochondrial metabolism and the NPC renders bromoxib a promising anticancer agent.
Collapse
Affiliation(s)
- Karina S. Krings
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Universitaetsstraße 1, 40225 Duesseldorf, Germany; (K.S.K.); (L.S.)
| | - Anastasia Ritchie
- Institute of Virology, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Universitaetsstraße 1, 40225 Duesseldorf, Germany; (A.R.)
| | - Laura Schmitt
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Universitaetsstraße 1, 40225 Duesseldorf, Germany; (K.S.K.); (L.S.)
| | - Judith Hatzfeld
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Universitaetsstraße 1, 40225 Duesseldorf, Germany; (K.S.K.); (L.S.)
| | - Gudrun Totzke
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Universitaetsstraße 1, 40225 Duesseldorf, Germany; (K.S.K.); (L.S.)
| | - Thomas Lenz
- Molecular Proteomics Laboratory, Biological-Medical-Research Center (BMFZ), Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Universitaetsstraße 1, 40225 Duesseldorf, Germany
| | - María José Mendiburo
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Universitaetsstraße 1, 40225 Duesseldorf, Germany; (K.S.K.); (L.S.)
| | - Björn Stork
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Universitaetsstraße 1, 40225 Duesseldorf, Germany; (K.S.K.); (L.S.)
| | - Nicole Teusch
- Institute of Pharmaceutical Biology and Biotechnology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, Universitaetsstraße 1, 40225 Duesseldorf, Germany; (N.T.)
| | - Peter Proksch
- Institute of Pharmaceutical Biology and Biotechnology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, Universitaetsstraße 1, 40225 Duesseldorf, Germany; (N.T.)
| | - Kai Stühler
- Molecular Proteomics Laboratory, Biological-Medical-Research Center (BMFZ), Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Universitaetsstraße 1, 40225 Duesseldorf, Germany
| | - Lisa Müller
- Institute of Virology, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Universitaetsstraße 1, 40225 Duesseldorf, Germany; (A.R.)
| | - Sebastian Wesselborg
- Institute for Molecular Medicine I, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, Universitaetsstraße 1, 40225 Duesseldorf, Germany; (K.S.K.); (L.S.)
- Center for Integrated Oncology Aachen-Bonn-Cologne-Duesseldorf (CIO ABCD), 40225 Duesseldorf, Germany
| |
Collapse
|
10
|
Adlakha A, Williams TJ, Shou X, Reed AK, Lenhard B, Armstrong-James D. Interferon-gamma rescues dendritic cell calcineurin-dependent responses to Aspergillus fumigatus via Stat3 to Stat1 switching. iScience 2025; 28:111535. [PMID: 39898039 PMCID: PMC11787545 DOI: 10.1016/j.isci.2024.111535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/11/2024] [Accepted: 12/03/2024] [Indexed: 02/04/2025] Open
Abstract
Invasive pulmonary aspergillosis is a lethal opportunistic fungal infection in transplant recipients receiving calcineurin inhibitors. We previously identified a role for the calcineurin pathway in innate immune responses to A. fumigatus and have used exogenous interferon-gamma successfully to treat aspergillosis in this setting. Here we show that calcineurin inhibitors block dendritic cell maturation in response to A. fumigatus, impairing the Th1 polarization of CD4 cells. Interferon gamma, an immunotherapeutic option for invasive aspergillosis, restored maturation and promoted Th1 polarization via a dendritic cell dependent effect that was co-dependent on T cell interaction. We find that interferon gamma activates alternative transcriptional pathways to calcineurin-NFAT for the augmentation of pathogen handling. Histone modification ChIP-Seq analysis revealed dominant control by an interferon gamma induced regulatory switch from STAT3 to STAT1 transcription factor binding underpinning these observations. These findings provide key insight into the mechanisms of immunotherapy in organ transplant recipients with invasive fungal diseases.
Collapse
Affiliation(s)
- Amit Adlakha
- Department of Infectious Disease, Faculty of Medicine, Imperial College, London SW7 2AZ, UK
- MRC London Institute of Medical Sciences, Imperial College, London W12 0NN, UK
- Department of Cardiothoracic Transplantation and Mechanical Support, Harefield Hospital, Royal Brompton & Harefield Hospitals, Part of Guy’s and St. Thomas' NHS Foundation Trust & Imperial College, London UB9 6JH, UK
| | - Thomas J. Williams
- Department of Infectious Disease, Faculty of Medicine, Imperial College, London SW7 2AZ, UK
- Department of Cardiothoracic Transplantation and Mechanical Support, Harefield Hospital, Royal Brompton & Harefield Hospitals, Part of Guy’s and St. Thomas' NHS Foundation Trust & Imperial College, London UB9 6JH, UK
| | - Xinxin Shou
- Department of Infectious Disease, Faculty of Medicine, Imperial College, London SW7 2AZ, UK
| | - Anna K. Reed
- Department of Cardiothoracic Transplantation and Mechanical Support, Harefield Hospital, Royal Brompton & Harefield Hospitals, Part of Guy’s and St. Thomas' NHS Foundation Trust & Imperial College, London UB9 6JH, UK
| | - Boris Lenhard
- MRC London Institute of Medical Sciences, Imperial College, London W12 0NN, UK
| | - Darius Armstrong-James
- Department of Infectious Disease, Faculty of Medicine, Imperial College, London SW7 2AZ, UK
- Department of Cardiothoracic Transplantation and Mechanical Support, Harefield Hospital, Royal Brompton & Harefield Hospitals, Part of Guy’s and St. Thomas' NHS Foundation Trust & Imperial College, London UB9 6JH, UK
| |
Collapse
|
11
|
Ye X, Liu R. Exercise-induced cytosolic calcium oscillations: mechanisms and modulation of T-cell function. Biochem Biophys Res Commun 2025; 748:151321. [PMID: 39826528 DOI: 10.1016/j.bbrc.2025.151321] [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: 08/11/2024] [Revised: 12/26/2024] [Accepted: 01/11/2025] [Indexed: 01/22/2025]
Abstract
This study investigated time-dependent changes in intracellular Ca2⁺ levels in T cells, regulatory mechanisms, and functional effects after acute exercise. Male C57BL/6 mice were assigned to control and exercise groups, with the latter sacrificed at different intervals post-exercise. Murine splenic lymphocytes were isolated, and cytosolic Ca2⁺ levels were measured using Fluo-3/AM. T-cell proliferation was assessed by flow cytometry and CFSE labeling, apoptosis by Annexin V/PI staining, and cytokine levels by CBA. RNA sequencing results were validated by qRT-PCR. The findings revealed that exercise significantly altered intracellular calcium oscillations in CD3+ cells, leading to reduced mitogen-stimulated proliferation, increased IL-6, IL-5, and IL-13 production, and decreased IL-2 secretion. Additionally, there was an increase in the apoptotic fraction of CD3+ cells, with upregulated expression of Cav1.1, Cav3.2, Cav3.3, SERCA2B, PKCθ, Bcl-xL, and FADD, and downregulated Ryr3 (p < 0.05). Transcriptomic analysis identified 607 differentially expressed genes involved in calcium ion binding and related pathways, including calcium signaling and cytokine-cytokine receptor interactions. Thus, acute exercise induces specific calcium oscillation patterns in T cells, mediated by PKCθ, affecting proliferation, apoptosis, and cytokine production. These changes are attributed to increased calcium influx through Cav1.1, Cav3.2, and Cav3.3 channels, decreased calcium reuptake via SERCA2B, and reduced calcium release through Ryr3. This research provides novel insights into how exercise modulates immune cell function by altering calcium levels, potential implications for enhancing immune responses or reducing inflammation.
Collapse
Affiliation(s)
- Xing Ye
- School of Physical Education, China University of Geosciences (Wuhan), Wuhan, China
| | - Renyi Liu
- School of Physical Education, China University of Geosciences (Wuhan), Wuhan, China.
| |
Collapse
|
12
|
Jiang W, Guan B, Sun H, Mi Y, Cai S, Wan R, Li X, Lian P, Li D, Zhao S. WNT11 Promotes immune evasion and resistance to Anti-PD-1 therapy in liver metastasis. Nat Commun 2025; 16:1429. [PMID: 39920102 PMCID: PMC11806061 DOI: 10.1038/s41467-025-56714-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: 06/25/2024] [Accepted: 01/27/2025] [Indexed: 02/09/2025] Open
Abstract
Liver metastasis (LM) poses a significant challenge in cancer treatment, with limited available therapeutic options and poor prognosis. Understanding the dynamics of tumor microenvironment (TME) and immune interactions is crucial for developing effective treatments. We find that WNT11 promoted CD8+ T-cell exclusion and suppression, which was correlated with poor prognosis in LM. Mechanistically, WNT11-overexpressing tumor cells directly reduce CD8+ T-cell recruitment and activity by decreasing CXCL10 and CCL4 expression through CAMKII-mediated β-catenin/AFF3 downregulation. WNT11-overexpressing tumor cells promote immunosuppressive macrophage polarization by inducing IL17D expression via the CAMKII/NF-κB pathway, which result in CD8+ T-cell suppression. Moreover, CAMKII inhibition increases the efficacy of anti-PD-1 therapy in mouse model of LM. Serum expression of WNT11 is identified as a potential minimally invasive biomarker in the management of colorectal cancer-LM with immunotherapy. Our findings highlight WNT11/CAMKII axis as a critical regulator of the TME and a promising target for immunotherapy in patients with LM.
Collapse
Affiliation(s)
- Weiliang Jiang
- Cancer Institute, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, No. 650 New Songjiang Road, Shanghai, China
| | - Bingjie Guan
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 650 New Songjiang Road, Shanghai, China
| | - Hongcheng Sun
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 650 New Songjiang Road, Shanghai, China
| | - Yushuai Mi
- Department of Gastrointestinal Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247 Beiyuan Road, Jinan, Shandong, China
| | - Sanjun Cai
- Cancer Institute, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dong'an Road, Shanghai, China
| | - Rong Wan
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, No. 650 New Songjiang Road, Shanghai, China
| | - Xinxiang Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dong'an Road, Shanghai, China
| | - Peng Lian
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dong'an Road, Shanghai, China
| | - Dawei Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dong'an Road, Shanghai, China.
| | - Senlin Zhao
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dong'an Road, Shanghai, China.
| |
Collapse
|
13
|
Zhou H, Chen P, Zhao C, Zou S, Wu H, Huang C, Hu H, Wu Q, Zhang C, Weng W. Fraxin inhibits ovariectomized-induced bone loss and osteoclastogenesis by suppressing ROS activity. Int Immunopharmacol 2025; 147:113871. [PMID: 39798467 DOI: 10.1016/j.intimp.2024.113871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 01/15/2025]
Abstract
Osteoporosis is characterized by increased osteoclast activity, which is strongly associated with increased levels of reactive oxygen species (ROS). Fraxin, a natural coumarin glycoside, has shown anti-inflammatory and antioxidant properties, but its effects on bone homeostasis are obscure. The effects of fraxin on osteoclast formation and activation were measured via an in vitro osteoclastogenesis assay. Mitochondrial and total ROS production were evaluated with the aid of MitoSOX Red and DCFH-DA, respectively. Osteoclast-related gene expression analysis was performed via qPCR. Key proteins related to osteoclast formation, ROS scavenging, and ROS-regulated signaling, such as mitogen-activated protein kinases (MAPKs), NF-κB pathways, and nuclear factor of activated T cells 1 (NFATc1) signaling, were detected via western blotting. An ovariectomized mouse model was used to evaluate the therapeutic effects of fraxin in vivo. Fraxin inhibited osteoclastogenesis and osteoclast-related gene expression. Mechanistically, fraxin restored the levels of ROS-scavenging enzymes to inhibit ROS accumulation, eventually downregulating ROS-regulated signaling. The measurement of Micro-CT and histological analyses revealed that fraxin treatment significantly reduced OVX-induced bone loss by decreasing the number of osteoclasts. Fraxin shows promise as a novel therapeutic agent for osteoclast-related bone diseases, especially osteoporosis.
Collapse
Affiliation(s)
- Han Zhou
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Pianpian Chen
- Department of Pharmacy, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, China
| | - Chuanyong Zhao
- Department of Orthopedics, The Third Affiliated Hospital of Wenzhou Medical University & Rui'an People's Hospital, Rui'an, China
| | - Siyuan Zou
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hao Wu
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chenhao Huang
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hongwei Hu
- The First Clinical Medical Institute of Wenzhou Medical College, Wenzhou, China
| | - Qianmo Wu
- The First Clinical Medical Institute of Wenzhou Medical College, Wenzhou, China
| | - ChunWu Zhang
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Weidong Weng
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
14
|
Miyano T, Suzuki A, Konta H, Sakamoto N. Hyperosmotic Stress Promotes the Nuclear Translocation of TFEB in Tubular Epithelial Cells Depending on Intracellular Ca 2+ Signals via TRPML Channels. Cell Mol Bioeng 2025; 18:39-52. [PMID: 39949488 PMCID: PMC11814421 DOI: 10.1007/s12195-024-00839-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 12/13/2024] [Indexed: 02/16/2025] Open
Abstract
Purpose We previously demonstrated that hyperosmotic stress, which acts as mechanical stress, induces autophagy of tubular epithelial cells. This study aims to elucidate the molecular mechanisms of hyperosmolarity-induced autophagy. The research question addresses how hyperosmotic stress activates autophagy through transcription factor EB (TFEB) and Ca2+ signaling pathways, contributing to understanding cellular responses to mechanical stress. Methods NRK-52E normal rat kidney cells were subjected to hyperosmotic stress using mannitol-containing medium. Fluorescence microscopy was utilized to observe TFEB nuclear translocation, a crucial event in autophagy regulation. An intracellular Ca2+ chelator, BAPTA-AM, and a calcineurin inhibitor were used to dissect the Ca2+ signaling pathway involved in TFEB translocation. The phosphorylation of p70S6K, a substrate of the mammalian target of rapamycin complex 1 kinase, was analyzed to explore its role in TFEB localization. Additionally, the function of transient receptor potential mucolipin 1 (TRPML1), an intracellular Ca2+ channel, was assessed using pharmacological inhibition to determine its impact on TFEB translocation and autophagy marker LC3-II levels. Results Mannitol-induced hyperosmotic stress promoted the nuclear translocation of TFEB, which was completely abolished by treatment with BAPTA-AM. Inhibition of calcineurin suppressed TFEB nuclear translocation under hyperosmolarity, indicating that a signaling pathway governed by intracellular Ca2+ is involved in TFEB's nuclear translocation. In contrast, hyperosmotic stress did not significantly alter p70S6K phosphorylation. Pharmacological inhibition of TRPML1 attenuated both TFEB nuclear translocation and LC3-II upregulation in response to hyperosmotic stress. Conclusions Hyperosmotic stress promotes TFEB nuclear localization, and TRPML1-induced activation of calcineurin is involved in the mechanism of hyperosmolarity-induced autophagy. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-024-00839-6.
Collapse
Affiliation(s)
- Takashi Miyano
- Department of Medical and Robotic Engineering Design, Tokyo University of Science, Tokyo, Japan
- Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Tokyo, Japan
| | - Atsushi Suzuki
- Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Tokyo, Japan
| | - Hisaaki Konta
- Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Tokyo, Japan
| | - Naoya Sakamoto
- Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Tokyo, Japan
| |
Collapse
|
15
|
Patalano SD, Fuxman Bass P, Fuxman Bass JI. Transcription factors in the development and treatment of immune disorders. Transcription 2025; 16:118-140. [PMID: 38100543 PMCID: PMC11970766 DOI: 10.1080/21541264.2023.2294623] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023] Open
Abstract
Immune function is highly controlled at the transcriptional level by the binding of transcription factors (TFs) to promoter and enhancer elements. Several TF families play major roles in immune gene expression, including NF-κB, STAT, IRF, AP-1, NRs, and NFAT, which trigger anti-pathogen responses, promote cell differentiation, and maintain immune system homeostasis. Aberrant expression, activation, or sequence of isoforms and variants of these TFs can result in autoimmune and inflammatory diseases as well as hematological and solid tumor cancers. For this reason, TFs have become attractive drug targets, even though most were previously deemed "undruggable" due to their lack of small molecule binding pockets and the presence of intrinsically disordered regions. However, several aspects of TF structure and function can be targeted for therapeutic intervention, such as ligand-binding domains, protein-protein interactions between TFs and with cofactors, TF-DNA binding, TF stability, upstream signaling pathways, and TF expression. In this review, we provide an overview of each of the important TF families, how they function in immunity, and some related diseases they are involved in. Additionally, we discuss the ways of targeting TFs with drugs along with recent research developments in these areas and their clinical applications, followed by the advantages and disadvantages of targeting TFs for the treatment of immune disorders.
Collapse
Affiliation(s)
- Samantha D. Patalano
- Biology Department, Boston University, Boston, MA, USA
- Molecular Biology, Cellular Biology and Biochemistry Program, Boston University, Boston, MA, USA
| | - Paula Fuxman Bass
- Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Juan I. Fuxman Bass
- Biology Department, Boston University, Boston, MA, USA
- Molecular Biology, Cellular Biology and Biochemistry Program, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| |
Collapse
|
16
|
Bercusson A, Williams TJ, Simmonds NJ, Alton EWFW, Griesenbach U, Shah A, Warris A, Armstrong-James D. Increased NFAT and NFκB signalling contribute to the hyperinflammatory phenotype in response to Aspergillus fumigatus in a mouse model of cystic fibrosis. PLoS Pathog 2025; 21:e1012784. [PMID: 39903773 PMCID: PMC11957335 DOI: 10.1371/journal.ppat.1012784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/31/2025] [Accepted: 01/16/2025] [Indexed: 02/06/2025] Open
Abstract
Aspergillus fumigatus (Af) is a major mould pathogen found ubiquitously in the air. It commonly infects the airways of people with cystic fibrosis (CF) leading to Aspergillus bronchitis or allergic bronchopulmonary aspergillosis. Resident alveolar macrophages and recruited neutrophils are important first lines of defence for clearance of Af in the lung. However, their contribution to the inflammatory phenotype in CF during Af infection is not well understood. Here, utilising CFTR deficient mice we describe a hyperinflammatory phenotype in both acute and allergic murine models of pulmonary aspergillosis. We show that during aspergillosis, CFTR deficiency leads to increased alveolar macrophage death and persistent inflammation of the airways in CF, accompanied by impaired fungal control. Utilising CFTR deficient murine cells and primary human CF cells we show that at a cellular level there is increased activation of NFκB and NFAT in response to Af which, as in in vivo models, is associated with increased cell death and reduced fungal control. Taken together, these studies indicate that CFTR deficiency promotes increased activation of inflammatory pathways, the induction of macrophage cell death and reduced fungal control contributing to the hyper-inflammatory of pulmonary aspergillosis phenotypes in CF.
Collapse
Affiliation(s)
- Amelia Bercusson
- Department of Infectious Disease, Faculty of Medicine, Imperial College, London, United Kingdom
- Cystic Fibrosis Unit, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Thomas J. Williams
- Department of Infectious Disease, Faculty of Medicine, Imperial College, London, United Kingdom
- Department of Cardiothoracic Transplantation and Mechanical Support, Harefield Hospital, Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Nicholas J. Simmonds
- Adult Cystic Fibrosis Centre, Royal Brompton Hospital, London, United Kingdom
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Eric WFW Alton
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Uta Griesenbach
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Anand Shah
- Department of Infectious Disease, Faculty of Medicine, Imperial College, London, United Kingdom
- Department of Respiratory Medicine, Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Adilia Warris
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Darius Armstrong-James
- Department of Infectious Disease, Faculty of Medicine, Imperial College, London, United Kingdom
- Department of Respiratory Medicine, Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
17
|
Wang Y, Zhuang X, Qi Y, Yiu L, Li Z, Chan YW, Liu X, Tsang SY. TRPC3-mediated NFATc1 calcium signaling promotes triple negative breast cancer migration through regulating glypican-6 and focal adhesion. Pflugers Arch 2025; 477:253-272. [PMID: 39436410 PMCID: PMC11762004 DOI: 10.1007/s00424-024-03030-y] [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: 04/20/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024]
Abstract
Canonical transient receptor potential isoform 3 (TRPC3), a calcium-permeable non-selective cation channel, has been reported to be upregulated in breast cancers and a modulator of cell migration. Calcium-sensitive transcription factor NFATc1, which is important for cell migration, was shown to be frequently activated in triple negative breast cancer (TNBC) biopsy tissues. However, whether TRPC3-mediated calcium influx would activate NFATc1 and affect the migration of TNBC cells, and, if yes, the underlying mechanisms involved, remain to be investigated. By immunostaining followed by confocal microscopy, TNBC lines MDA-MB-231 and BT-549 were both found to express TRPC3 on their plasma membrane while ER+ line MCF-7 and HER2+ line SK-BR3 do not. Blockade of TRPC3 by pharmacological inhibitor Pyr3 or stable knockdown of TRPC3 by lentiviral vector both inhibited cell migration as measured by wound healing assay. Importantly, blocking TRPC3 by Pyr3 or knockdown of TRPC3 both caused the translocation of NFATc1 from the nucleus to the cytosol as revealed by confocal microscopy. Interestingly, NFATc1 was found to bind to the promoter of glypican 6 (GPC6) as determined by chromatin immunoprecipitation assay. Consistently, knockdown of TRPC3 decreased the expression of GPC6 as revealed by western blotting. Moreover, long-term knockdown of GPC6 by lentiviral vector also consistently decreased the migration of TNBC cells. Intriguingly, GPC6 proteins physically interact with vinculin in MDA-MB-231 as determined by co-immunoprecipitation. Blockade of TRPC3, knockdown of TRPC3 or knockdown of GPC6 all induced larger, stabilized actin-bound peripheral focal adhesion (FA) formations in TNBC cells as determined by co-staining of actin and vinculin followed by confocal microscopy. These large, stabilized actin-bound peripheral FAs indicated a defective FA turnover, and were reported to be responsible for impairing directed cell migration. Our results suggest that, in TNBC cells, calcium influx through TRPC3 channel positively regulates NFATc1 nuclear translocation and GPC6 expression, which maintains the dynamics of FA turnover and optimal cell migration. Our study reveals a novel TRPC3-NFATc1-GPC6-vinculin signaling cascade in maintaining the migration of TNBC cells.
Collapse
Affiliation(s)
- Yan Wang
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiaosheng Zhuang
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yanxiang Qi
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lung Yiu
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhenping Li
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yuk Wah Chan
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xianji Liu
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Suk Ying Tsang
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Key Laboratory for Regenerative Medicine, Ministry of Education, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
18
|
Yip AJW, Lee YZ, Kow ASF, Wong CSA, Lee MT, Tham CL, Tan JW. Current utilization trend of immortalized mast cell lines in allergy research: a systematic review. Immunol Res 2025; 73:41. [PMID: 39838115 PMCID: PMC11750950 DOI: 10.1007/s12026-024-09562-w] [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: 02/25/2024] [Accepted: 11/07/2024] [Indexed: 01/23/2025]
Abstract
Today, in the modern world, allergic diseases, also described as atopic allergies, are classified as a type of multifactorial disorder due to the complex interplay between genetics, environment, and socioeconomic factors that influence the disease's manifestation, severity, and one's predisposition to allergic diseases. It is undeniable that many reported studies have pointed out that the mast cell is one of the leading key players involved in triggering an allergic reaction. To improve our understanding of the molecular and cellular mechanisms underlying allergy, various mast cell lines have been employed in vitro to study the pathogenesis of allergic diseases for the past decades. However, there is no consensus on many fundamental aspects associated with their use, such as the effects of culture media composition and the type of inducer used for cell degranulation. As the standardization of research protocols and disease models is crucial, we present the outcome of a systematic review of scientific articles using three major immortalized in vitro mast cell lines (HMC-1, LAD2, and RBL-2H3) to study allergy. This systematic review described the cell source, culture conditions, inducers used for degranulation, and mediators released for examination. We hope that the present systematic review may help to standardize the use of immortalized in vitro mast cell lines in allergy research and serve as a user's guide to understand the fundamental aspects of allergy as well to develop an effective allergy therapy in the future for the betterment of human good health and wellbeing.
Collapse
Affiliation(s)
- Ashley Jia Wen Yip
- School of Science, Monash University Malaysia, Jalan Lagoon SelatanSubang Jaya, 47500, Bandar Sunway, Selangor, Malaysia
| | - Yu Zhao Lee
- Faculty of Medicine and Health Sciences, UCSI University, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Audrey Siew Foong Kow
- Faculty of Pharmaceutical Sciences, UCSI University, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Carisa Su-Ann Wong
- School of Science, Monash University Malaysia, Jalan Lagoon SelatanSubang Jaya, 47500, Bandar Sunway, Selangor, Malaysia
| | - Ming-Tatt Lee
- Faculty of Pharmaceutical Sciences, UCSI University, Cheras, 56000, Kuala Lumpur, Malaysia
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Chau Ling Tham
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Natural Medicine and Product Research Laboratory (NaturMeds), Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Ji Wei Tan
- School of Science, Monash University Malaysia, Jalan Lagoon SelatanSubang Jaya, 47500, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
19
|
Hirsch MG, Pal S, Rashidi Mehrabadi F, Malikic S, Gruen C, Sassano A, Pérez-Guijarro E, Merlino G, Sahinalp SC, Molloy EK, Day CP, Przytycka TM. Stochastic modeling of single-cell gene expression adaptation reveals non-genomic contribution to evolution of tumor subclones. Cell Syst 2025; 16:101156. [PMID: 39701099 PMCID: PMC11867576 DOI: 10.1016/j.cels.2024.11.013] [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: 02/24/2024] [Revised: 08/30/2024] [Accepted: 11/18/2024] [Indexed: 12/21/2024]
Abstract
Cancer progression is an evolutionary process driven by the selection of cells adapted to gain growth advantage. We present a formal study on the adaptation of gene expression in subclonal evolution. We model evolutionary changes in gene expression as stochastic Ornstein-Uhlenbeck processes, jointly leveraging the evolutionary history of subclones and single-cell expression data. Applying our model to sublines derived from single cells of a mouse melanoma revealed that sublines with distinct phenotypes are underlined by different patterns of gene expression adaptation, indicating non-genetic mechanisms of cancer evolution. Sublines previously observed to be resistant to anti-CTLA4 treatment showed adaptive expression of genes related to invasion and non-canonical Wnt signaling, whereas sublines that responded to treatment showed adaptive expression of genes related to proliferation and canonical Wnt signaling. Our results suggest that clonal phenotypes emerge as the result of specific adaptivity patterns of gene expression. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- M G Hirsch
- National Library of Medicine (NLM), National Institutes of Health (NIH), Bethesda, MD 20892, USA; Department of Computer Science, University of Maryland, College Park, MD 20742, USA
| | - Soumitra Pal
- Neurobiology Neurodegeneration and Repair Lab, National Eye Institute (NEI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Farid Rashidi Mehrabadi
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA; Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Salem Malikic
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Charli Gruen
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Antonella Sassano
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Eva Pérez-Guijarro
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA; Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid (IIBM, CSIC-UAM), Madrid 28029, Spain
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - S Cenk Sahinalp
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Erin K Molloy
- Department of Computer Science, University of Maryland, College Park, MD 20742, USA; University of Maryland Institute for Advanced Computer Studies, College Park, MD 20742, USA
| | - Chi-Ping Day
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Teresa M Przytycka
- National Library of Medicine (NLM), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
20
|
Dome PA, Jeong P, Nam G, Jang H, Rivera A, Floyd Averette A, Park E, Liao TC, Ciofani M, Wu J, Chi JTA, Venters RA, Park HJ, Steinbach WJ, Juvvadi PR, Heitman J, Hong J. Structure-guided design and synthesis of C22- and C32-modified FK520 analogs with enhanced activity against human pathogenic fungi. Proc Natl Acad Sci U S A 2025; 122:e2419883121. [PMID: 39739817 PMCID: PMC11725869 DOI: 10.1073/pnas.2419883121] [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: 10/02/2024] [Accepted: 11/22/2024] [Indexed: 01/02/2025] Open
Abstract
Invasive fungal infections are a leading cause of death worldwide. Translating molecular insights into clinical benefits is challenging because fungal pathogens and their hosts share similar eukaryotic physiology. Consequently, current antifungal treatments have limited efficacy, may be poorly fungicidal in the host, can exhibit toxicity, and are increasingly compromised by emerging resistance. We have established that the phosphatase calcineurin (CaN) is required for invasive fungal disease and an attractive target for antifungal drug development. CaN is a druggable target, and there is vast clinical experience with the CaN inhibitors FK506 and cyclosporin A (CsA). However, while FK506 and its natural analog FK520 exhibit antifungal activity, they are also immunosuppressive in the host and thus not fungal-selective. We leverage our pathogenic fungal CaN-FK506-FKBP12 complex X-ray structures and biophysical data to support structure-based ligand design as well as structure-activity relationship analyses of broad-spectrum FK506/FK520 derivatives with potent antifungal activity and reduced immunosuppressive activity. Here, we apply molecular docking studies to develop antifungal C22- or C32-modified FK520 derivatives with improved therapeutic index scores. Among them, the C32-modified FK520 derivative JH-FK-44 (7) demonstrates a significantly improved therapeutic index compared to JH-FK-08, our lead compound to date. NMR binding studies with C32-derivatives are consistent with our hypothesis that C32 modifications disrupt the hydrogen bonding network in the human complex while introducing favorable electrostatic and cation-π interactions with the fungal FKBP12 R86 residue. These findings further reinforce calcineurin inhibition as a promising strategy for antifungal therapy.
Collapse
Affiliation(s)
| | | | - Gibeom Nam
- School of Pharmacy, Sungkyunkwan University, Suwon16419, Republic of Korea
| | - Hongjun Jang
- Department of Chemistry, Duke University, Durham, NC27708
| | - Angela Rivera
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC27710
| | - Anna Floyd Averette
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | - Eunchong Park
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC27710
| | - Tzu-Chieh Liao
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC27710
| | - Maria Ciofani
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC27710
| | - Jianli Wu
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | - Jen-Tsan Ashley Chi
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | - Ronald A. Venters
- Duke University Nuclear Magnetic Resonance Center, Duke University, Durham, NC27710
- Department of Radiology, Duke University Medical Center, Durham, NC27710
| | - Hyun-Ju Park
- School of Pharmacy, Sungkyunkwan University, Suwon16419, Republic of Korea
| | - William J. Steinbach
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR72202
| | - Praveen R. Juvvadi
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR72202
| | - Joseph Heitman
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC27710
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC27710
| | - Jiyong Hong
- Department of Chemistry, Duke University, Durham, NC27708
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC27710
| |
Collapse
|
21
|
Yuan Y, Liu H, Dai Z, He C, Qin S, Su Z. From Physiology to Pathology of Astrocytes: Highlighting Their Potential as Therapeutic Targets for CNS Injury. Neurosci Bull 2025; 41:131-154. [PMID: 39080102 PMCID: PMC11748647 DOI: 10.1007/s12264-024-01258-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/15/2024] [Indexed: 01/19/2025] Open
Abstract
In the mammalian central nervous system (CNS), astrocytes are the ubiquitous glial cells that have complex morphological and molecular characteristics. These fascinating cells play essential neurosupportive and homeostatic roles in the healthy CNS and undergo morphological, molecular, and functional changes to adopt so-called 'reactive' states in response to CNS injury or disease. In recent years, interest in astrocyte research has increased dramatically and some new biological features and roles of astrocytes in physiological and pathological conditions have been discovered thanks to technological advances. Here, we will review and discuss the well-established and emerging astroglial biology and functions, with emphasis on their potential as therapeutic targets for CNS injury, including traumatic and ischemic injury. This review article will highlight the importance of astrocytes in the neuropathological process and repair of CNS injury.
Collapse
Affiliation(s)
- Yimin Yuan
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
- Department of Pain Medicine, School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
| | - Hong Liu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Ziwei Dai
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Cheng He
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Shangyao Qin
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China.
| | - Zhida Su
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
22
|
Tabuchi A, Poole DC, Kano Y. Intracellular Ca 2+ After Eccentric Muscle Contractions: Key Role for Ryanodine Receptors. Exerc Sport Sci Rev 2025; 53:23-30. [PMID: 39262047 DOI: 10.1249/jes.0000000000000348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Eccentric contractions (ECC) induce excessive intracellular calcium ion (Ca 2+ ) accumulation and muscle structural damage in localized regions of the muscle fibers. In this investigation, we present the novel hypothesis that the ryanodine receptor (RyR) plays a central role in evoking a Ca 2+ dynamics profile that is markedly distinguishable from other muscle adaptive responses.
Collapse
Affiliation(s)
| | - David C Poole
- Departments of Anatomy and Physiology and Kinesiology, Kansas State University, Manhattan, KS
| | | |
Collapse
|
23
|
Shumanska M, Lodygin D, Gibhardt CS, Ickes C, Stejerean-Todoran I, Krause LCM, Pahl K, Jacobs LJHC, Paluschkiwitz A, Liu S, Boshnakovska A, Voigt N, Legler TJ, Haubrock M, Mitkovski M, Poschmann G, Rehling P, Dennerlein S, Riemer J, Flügel A, Bogeski I. Mitochondrial calcium uniporter complex controls T-cell-mediated immune responses. EMBO Rep 2025; 26:407-442. [PMID: 39623165 PMCID: PMC11772621 DOI: 10.1038/s44319-024-00313-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 01/29/2025] Open
Abstract
T-cell receptor (TCR)-induced Ca2+ signals are essential for T-cell activation and function. In this context, mitochondria play an important role and take up Ca2+ to support elevated bioenergetic demands. However, the functional relevance of the mitochondrial-Ca2+-uniporter (MCU) complex in T-cells was not fully understood. Here, we demonstrate that TCR activation causes rapid mitochondrial Ca2+ (mCa2+) uptake in primary naive and effector human CD4+ T-cells. Compared to naive T-cells, effector T-cells display elevated mCa2+ and increased bioenergetic and metabolic output. Transcriptome and proteome analyses reveal molecular determinants involved in the TCR-induced functional reprogramming and identify signalling pathways and cellular functions regulated by MCU. Knockdown of MCUa (MCUaKD), diminishes mCa2+ uptake, mitochondrial respiration and ATP production, as well as T-cell migration and cytokine secretion. Moreover, MCUaKD in rat CD4+ T-cells suppresses autoimmune responses in an experimental autoimmune encephalomyelitis (EAE) multiple sclerosis model. In summary, we demonstrate that mCa2+ uptake through MCU is essential for proper T-cell function and has a crucial role in autoimmunity. T-cell specific MCU inhibition is thus a potential tool for targeting autoimmune disorders.
Collapse
Affiliation(s)
- Magdalena Shumanska
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Dmitri Lodygin
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Christine S Gibhardt
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Christian Ickes
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Ioana Stejerean-Todoran
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Lena C M Krause
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Kira Pahl
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Lianne J H C Jacobs
- Redox Metabolism, Institute of Biochemistry and CECAD, University of Cologne, Cologne, Germany
| | - Andrea Paluschkiwitz
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Shuya Liu
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Angela Boshnakovska
- Department of Cellular Biochemistry, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Tobias J Legler
- Department of Transfusion Medicine, University Medical Centre, Göttingen, Germany
| | - Martin Haubrock
- Department of Medical Bioinformatics, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Miso Mitkovski
- City Campus Light Microscopy Facility, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Gereon Poschmann
- Institute for Molecular Medicine, Proteome Research, University Hospital and Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Peter Rehling
- Department of Cellular Biochemistry, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Sven Dennerlein
- Department of Cellular Biochemistry, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Jan Riemer
- Redox Metabolism, Institute of Biochemistry and CECAD, University of Cologne, Cologne, Germany
| | - Alexander Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Ivan Bogeski
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany.
| |
Collapse
|
24
|
Arafat Hossain M. A comprehensive review of immune checkpoint inhibitors for cancer treatment. Int Immunopharmacol 2024; 143:113365. [PMID: 39447408 DOI: 10.1016/j.intimp.2024.113365] [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: 08/25/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Immunology-based therapies are emerging as an effective cancer treatment, using the body's immune system to target tumors. Immune checkpoints, which regulate immune responses to prevent tissue damage and autoimmunity, are often exploited by cancer cells to avoid destruction. The discovery of checkpoint proteins like PD-1/PD-L1 and CTLA-4 was pivotal in developing cancer immunotherapy. Immune checkpoint inhibitors (ICIs) have shown great success, with FDA-approved drugs like PD-1 inhibitors (Nivolumab, Pembrolizumab, Cemiplimab), PD-L1 inhibitors (Atezolizumab, Durvalumab, Avelumab), and CTLA-4 inhibitors (Ipilimumab, Tremelimumab), alongside LAG-3 inhibitor Relatlimab. Research continues on new checkpoints like TIM-3, VISTA, B7-H3, BTLA, and TIGIT. Biomarkers like PDL-1 expression, tumor mutation burden, interferon-γ presence, microbiome composition, and extracellular matrix characteristics play a crucial role in predicting responses to immunotherapy with checkpoint inhibitors. Despite their effectiveness, not all patients experience the same level of benefit, and organ-specific immune-related adverse events (irAEs) such as rash or itching, colitis, diarrhea, hyperthyroidism, and hypothyroidism may occur. Given the rapid advancements in this field and the variability in patient outcomes, there is an urgent need for a comprehensive review that consolidates the latest findings on immune checkpoint inhibitors, covering their clinical status, biomarkers, resistance mechanisms, strategies to overcome resistance, and associated adverse effects. This review aims to fill this gap by providing an analysis of the current clinical status of ICIs, emerging biomarkers, mechanisms of resistance, strategies to enhance therapeutic efficacy, and assessment of adverse effects. This review is crucial to furthering our understanding of ICIs and optimizing their application in cancer therapy.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh.
| |
Collapse
|
25
|
Costa FP, Wiedenmann B, Schöll E, Tuszynski J. Emerging cancer therapies: targeting physiological networks and cellular bioelectrical differences with non-thermal systemic electromagnetic fields in the human body - a comprehensive review. FRONTIERS IN NETWORK PHYSIOLOGY 2024; 4:1483401. [PMID: 39720338 PMCID: PMC11666389 DOI: 10.3389/fnetp.2024.1483401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/22/2024] [Indexed: 12/26/2024]
Abstract
A steadily increasing number of publications support the concept of physiological networks, and how cellular bioelectrical properties drive cell proliferation and cell synchronization. All cells, especially cancer cells, are known to possess characteristic electrical properties critical for physiological behavior, with major differences between normal and cancer cell counterparts. This opportunity can be explored as a novel treatment modality in Oncology. Cancer cells exhibit autonomous oscillations, deviating from normal rhythms. In this context, a shift from a static view of cellular processes is required for a better understanding of the dynamic connections between cellular metabolism, gene expression, cell signaling and membrane polarization as states in constant flux in realistic human models. In oncology, radiofrequency electromagnetic fields have produced sustained responses and improved quality of life in cancer patients with minimal side effects. This review aims to show how non-thermal systemic radiofrequency electromagnetic fields leads to promising therapeutic responses at cellular and tissue levels in humans, supporting this newly emerging cancer treatment modality with early favorable clinical experience specifically in advanced cancer.
Collapse
Affiliation(s)
| | | | - Eckehard Schöll
- Institut für Theoretische Physik, Technische Universität Berlin, Berlin, Germany
| | - Jack Tuszynski
- Department of Physics, University of Alberta, Edmonton, AB, Canada
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Turin, Italy
- Department of Data Science and Engineering, The Silesian University of Technology, Gliwice, Poland
| |
Collapse
|
26
|
Young M, Booth DM, Smith D, Tigano M, Hajnόczky G, Joseph SK. Transcriptional regulation in the absence of inositol trisphosphate receptor calcium signaling. Front Cell Dev Biol 2024; 12:1473210. [PMID: 39712573 PMCID: PMC11659226 DOI: 10.3389/fcell.2024.1473210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024] Open
Abstract
The activation of IP3 receptor (IP3R) Ca2+ channels generates agonist-mediated Ca2+ signals that are critical for the regulation of a wide range of biological processes. It is therefore surprising that CRISPR induced loss of all three IP3R isoforms (TKO) in HEK293 and HeLa cell lines yields cells that can survive, grow and divide, albeit more slowly than wild-type cells. In an effort to understand the adaptive mechanisms involved, we have examined the activity of key Ca2+ dependent transcription factors (NFAT, CREB and AP-1) and signaling pathways using luciferase-reporter assays, phosphoprotein immunoblots and whole genome transcriptomic studies. In addition, the diacylglycerol arm of the signaling pathway was investigated with protein kinase C (PKC) inhibitors and siRNA knockdown. The data showed that agonist-mediated NFAT activation was lost but CREB activation was maintained in IP3R TKO cells. Under base-line conditions transcriptome analysis indicated the differential expression of 828 and 311 genes in IP3R TKO HEK293 or HeLa cells, respectively, with only 18 genes being in common. Three main adaptations in TKO cells were identified in this study: 1) increased basal activity of NFAT, CREB and AP-1; 2) an increased reliance on Ca2+- insensitive PKC isoforms; and 3) increased production of reactive oxygen species and upregulation of antioxidant defense enzymes. We suggest that whereas wild-type cells rely on a Ca2+ and DAG signal to respond to stimuli, the TKO cells utilize the adaptations to allow key signaling pathways (e.g., PKC, Ras/MAPK, CREB) to transition to the activated state using a DAG signal alone.
Collapse
Affiliation(s)
- Michael Young
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - David M. Booth
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - David Smith
- Center for Single Cell Biology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Marco Tigano
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Gyӧrgy Hajnόczky
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Suresh K. Joseph
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
27
|
Ji Y, Chen C, Lu P, Wang Z, Chen H, Sun L, Fei S, Ju X, Tan R, Gu M. Nuclear factor of activated T cell cytoplasmic 1 (NFATc1) insertion gene polymorphism as a possible trigger in acute T cell-mediated rejection (aTCMR) after kidney transplantation. Transpl Immunol 2024; 87:102139. [PMID: 39461381 DOI: 10.1016/j.trim.2024.102139] [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: 07/04/2024] [Revised: 10/04/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND To investigate the potential regulatory role of gene insertion or deletion (in/del) polymorphism in the occurrence of acute T cell-mediated rejection (aTCMR) after kidney transplantation. METHODS We retrospectively analyzed the 5-year follow-up data of 133 recipients who underwent renal transplantation at the First Affiliated Hospital of Nanjing Medical University between February 1, 2010, and December 1, 2015. With target sequencing based on next-generation sequencing (NGS), tagger in/dels selection involved calculating the Hardy-Weinberg equilibrium (HWE), Minor Allele Frequency (MAF), and the linkage disequilibrium (LD) blocks. Significant in/dels associated with aTCMR were identified by intersecting the results obtained through analysis of covariance (ANCOVA) of clinical cofounders and model analysis in Rstudio using the "SNPassoc" package. Additionally, logistic models were employed to assess the associations between genotypes and the aTCMR occurrence in 5 years after surgery. RESULTS NFATc1 rs55741427 insertion was identified to be significantly associated with the post-surgery aTCMR(OR = 2.66, P < 0.001). We constructed a conclusive model containing the occurrence of delayed graft function (DGF) and the insertion polymorphism of rs55741427, showing a favorable predictive ability (AUC = 0.766) for aTCMR after surgery. Based on the receiver operating characteristic (ROC) curve, all cases were stratified into aTCMR high-risk and low-risk groups. Kaplan-Meier curves for two groups revealed that the aTCMR high-risk group exhibited a more unfavorable graft survival outcome (P = 0.0048). CONCLUSION Insertion mutation of rs55741427 was found to be statistically correlated with the post-surgery aTCMR during 5 years of follow-up. Our model identified DGF and insertion of rs55741427 as two crucial aTCMR-related hazards, and aTCMR high-risk group showed a worse graft prognosis.
Collapse
Affiliation(s)
- Yisheng Ji
- Deparment of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China; The First Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Congcong Chen
- Deparment of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China; The First Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Pei Lu
- Deparment of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zijie Wang
- Deparment of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Chen
- Deparment of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Li Sun
- Deparment of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shuang Fei
- Deparment of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaobing Ju
- Deparment of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Ruoyun Tan
- Deparment of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Min Gu
- Deparment of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of Urology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
28
|
Lawler NB, Bhatt U, Agarwal V, Evans CW, Kaluskar P, Amos SE, Chen K, Yao Y, Jiang H, Choi YS, Zheng M, Spagnoli D, Suarez‐Martinez I, Zetterlund PB, Wallace VP, Harvey AR, Hodgetts SI, Iyer KS. Transcriptomic Analysis Reveals the Heterogeneous Role of Conducting Films Upon Electrical Stimulation. Adv Healthc Mater 2024; 13:e2400364. [PMID: 39221662 PMCID: PMC11670273 DOI: 10.1002/adhm.202400364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/17/2024] [Indexed: 09/04/2024]
Abstract
Central nervous system (CNS) injuries and neurodegenerative diseases have markedly poor prognoses and can result in permanent dysfunction due to the general inability of CNS neurons to regenerate. Differentiation of transplanted stem cells has emerged as a therapeutic avenue to regenerate tissue architecture in damaged areas. Electrical stimulation is a promising approach for directing the differentiation outcomes and pattern of outgrowth of transplanted stem cells, however traditional inorganic bio-electrodes can induce adverse effects such as inflammation. This study demonstrates the implementation of two organic thin films, a polymer/reduced graphene oxide nanocomposite (P(rGO)) and PEDOT:PSS, that have favorable properties for implementation as conductive materials for electrical stimulation, as well as an inorganic indium tin oxide (ITO) conductive film. Transcriptomic analysis reveals that electrical stimulation improves neuronal differentiation of SH-SY5Y cells on all three films, with the greatest effect for P(rGO). Unique material- and electrical stimuli-mediated effects are observed, associated with differentiation, cell-substrate adhesion, and translation. The work demonstrates that P(rGO) and PEDOT:PSS are highly promising organic materials for the development of biocompatible, conductive scaffolds that will enhance electrically-aided stem cell therapeutics for CNS injuries and neurodegenerative diseases.
Collapse
Affiliation(s)
- Nicholas B. Lawler
- School of Molecular SciencesThe University of Western AustraliaPerthWA6009Australia
- School of PhysicsMathematics and ComputingThe University of Western AustraliaPerthWA6009Australia
- ARC Training Centre for Next‐Gen Technologies in Biomedical AnalysisSchool of Molecular SciencesThe University of Western AustraliaPerthWA6009Australia
| | - Uditi Bhatt
- School of Molecular SciencesThe University of Western AustraliaPerthWA6009Australia
- ARC Training Centre for Next‐Gen Technologies in Biomedical AnalysisSchool of Molecular SciencesThe University of Western AustraliaPerthWA6009Australia
| | - Vipul Agarwal
- Cluster for Advanced Macromolecular Design (CAMD)School of Chemical EngineeringUniversity of New South WalesSydneyNSW2052Australia
| | - Cameron W. Evans
- School of Molecular SciencesThe University of Western AustraliaPerthWA6009Australia
- ARC Training Centre for Next‐Gen Technologies in Biomedical AnalysisSchool of Molecular SciencesThe University of Western AustraliaPerthWA6009Australia
| | - Priya Kaluskar
- School of Molecular SciencesThe University of Western AustraliaPerthWA6009Australia
- ARC Training Centre for Next‐Gen Technologies in Biomedical AnalysisSchool of Molecular SciencesThe University of Western AustraliaPerthWA6009Australia
- Perron Institute for Neurological and Translational SciencePerthWA6009Australia
- Centre for Orthopaedic ResearchThe UWA Medical SchoolThe University of Western AustraliaPerthWA6009Australia
- Australian Research Council Centre for Personalised Therapeutics TechnologiesUniversity of MelbourneMelbourneVictoria3010Australia
| | - Sebastian E. Amos
- School of Human SciencesThe University of Western AustraliaPerthWA6009Australia
| | - Kai Chen
- School of Molecular SciencesThe University of Western AustraliaPerthWA6009Australia
- ARC Training Centre for Next‐Gen Technologies in Biomedical AnalysisSchool of Molecular SciencesThe University of Western AustraliaPerthWA6009Australia
- Department of ChemistryThe University of Hong KongHong KongChina
| | - Yin Yao
- Electron Microscope UnitMark Wainwright Analytical CentreUniversity of New South WalesSydneyNSW2052Australia
| | - Haibo Jiang
- School of Molecular SciencesThe University of Western AustraliaPerthWA6009Australia
- ARC Training Centre for Next‐Gen Technologies in Biomedical AnalysisSchool of Molecular SciencesThe University of Western AustraliaPerthWA6009Australia
- Department of ChemistryThe University of Hong KongHong KongChina
| | - Yu Suk Choi
- ARC Training Centre for Next‐Gen Technologies in Biomedical AnalysisSchool of Molecular SciencesThe University of Western AustraliaPerthWA6009Australia
- School of Human SciencesThe University of Western AustraliaPerthWA6009Australia
| | - Minghao Zheng
- Perron Institute for Neurological and Translational SciencePerthWA6009Australia
- Centre for Orthopaedic ResearchThe UWA Medical SchoolThe University of Western AustraliaPerthWA6009Australia
| | - Dino Spagnoli
- School of Molecular SciencesThe University of Western AustraliaPerthWA6009Australia
| | | | - Per B. Zetterlund
- Cluster for Advanced Macromolecular Design (CAMD)School of Chemical EngineeringUniversity of New South WalesSydneyNSW2052Australia
| | - Vincent P. Wallace
- School of PhysicsMathematics and ComputingThe University of Western AustraliaPerthWA6009Australia
| | - Alan R. Harvey
- Perron Institute for Neurological and Translational SciencePerthWA6009Australia
- School of Human SciencesThe University of Western AustraliaPerthWA6009Australia
| | - Stuart I. Hodgetts
- Perron Institute for Neurological and Translational SciencePerthWA6009Australia
- School of Human SciencesThe University of Western AustraliaPerthWA6009Australia
| | - K. Swaminathan Iyer
- School of Molecular SciencesThe University of Western AustraliaPerthWA6009Australia
- ARC Training Centre for Next‐Gen Technologies in Biomedical AnalysisSchool of Molecular SciencesThe University of Western AustraliaPerthWA6009Australia
| |
Collapse
|
29
|
Dome PA, Jeong P, Nam G, Jang H, Rivera A, Averette AF, Park E, Liao TC, Ciofani M, Wu J, Chi JTA, Venters RA, Park HJ, Steinbach WJ, Juvvadi PR, Heitman J, Hong J. Structure-guided design and synthesis of C22- and C32-modified FK520 analogs with enhanced activity against human pathogenic fungi. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.27.615491. [PMID: 39386510 PMCID: PMC11463406 DOI: 10.1101/2024.09.27.615491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Invasive fungal infections are a leading cause of death worldwide. Translating molecular insights into clinical benefits is challenging because fungal pathogens and their hosts share similar eukaryotic physiology. Consequently, current antifungal treatments have limited efficacy, may be poorly fungicidal in the host, can exhibit toxicity, and are increasingly compromised by emerging resistance. We have established that the phosphatase calcineurin (CaN) is required for invasive fungal disease and an attractive target for antifungal drug development. CaN is a druggable target, and there is vast clinical experience with the CaN inhibitors FK506 and cyclosporin A (CsA). However, while FK506 and its natural analog FK520 exhibit antifungal activity, they are also immunosuppressive in the host and thus not fungal-selective. We leverage our pathogenic fungal CaN-FK506-FKBP12 complex X-ray structures and biophysical data to support structure-based ligand design as well as structure-activity relationship analyses of broad-spectrum FK506/FK520 derivatives with potent antifungal activity and reduced immunosuppressive activity. Here we apply molecular docking studies to develop antifungal C22- or C32-modified FK520 derivatives with improved therapeutic index scores. Among them, the C32-modified FK520 derivative JH-FK-44 ( 7 ) demonstrates a significantly improved therapeutic index compared to JH-FK-08, our lead compound to date. NMR binding studies with C32-derivatives are consistent with our hypothesis that C32 modifications disrupt the hydrogen bonding network in the human complex while introducing favorable electrostatic and cation-π interactions with the fungal FKBP12 R86 residue. These findings further reinforce calcineurin inhibition as a promising strategy for antifungal therapy. Significance Invasive fungal infections cause significant mortality worldwide, and current antifungal treatments are often ineffective, toxic, or face growing resistance. This research identifies calcineurin (CaN), a critical protein for fungal survival, as a potential target for developing new antifungal drugs. Although existing CaN inhibitors such as FK506 (tacrolimus) and FK520 (ascomycin) possess antifungal properties, their immunosuppressive effects limit their clinical utility. By studying the structure of human and fungal FKBP12-FK506 or FK520 complexes with CaN, we have designed and synthesized modified FK520 derivatives with strong antifungal activity and reduced immunosuppressive effects. These new derivatives are expected to have significantly improved therapeutic profiles, offering hope for more effective and safer antifungal treatments.
Collapse
|
30
|
Chimura T, Manabe T. Ca2+-PP2B-PSD-95 axis: A novel regulatory mechanism of the phosphorylation state of Serine 295 of PSD-95. PLoS One 2024; 19:e0313441. [PMID: 39509447 PMCID: PMC11542788 DOI: 10.1371/journal.pone.0313441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024] Open
Abstract
The phosphorylation state of PSD-95 at Serine 295 (Ser295) is important for the regulation of synaptic plasticity. Although the activation of NMDA receptors (NMDARs), which initiates an intracellular calcium signaling cascade, decreases phosphorylated Ser295 (pS295) of PSD-95, the molecular mechanisms are not fully understood. We found that the calcium-activated protein phosphatase PP2B dephosphorylated pS295 not only in basal conditions but also in NMDAR-activated conditions in cultured neurons. The biochemical assay also revealed the dephosphorylation of pS295 by PP2B, consistently supporting the results obtained using neurons. The newly identified calcium signaling cascade "Ca2+-PP2B-PSD-95 axis" would play an important role in the molecular mechanism for NMDA receptor-dependent plasticity.
Collapse
Affiliation(s)
- Takahiko Chimura
- Department of Basic Medical Sciences, Institute of Medical Science, Division of Neuronal Network, University of Tokyo, Tokyo, Japan
| | - Toshiya Manabe
- Department of Basic Medical Sciences, Institute of Medical Science, Division of Neuronal Network, University of Tokyo, Tokyo, Japan
| |
Collapse
|
31
|
Mackiewicz J, Tomczak J, Lisek M, Sakowicz A, Guo F, Boczek T. NFATc4 Knockout Promotes Neuroprotection and Retinal Ganglion Cell Regeneration After Optic Nerve Injury. Mol Neurobiol 2024; 61:9383-9401. [PMID: 38639863 PMCID: PMC11496353 DOI: 10.1007/s12035-024-04129-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/19/2024] [Indexed: 04/20/2024]
Abstract
Retinal ganglion cells (RGCs), neurons transmitting visual information via the optic nerve, fail to regenerate their axons after injury. The progressive loss of RGC function underlies the pathophysiology of glaucoma and other optic neuropathies, often leading to irreversible blindness. Therefore, there is an urgent need to identify the regulators of RGC survival and the regenerative program. In this study, we investigated the role of the family of transcription factors known as nuclear factor of activated T cells (NFAT), which are expressed in the retina; however, their role in RGC survival after injury is unknown. Using the optic nerve crush (ONC) model, widely employed to study optic neuropathies and central nervous system axon injury, we found that NFATc4 is specifically but transiently up-regulated in response to mechanical injury. In the injured retina, NFATc4 immunolocalized primarily to the ganglionic cell layer. Utilizing NFATc4-/- and NFATc3-/- mice, we demonstrated that NFATc4, but not NFATc3, knockout increased RGC survival, improved retina function, and delayed axonal degeneration. Microarray screening data, along with decreased immunostaining of cleaved caspase-3, revealed that NFATc4 knockout was protective against ONC-induced degeneration by suppressing pro-apoptotic signaling. Finally, we used lentiviral-mediated NFATc4 delivery to the retina of NFATc4-/- mice and reversed the pro-survival effect of NFATc4 knockout, conclusively linking the enhanced survival of injured RGCs to NFATc4-dependent mechanisms. In summary, this study is the first to demonstrate that NFATc4 knockout may confer transient RGC neuroprotection and decelerate axonal degeneration after injury, providing a potent therapeutic strategy for optic neuropathies.
Collapse
Affiliation(s)
- Joanna Mackiewicz
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland
| | - Julia Tomczak
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland
| | - Malwina Lisek
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland
| | - Agata Sakowicz
- Department of Medical Biotechnology, Medical University of Lodz, Lodz, Poland
| | - Feng Guo
- Department of Pharmaceutical Toxicology, China Medical University, Shenyang, China.
| | - Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
32
|
Parekh AB. House dust mite allergens, store-operated Ca 2+ channels and asthma. J Physiol 2024; 602:6021-6038. [PMID: 38054814 DOI: 10.1113/jp284931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/26/2023] [Indexed: 12/07/2023] Open
Abstract
The house dust mite is the principal source of aero-allergen worldwide. Exposure to mite-derived allergens is associated with the development of asthma in susceptible individuals, and the majority of asthmatics are allergic to the mite. Mite-derived allergens are functionally diverse and activate multiple cell types within the lung that result in chronic inflammation. Allergens activate store-operated Ca2+ release-activated Ca2+ (CRAC) channels, which are widely expressed in multiple cell types within the lung that are associated with the pathogenesis of asthma. Opening of CRAC channels stimulates Ca2+-dependent transcription factors, including nuclear factor of activated T cells and nuclear factor-κB, which drive expression of a plethora of pro-inflammatory cytokines and chemokines that help to sustain chronic inflammation. Here, I describe drivers of asthma, properties of mite-derived allergens, how the allergens are recognized by cells, the signalling pathways used by the receptors and how these are transduced into functional effects, with a focus on CRAC channels. In vivo experiments that demonstrate the effectiveness of targeting CRAC channels as a potential new therapy for treating mite-induced asthma are also discussed, in tandem with other possible approaches.
Collapse
Affiliation(s)
- Anant B Parekh
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, US National Institutes of Health, Department of Health and Human Services, Research Triangle Park, Durham, NC, USA
| |
Collapse
|
33
|
Das P, Aballay A, Singh J. Calcineurin inhibition enhances Caenorhabditis elegans lifespan by defecation defects-mediated calorie restriction and nuclear hormone signaling. eLife 2024; 12:RP89572. [PMID: 39485281 PMCID: PMC11530235 DOI: 10.7554/elife.89572] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024] Open
Abstract
Calcineurin is a highly conserved calcium/calmodulin-dependent serine/threonine protein phosphatase with diverse functions. Inhibition of calcineurin is known to enhance the lifespan of Caenorhabditis elegans through multiple signaling pathways. Aiming to study the role of calcineurin in regulating innate immunity, we discover that calcineurin is required for the rhythmic defecation motor program (DMP) in C. elegans. Calcineurin inhibition leads to defects in the DMP, resulting in intestinal bloating, rapid colonization of the gut by bacteria, and increased susceptibility to bacterial infection. We demonstrate that intestinal bloating caused by calcineurin inhibition mimics the effects of calorie restriction, resulting in enhanced lifespan. The TFEB ortholog, HLH-30, is required for lifespan extension mediated by calcineurin inhibition. Finally, we show that the nuclear hormone receptor, NHR-8, is upregulated by calcineurin inhibition and is necessary for the increased lifespan. Our studies uncover a role for calcineurin in the C. elegans DMP and provide a new mechanism for calcineurin inhibition-mediated longevity extension.
Collapse
Affiliation(s)
- Priyanka Das
- Department of Biological Sciences, Indian Institute of Science Education and ResearchMohaliIndia
| | - Alejandro Aballay
- Department of Genetics, The University of Texas MD Anderson Cancer CenterHoustonUnited States
| | - Jogender Singh
- Department of Biological Sciences, Indian Institute of Science Education and ResearchMohaliIndia
| |
Collapse
|
34
|
Flynn MJ, Harper NW, Li R, Zhu LJ, Lee MJ, Benanti JA. Calcineurin promotes adaptation to chronic stress through two distinct mechanisms. Mol Biol Cell 2024; 35:ar123. [PMID: 39083354 PMCID: PMC11481702 DOI: 10.1091/mbc.e24-03-0122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024] Open
Abstract
Adaptation to environmental stress requires coordination between stress-defense programs and cell cycle progression. The immediate response to many stressors has been well characterized, but how cells survive in challenging environments long term is unknown. Here, we investigate the role of the stress-activated phosphatase calcineurin (CN) in adaptation to chronic CaCl2 stress in Saccharomyces cerevisiae. We find that prolonged exposure to CaCl2 impairs mitochondrial function and demonstrate that cells respond to this stressor using two CN-dependent mechanisms-one that requires the downstream transcription factor Crz1 and another that is Crz1 independent. Our data indicate that CN maintains cellular fitness by promoting cell cycle progression and preventing CaCl2-induced cell death. When Crz1 is present, transient CN activation suppresses cell death and promotes adaptation despite high levels of mitochondrial loss. However, in the absence of Crz1, prolonged activation of CN prevents mitochondrial loss and further cell death by upregulating glutathione biosynthesis genes thereby mitigating damage from reactive oxygen species. These findings illustrate how cells maintain long-term fitness during chronic stress and suggest that CN promotes adaptation in challenging environments by multiple mechanisms.
Collapse
Affiliation(s)
- Mackenzie J. Flynn
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605
- Interdisciplinary Graduate Program, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01605
| | - Nicholas W. Harper
- Interdisciplinary Graduate Program, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01605
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605
| | - Rui Li
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605
| | - Lihua Julie Zhu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605
| | - Michael J. Lee
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605
| | - Jennifer A. Benanti
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605
| |
Collapse
|
35
|
Faiz M, Kalev‐Zylinska ML, Dunstan‐Harrison C, Singleton DC, Hay MP, Ledgerwood EC. Megakaryocyte maturation involves activation of the adaptive unfolded protein response. Genes Cells 2024; 29:889-901. [PMID: 39138929 PMCID: PMC11555628 DOI: 10.1111/gtc.13151] [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: 02/12/2024] [Revised: 07/21/2024] [Accepted: 07/26/2024] [Indexed: 08/15/2024]
Abstract
Endoplasmic reticulum stress triggers the unfolded protein response (UPR) to promote cell survival or apoptosis. Transient endoplasmic reticulum stress activation has been reported to trigger megakaryocyte production, and UPR activation has been reported as a feature of megakaryocytic cancers. However, the role of UPR signaling in megakaryocyte biology is not fully understood. We studied the involvement of UPR in human megakaryocytic differentiation using PMA (phorbol 12-myristate 13-acetate)-induced maturation of megakaryoblastic cell lines and thrombopoietin-induced differentiation of human peripheral blood-derived progenitors. Our results demonstrate that an adaptive UPR is a feature of megakaryocytic differentiation and that this response is not associated with ER stress-induced apoptosis. Differentiation did not alter the response to the canonical endoplasmic reticulum stressors DTT or thapsigargin. However, thapsigargin, but not DTT, inhibited differentiation, consistent with the involvement of Ca2+ signaling in megakaryocyte differentiation.
Collapse
Affiliation(s)
- Mifra Faiz
- Department of BiochemistrySchool of Biomedical Sciences, University of OtagoDunedinNew Zealand
| | - Maggie L. Kalev‐Zylinska
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine & PathologyFaculty of Medical and Health Sciences, The University of AucklandAucklandNew Zealand
| | | | - Dean C. Singleton
- Auckland Cancer Society Research CentreFaculty of Medical and Health Sciences, The University of AucklandAucklandNew Zealand
| | - Michael P. Hay
- Auckland Cancer Society Research CentreFaculty of Medical and Health Sciences, The University of AucklandAucklandNew Zealand
| | - Elizabeth C. Ledgerwood
- Department of BiochemistrySchool of Biomedical Sciences, University of OtagoDunedinNew Zealand
| |
Collapse
|
36
|
Rodriguez-Rodriguez C, González-Mancha N, Ochoa-Echeverría A, Mérida I. Sorting nexin 27-dependent regulation of Lck and CD4 tunes the initial stages of T-cell activation. J Leukoc Biol 2024; 116:793-806. [PMID: 38648515 DOI: 10.1093/jleuko/qiae086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/27/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
Sorting nexin 27 is a unique member of the sorting nexin family of proteins that mediates the endosome-to-plasma membrane trafficking of cargos bearing a PSD95/Dlg1/ZO-1 (PDZ)-binding motif. In brain, sorting nexin 27 regulates synaptic plasticity, and its dysregulation contributes to cognitive impairment and neuronal degeneration. In T lymphocytes, sorting nexin 27 partners with diacylglycerol kinase ζ to facilitate polarized traffic and signaling at the immune synapse. By silencing sorting nexin 27 expression in a human T-cell line, we demonstrate that sorting nexin 27 is a key regulator of the early T-cell tyrosine-based signaling cascade. Sorting nexin 27 transcriptionally controls CD4 abundance in resting conditions and that of its associated molecule, Lck. This guarantees the adequate recruitment of Lck at the immune synapse, which is indispensable for subsequent activation of tyrosine phosphorylation-regulated events. In contrast, reduced sorting nexin 27 expression enhances NF-κB-dependent induction of CXCR4 and triggers production of lytic enzymes and proinflammatory cytokines. These results provide mechanistic explanation to previously described sorting nexin 27 function in the control of immune synapse organization and indicate that impaired sorting nexin 27 expression contributes to CD4 T-cell dysfunction.
Collapse
Affiliation(s)
- Cristina Rodriguez-Rodriguez
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology (CNB-CSIC), UAM Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| | - Natalia González-Mancha
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology (CNB-CSIC), UAM Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| | - Ane Ochoa-Echeverría
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology (CNB-CSIC), UAM Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| | - Isabel Mérida
- Department of Immunology and Oncology, Spanish National Centre for Biotechnology (CNB-CSIC), UAM Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| |
Collapse
|
37
|
Srinivasan S, Armitage J, Nilsson J, Waithman J. Transcriptional rewiring in CD8 + T cells: implications for CAR-T cell therapy against solid tumours. Front Immunol 2024; 15:1412731. [PMID: 39399500 PMCID: PMC11466849 DOI: 10.3389/fimmu.2024.1412731] [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: 04/05/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024] Open
Abstract
T cells engineered to express chimeric-antigen receptors (CAR-T cells) can effectively control relapsed and refractory haematological malignancies in the clinic. However, the successes of CAR-T cell therapy have not been recapitulated in solid tumours due to a range of barriers such as immunosuppression, poor infiltration, and tumour heterogeneity. Numerous strategies are being developed to overcome these barriers, which include improving culture conditions and manufacturing protocols, implementing novel CAR designs, and novel approaches to engineering the T cell phenotype. In this review, we describe the various emerging strategies to improve CAR T cell therapy for solid tumours. We specifically focus on new strategies to modulate cell function and fate that have precipitated from the growing knowledge of transcriptional circuits driving T cell differentiation, with the ultimate goal of driving more productive anti-tumour T cell immunity. Evidence shows that enrichment of particular phenotypic subsets of T cells in the initial cell product correlates to improved therapeutic responses and clinical outcomes. Furthermore, T cell exhaustion and poor persistence are major factors limiting therapeutic efficacy. The latest preclinical work shows that targeting specific master regulators and transcription factors can overcome these key barriers, resulting in superior T cell therapeutic products. This can be achieved by targeting key transcriptional circuits promoting memory-like phenotypes or sustaining key effector functions within the hostile tumour microenvironment. Additional discussion points include emerging considerations for the field such as (i) targeting permutations of transcription factors, (ii) transient expression systems, (iii) tissue specificity, and (iv) expanding this strategy beyond CAR-T cell therapy and cancer.
Collapse
Affiliation(s)
- Shamini Srinivasan
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Jesse Armitage
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
- Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Jonas Nilsson
- Melanoma Discovery Lab, Harry Perkins Institute of Medical Research, Centre of Medical Research, The University of Western Australia, Perth, WA, Australia
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Jason Waithman
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
- Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| |
Collapse
|
38
|
Falah G, Kurolap A, Paperna T, Ekhilevitch N, Moustafa N, Damouny-Naoum N, Amir Y, Sharvit L, Moghrabi R, Hassoun G, Fares F, Baris Feldman H, Atzmon G. The d3GHR carrier epigenome in Druze clan longevity. Sci Rep 2024; 14:21419. [PMID: 39271799 PMCID: PMC11399368 DOI: 10.1038/s41598-024-72240-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
The Druze are a distinct group known for their close community, traditions, and consanguineous marriages, dating back to the eleventh century. This practice has led to unique genetic variations, impacting both pathology and gene-associated phenotypes. Some Druze clans, particularly those with exceptional long-lived family heads (ELLI), attracted attention. Given that the bulk of these ELLI were men, the d3GHR polymorphism was the first obvious possibility. Among the 73 clan members, 8.2% carried the d3GHR isoform, with nearly 11% being males. There was a significant age-related increase (p = 0.04) in this isoform among males, leading to examination of potential environmental mediators affecting gene regulation among these carriers during life (namely epigenetic). We focused on DNA methylation due to its crucial role in gene regulation, development, and disease progression. We analyzed DNA samples from 14 clan members with different GHR genotypes, finding a significant (p < 0.05) negative correlation between DNA methylation levels and age. Employing a biological age clock, we observed a significant + 4.229 years favoring the d3GHR group over the WT and heterozygous groups. In conclusion, this study highlights the advantage of d3GHR carriers among this unique Druze clan and underscores the importance of genotype-environment interaction in epigenetic regulation and its impact on health.
Collapse
Affiliation(s)
- Ghadeer Falah
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Alina Kurolap
- The Genetics Institute and Genomics Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Tamar Paperna
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
| | - Nina Ekhilevitch
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
| | - Nivin Moustafa
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
| | | | - Yam Amir
- The Genetics Institute and Genomics Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Lital Sharvit
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Rihan Moghrabi
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
| | - Gamal Hassoun
- Institute of Allergy, Clinical Immunology & AIDS, Rambam Health Care Campus, Haifa, Israel
| | - Fuad Fares
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Hagit Baris Feldman
- The Genetics Institute and Genomics Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Gil Atzmon
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel.
- Departments of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
39
|
Prikhodko O, Freund RK, Sullivan E, Kennedy MJ, Dell'Acqua ML. Amyloid-β Causes NMDA Receptor Dysfunction and Dendritic Spine Loss through mGluR1 and AKAP150-Anchored Calcineurin Signaling. J Neurosci 2024; 44:e0675242024. [PMID: 39134419 PMCID: PMC11391497 DOI: 10.1523/jneurosci.0675-24.2024] [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: 04/10/2024] [Revised: 07/08/2024] [Accepted: 07/30/2024] [Indexed: 09/13/2024] Open
Abstract
Neuronal excitatory synapses are primarily located on small dendritic protrusions called spines. During synaptic plasticity underlying learning and memory, Ca2+ influx through postsynaptic NMDA-type glutamate receptors (NMDARs) initiates signaling pathways that coordinate changes in dendritic spine structure and synaptic function. During long-term potentiation (LTP), high levels of NMDAR Ca2+ influx promote increases in both synaptic strength and dendritic spine size through activation of Ca2+-dependent protein kinases. In contrast, during long-term depression (LTD), low levels of NMDAR Ca2+ influx promote decreased synaptic strength and spine shrinkage and elimination through activation of the Ca2+-dependent protein phosphatase calcineurin (CaN), which is anchored at synapses via the scaffold protein A-kinase anchoring protein (AKAP)150. In Alzheimer's disease (AD), the pathological agent amyloid-β (Aβ) may impair learning and memory through biasing NMDAR Ca2+ signaling pathways toward LTD and spine elimination. By employing AKAP150 knock-in mice of both sexes with a mutation that disrupts CaN anchoring to AKAP150, we revealed that local, postsynaptic AKAP-CaN-LTD signaling was required for Aβ-mediated impairment of NMDAR synaptic Ca2+ influx, inhibition of LTP, and dendritic spine loss. Additionally, we found that Aβ acutely engages AKAP-CaN signaling through activation of G-protein-coupled metabotropic glutamate receptor 1 (mGluR1) leading to dephosphorylation of NMDAR GluN2B subunits, which decreases Ca2+ influx to favor LTD over LTP, and cofilin, which promotes F-actin severing to destabilize dendritic spines. These findings reveal a novel interplay between NMDAR and mGluR1 signaling that converges on AKAP-anchored CaN to coordinate dephosphorylation of postsynaptic substrates linked to multiple aspects of Aβ-mediated synaptic dysfunction.
Collapse
Affiliation(s)
- Olga Prikhodko
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Ronald K Freund
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Alzheimer's and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Emily Sullivan
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Matthew J Kennedy
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Neurotechnology Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Neurotechnology Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Alzheimer's and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| |
Collapse
|
40
|
He M, Zong X, Xu B, Qi W, Huang W, Djekidel MN, Zhang Y, Pagala VR, Li J, Hao X, Guy C, Bai L, Cross R, Li C, Peng J, Feng Y. Dynamic Foxp3-chromatin interaction controls tunable Treg cell function. J Exp Med 2024; 221:e20232068. [PMID: 38935023 PMCID: PMC11211070 DOI: 10.1084/jem.20232068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 04/11/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Nuclear factor Foxp3 determines regulatory T (Treg) cell fate and function via mechanisms that remain unclear. Here, we investigate the nature of Foxp3-mediated gene regulation in suppressing autoimmunity and antitumor immune response. Contrasting with previous models, we find that Foxp3-chromatin binding is regulated by Treg activation states, tumor microenvironment, and antigen and cytokine stimulations. Proteomics studies uncover dynamic proteins within Foxp3 proximity upon TCR or IL-2 receptor signaling in vitro, reflecting intricate interactions among Foxp3, signal transducers, and chromatin. Pharmacological inhibition and genetic knockdown experiments indicate that NFAT and AP-1 protein Batf are required for enhanced Foxp3-chromatin binding in activated Treg cells and tumor-infiltrating Treg cells to modulate target gene expression. Furthermore, mutations at the Foxp3 DNA-binding domain destabilize the Foxp3-chromatin association. These representative settings delineate context-dependent Foxp3-chromatin interaction, suggesting that Foxp3 associates with chromatin by hijacking DNA-binding proteins resulting from Treg activation or differentiation, which is stabilized by direct Foxp3-DNA binding, to dynamically regulate Treg cell function according to immunological contexts.
Collapse
Affiliation(s)
- Minghong He
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Xinying Zong
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Beisi Xu
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Wenjie Qi
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Wenjun Huang
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | | | - Yang Zhang
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Vishwajeeth R. Pagala
- Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Jun Li
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Xiaolei Hao
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Clifford Guy
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Lu Bai
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Richard Cross
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Chunliang Li
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Junmin Peng
- Department of Structure Biology and Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Yongqiang Feng
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
41
|
Ivanovski F, Meško M, Lebar T, Rupnik M, Lainšček D, Gradišek M, Jerala R, Benčina M. Ultrasound-mediated spatial and temporal control of engineered cells in vivo. Nat Commun 2024; 15:7369. [PMID: 39191796 DOI: 10.1038/s41467-024-51620-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Remote regulation of cells in deep tissue remains a significant challenge. Low-intensity pulsed ultrasound offers promise for in vivo therapies due to its non-invasive nature and precise control. This study uses pulsed ultrasound to control calcium influx in mammalian cells and engineers a therapeutic cellular device responsive to acoustic stimulation in deep tissue without overexpressing calcium channels or gas vesicles. Pulsed ultrasound parameters are established to induce calcium influx in HEK293 cells. Additionally, cells are engineered to express a designed calcium-responsive transcription factor controlling the expression of a selected therapeutic gene, constituting a therapeutic cellular device. The engineered sonogenetic system's functionality is demonstrated in vivo in mice, where an implanted anti-inflammatory cytokine-producing cellular device effectively alleviates acute colitis, as shown by improved colonic morphology and histopathology. This approach provides a powerful tool for precise, localized control of engineered cells in deep tissue, showcasing its potential for targeted therapeutic delivery.
Collapse
Affiliation(s)
- Filip Ivanovski
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, Slovenia
- Interfaculty Doctoral Study of Biomedicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia
| | - Maja Meško
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, Slovenia
| | - Tina Lebar
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, Slovenia
| | - Marko Rupnik
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, Slovenia
| | - Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, Slovenia
| | - Miha Gradišek
- Faculty of Electrical Engineering, University of Ljubljana, Tržaška c. 25, Ljubljana, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, Slovenia.
- CTGCT, Centre of Technology of Gene and Cell Therapy, Hajdrihova 19, Ljubljana, Slovenia.
| | - Mojca Benčina
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, Slovenia.
- CTGCT, Centre of Technology of Gene and Cell Therapy, Hajdrihova 19, Ljubljana, Slovenia.
- University of Ljubljana, Kongresni trg 12, 1000, Ljubljana, Slovenia.
| |
Collapse
|
42
|
Otsuka S, Dutta D, Wu CJ, Alam MS, Ashwell JD. Calcineurin is an adaptor required for assembly of the TCR signaling complex. Cell Rep 2024; 43:114568. [PMID: 39088318 PMCID: PMC11407306 DOI: 10.1016/j.celrep.2024.114568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/25/2024] [Accepted: 07/17/2024] [Indexed: 08/03/2024] Open
Abstract
The serine/threonine phosphatase calcineurin is a component of the T cell receptor (TCR) signalosome, where it promotes T cell activation by dephosphorylating LckS59. Using small interfering RNA (siRNA)-mediated knockdown and CRISPR-Cas9-targeted genetic disruption of the calcineurin A chain α and β isoforms, we find that calcineurin also functions as an adaptor in TCR-signaled human T cells. Unlike inhibition of its phosphatase activity, in the absence of calcineurin A, TCR signaling results in attenuated actin rearrangement, markedly reduced TCR-Lck microcluster formation and recruitment of the adaptor RhoH, and diminished phosphorylation of critical targets downstream of Lck such as TCRζ and ZAP-70. Reconstitution of deficient T cells with either calcineurin Aα or Aβ restores TCR microcluster formation and signaling, as does reconstitution with a phosphatase-inactive Aα chain. These results assign a non-enzymatic adaptor function to calcineurin in the formation and stabilization of a functional TCR signaling complex.
Collapse
Affiliation(s)
- Shizuka Otsuka
- Laboratory of Immune Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Debjani Dutta
- Laboratory of Immune Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chuan-Jin Wu
- Laboratory of Immune Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Muhammad S Alam
- Laboratory of Immune Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jonathan D Ashwell
- Laboratory of Immune Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
43
|
Chuan J, Li W, Pan S, Jiang Z, Shi J, Yang Z. Progress in the development of modulators targeting Frizzleds. Pharmacol Res 2024; 206:107286. [PMID: 38936522 DOI: 10.1016/j.phrs.2024.107286] [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: 03/19/2024] [Revised: 06/08/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
The Frizzleds (FZDs) receptors on the cell surface belong to the class F of G protein-coupled receptors (GPCRs) which are the major receptors of WNT protein that mediates the classical WNT signaling pathway and other non-classical pathways. Besides, the FZDs also play a core role in tissue regeneration and tumor occurrence. With the structure and mechanism of FZDs activation becoming clearer, a series of FZDs modulators (inhibitors and agonists) have been developed, with the hope of bringing benefits to the treatment of cancer and degenerative diseases. Most of the FZDs inhibitors (small molecules, antibodies or designed protein inhibitors) block WNT signaling through binding to the cysteine-rich domain (CRD) of FZDs. Several small molecules impede FZDs activation by targeting to the third intracellular domain or the transmembrane domain of FZDs. However, three small molecules (FZM1.8, SAG1.3 and purmorphamine) activate the FZDs through direct interaction with the transmembrane domain. Another type of FZDs agonists are bivalent or tetravalent antibodies which activate the WNT signaling via inducing FZD-LRP5/6 heterodimerization. In this article, we reviewed the FZDs modulators reported in recent years, summarized the critical molecules' discovery processes and the elucidated relevant structural and pharmacological mechanisms. We believe the summaried molecular mechanisms of the relevant modulators could provide important guidance and reference for the future development of FZD modulators.
Collapse
Affiliation(s)
- Junlan Chuan
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei Li
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, No. 9, Section 4, Renmin South Road, Chengdu 610041, China; The University of Chinese Academy of Sciences, 380 Huaibeizhuang, Huairou District, Beijing 101408, China
| | - Shengliu Pan
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, No. 9, Section 4, Renmin South Road, Chengdu 610041, China; The University of Chinese Academy of Sciences, 380 Huaibeizhuang, Huairou District, Beijing 101408, China
| | - Zhongliang Jiang
- Hematology Department, Miller School of Medicine, University of Miami, USA
| | - Jianyou Shi
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Zhenglin Yang
- Research Unit for Blindness Prevention, Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; Jinfeng Laboratory, Chongqing, China.
| |
Collapse
|
44
|
Ponticelli C, Reggiani F, Moroni G. Autophagy: A Silent Protagonist in Kidney Transplantation. Transplantation 2024; 108:1532-1541. [PMID: 37953477 DOI: 10.1097/tp.0000000000004862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Autophagy is a lysosome-dependent regulated mechanism that recycles unnecessary cytoplasmic components. It is now known that autophagy dysfunction may have a pathogenic role in several human diseases and conditions, including kidney transplantation. Both defective and excessive autophagy may induce or aggravate several complications of kidney transplantation, such as ischemia-reperfusion injury, alloimmune response, and immunosuppressive treatment and side effects. Although it is still complicated to measure autophagy levels in clinical practice, more attention should be paid to the factors that may influence autophagy. In kidney transplantation, the association of low doses of a mammalian target of rapamycin inhibitor with low doses of a calcineurin inhibitor may be of benefit for autophagy modulation. However, further studies are needed to explore the role of other autophagy regulators.
Collapse
Affiliation(s)
| | - Francesco Reggiani
- Nephrology and Dialysis Unit, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Gabriella Moroni
- Nephrology and Dialysis Unit, IRCCS Humanitas Research Hospital, Milan, Italy
| |
Collapse
|
45
|
Chen S, Yi W, Zhou H, Jiang H, Lan P, Chen Z. FOS+ Macrophages Promote Chronic Rejection of Cardiac Transplantation. EXP CLIN TRANSPLANT 2024; 22:540-550. [PMID: 39223812 DOI: 10.6002/ect.2024.0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
OBJECTIVES Chronic rejection remains the leading cause of progressive decline in graft function. Accumulating evidence indicates that macrophages participate in chronic rejection dependent on CD40-CD40L. The FOS family members are critical in inflammatory and immune responses. However, the mechanisms underlying the role of FOS family members in chronic rejection remain unclear. In this study, we aimed to elucidate the role and underlying mechanisms of FOS-positive macrophages regulated by CD40 that mediate chronic allograft rejection. MATERIALS AND METHODS We downloaded publicly accessible chronic rejection kidney transplant single-cell sequencing datasets from the gene expression omnibus database. Differentially expressed genes between the CD40hi and CD40low macrophage chronic rejection groups were analyzed. We established a chronic rejection mouse model by using CTLA-4-Ig. We treated bone marrow-derived macrophages with an anti-CD40 antibody. We assessed expression of the FOS family by flow cytometry, real-time quantitative polymerase chain reaction, Western blotting, and immunofluorescence. We identified altered signaling pathways by using RNA sequencing analysis. We detected DNA specifically bound to transcription factors by using ChIP-sequencing, with detection of the degree of graft fibrosis and survival. RESULTS FOS was highly expressed on CD40hi macrophages in patients with chronic transplantrejection. Mechanistically, we showed that CD40 activated NF-κB2 translocation into the nucleus to upregulate c-Fos and FosB expression, thus promoting chronic rejection of cardiac transplant.We showed thatNF-κB2 regulated c-Fos and FosB expression by binding to the c-fos and fosb promoter regions. Inhibition of c-Fos/activator protein-1 decreased graft fibrosis and prolonged graft survival. CONCLUSIONS CD40 may activate transcription factor NF-κB2 translocation into the nucleus of macrophages to upregulate c-Fos and FosB expression, thus promoting chronic rejection of cardiac transplant. Inhibition of c-Fos/activator protein-1 decreased grafts fibrosis and prolonged graft survival.
Collapse
Affiliation(s)
- Shi Chen
- >From the Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China Key Laboratory of Organ Transplantation, Ministry of Education; the NHC Key Laboratory of Organ Transplantation; and the Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | | | | | | | | | | |
Collapse
|
46
|
Luna-Angulo A, Landa-Solís C, Escobar-Cedillo RE, Estrada-Mena FJ, Sánchez-Chapul L, Gómez-Díaz B, Carrillo-Mora P, Avilés-Arnaut H, Jiménez-Hernández L, Jiménez-Hernández DA, Miranda-Duarte A. Pharmacological Treatments and Therapeutic Targets in Muscle Dystrophies Generated by Alterations in Dystrophin-Associated Proteins. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1060. [PMID: 39064489 PMCID: PMC11279157 DOI: 10.3390/medicina60071060] [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: 05/29/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024]
Abstract
Muscular dystrophies (MDs) are a heterogeneous group of diseases of genetic origin characterized by progressive skeletal muscle degeneration and weakness. There are several types of MDs, varying in terms of age of onset, severity, and pattern of the affected muscles. However, all of them worsen over time, and many patients will eventually lose their ability to walk. In addition to skeletal muscle effects, patients with MDs may present cardiac and respiratory disorders, generating complications that could lead to death. Interdisciplinary management is required to improve the surveillance and quality of life of patients with an MD. At present, pharmacological therapy is only available for Duchene muscular dystrophy (DMD)-the most common type of MD-and is mainly based on the use of corticosteroids. Other MDs caused by alterations in dystrophin-associated proteins (DAPs) are less frequent but represent an important group within these diseases. Pharmacological alternatives with clinical potential in patients with MDs and other proteins associated with dystrophin have been scarcely explored. This review focuses on drugs and molecules that have shown beneficial effects, mainly in experimental models involving alterations in DAPs. The mechanisms associated with the effects leading to promising results regarding the recovery or maintenance of muscle strength and reduction in fibrosis in the less-common MDs (i.e., with respect to DMD) are explored, and other therapeutic targets that could contribute to maintaining the homeostasis of muscle fibers, involving different pathways, such as calcium regulation, hypertrophy, and maintenance of satellite cell function, are also examined. It is possible that some of the drugs explored here could be used to affordably improve the muscular function of patients until a definitive treatment for MDs is developed.
Collapse
Affiliation(s)
- Alexandra Luna-Angulo
- División de Neurociencias Clinicas, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Calzada México-Xochimilco, No. 289, Arenal de Guadalupe, Tlalpan, Ciudad de México 14389, Mexico
| | - Carlos Landa-Solís
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, División de Biotecnología, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Calzada México-Xochimilco, No. 289, Arenal de Guadalupe, Tlalpan, Ciudad de México 14389, Mexico
| | - Rosa Elena Escobar-Cedillo
- Departamento de Electromiografía y Distrofia Muscular, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Calzada México-Xochimilco, No. 289, Arenal de Guadalupe, Tlalpan, Ciudad de México 14389, Mexico
| | - Francisco Javier Estrada-Mena
- Laboratorio de Biología Molecular, Universidad Panamericana, Facultad de Ciencias de la Salud, Augusto Rodin 498, Ciudad de México 03920, Mexico
| | - Laura Sánchez-Chapul
- División de Neurociencias Clinicas, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Calzada México-Xochimilco, No. 289, Arenal de Guadalupe, Tlalpan, Ciudad de México 14389, Mexico
| | - Benjamín Gómez-Díaz
- Departamento de Medicina Genómica, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Calzada México-Xochimilco, No. 289, Arenal de Guadalupe, Tlalpan, Ciudad de México 14389, Mexico
| | - Paul Carrillo-Mora
- División de Neurociencias Clinicas, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Calzada México-Xochimilco, No. 289, Arenal de Guadalupe, Tlalpan, Ciudad de México 14389, Mexico
| | - Hamlet Avilés-Arnaut
- Facultad de Ciencias Biológicas de la Universidad Autónoma de Nuevo Leon, Av. Universidad s/n Ciudad Universitaria, San Nicolas de los Garza 66455, Mexico
| | | | | | - Antonio Miranda-Duarte
- Departamento de Medicina Genómica, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Calzada México-Xochimilco, No. 289, Arenal de Guadalupe, Tlalpan, Ciudad de México 14389, Mexico
| |
Collapse
|
47
|
Ahn T, Bae EA, Seo H. Decoding and overcoming T cell exhaustion: Epigenetic and transcriptional dynamics in CAR-T cells against solid tumors. Mol Ther 2024; 32:1617-1627. [PMID: 38582965 PMCID: PMC11184340 DOI: 10.1016/j.ymthe.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 02/14/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024] Open
Abstract
T cell exhaustion, which is observed in various chronic infections and malignancies, is characterized by elevated expression of multiple inhibitory receptors, impaired effector functions, decreased proliferation, and reduced cytokine production. Notably, while adoptive T cell therapies, such as chimeric antigen receptor (CAR)-T therapy, have shown promise in treating cancer and other diseases, the efficacy of these therapies is often compromised by T cell exhaustion. It is imperative, therefore, to understand the mechanisms underlying this exhaustion to promote advances in T cell-related therapies. Here, we divided exhausted T cells into three distinct subsets according to their developmental and functional profiles: stem-like progenitor cells, intermediately exhausted cells, and terminally exhausted cells. These subsets are carefully regulated by synergistic mechanisms that involve transcriptional and epigenetic modulators. Key transcription factors, such as TCF1, BACH2, and TOX, are crucial for defining and sustaining exhaustion phenotypes. Concurrently, epigenetic regulators, such as TET2 and DNMT3A, shape the chromatin dynamics that direct T cell fate. The interplay of these molecular drivers has recently been highlighted in CAR-T research, revealing promising therapeutic directions. Thus, a profound understanding of exhausted T cell hierarchies and their molecular complexities may reveal innovative and improved tumor treatment strategies.
Collapse
Affiliation(s)
- Taeyoung Ahn
- Laboratory of Cell & Gene Therapy, Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun-Ah Bae
- Laboratory of Immunology, Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyungseok Seo
- Laboratory of Cell & Gene Therapy, Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
48
|
Kennedy PH, Alborzian Deh Sheikh A, Balakar M, Jones AC, Olive ME, Hegde M, Matias MI, Pirete N, Burt R, Levy J, Little T, Hogan PG, Liu DR, Doench JG, Newton AC, Gottschalk RA, de Boer CG, Alarcón S, Newby GA, Myers SA. Post-translational modification-centric base editor screens to assess phosphorylation site functionality in high throughput. Nat Methods 2024; 21:1033-1043. [PMID: 38684783 PMCID: PMC11804830 DOI: 10.1038/s41592-024-02256-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 03/20/2024] [Indexed: 05/02/2024]
Abstract
Signaling pathways that drive gene expression are typically depicted as having a dozen or so landmark phosphorylation and transcriptional events. In reality, thousands of dynamic post-translational modifications (PTMs) orchestrate nearly every cellular function, and we lack technologies to find causal links between these vast biochemical pathways and genetic circuits at scale. Here we describe the high-throughput, functional assessment of phosphorylation sites through the development of PTM-centric base editing coupled to phenotypic screens, directed by temporally resolved phosphoproteomics. Using T cell activation as a model, we observe hundreds of unstudied phosphorylation sites that modulate NFAT transcriptional activity. We identify the phosphorylation-mediated nuclear localization of PHLPP1, which promotes NFAT but inhibits NFκB activity. We also find that specific phosphosite mutants can alter gene expression in subtle yet distinct patterns, demonstrating the potential for fine-tuning transcriptional responses. Overall, base editor screening of PTM sites provides a powerful platform to dissect PTM function within signaling pathways.
Collapse
Affiliation(s)
- Patrick H Kennedy
- Laboratory for Immunochemical Circuits, La Jolla Institute for Immunology, La Jolla, CA, USA
- Center of Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Amin Alborzian Deh Sheikh
- Laboratory for Immunochemical Circuits, La Jolla Institute for Immunology, La Jolla, CA, USA
- Center of Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Alexander C Jones
- Department of Pharmacology, University of California San Diego, San Diego, CA, USA
- Biomedical Sciences Graduate Program, University of California San Diego, San Diego, CA, USA
| | | | - Mudra Hegde
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Maria I Matias
- Laboratory for Immunochemical Circuits, La Jolla Institute for Immunology, La Jolla, CA, USA
- Center of Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Natan Pirete
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rajan Burt
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonathan Levy
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Tamia Little
- Laboratory for Immunochemical Circuits, La Jolla Institute for Immunology, La Jolla, CA, USA
- Center of Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Patrick G Hogan
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA, USA
- Program in Immunology, University of California San Diego, San Diego, CA, USA
- Moores Cancer Center, University of California San Diego Health, La Jolla, CA, USA
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - John G Doench
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alexandra C Newton
- Department of Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Rachel A Gottschalk
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Carl G de Boer
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Suzie Alarcón
- La Jolla Institute for Immunology, La Jolla, CA, USA
- AUGenomics, San Diego, CA, USA
| | - Gregory A Newby
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Samuel A Myers
- Laboratory for Immunochemical Circuits, La Jolla Institute for Immunology, La Jolla, CA, USA.
- Center of Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA.
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA, USA.
- Department of Pharmacology, University of California San Diego, San Diego, CA, USA.
- Program in Immunology, University of California San Diego, San Diego, CA, USA.
- Moores Cancer Center, University of California San Diego Health, La Jolla, CA, USA.
| |
Collapse
|
49
|
Gülow K, Tümen D, Heumann P, Schmid S, Kandulski A, Müller M, Kunst C. Unraveling the Role of Reactive Oxygen Species in T Lymphocyte Signaling. Int J Mol Sci 2024; 25:6114. [PMID: 38892300 PMCID: PMC11172744 DOI: 10.3390/ijms25116114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Reactive oxygen species (ROS) are central to inter- and intracellular signaling. Their localized and transient effects are due to their short half-life, especially when generated in controlled amounts. Upon T cell receptor (TCR) activation, regulated ROS signaling is primarily initiated by complexes I and III of the electron transport chain (ETC). Subsequent ROS production triggers the activation of nicotinamide adenine dinucleotide phosphate oxidase 2 (NADPH oxidase 2), prolonging the oxidative signal. This signal then engages kinase signaling cascades such as the mitogen-activated protein kinase (MAPK) pathway and increases the activity of REDOX-sensitive transcription factors such as nuclear factor-kappa B (NF-κB) and activator protein-1 (AP-1). To limit ROS overproduction and prevent oxidative stress, nuclear factor erythroid 2-related factor 2 (Nrf2) and antioxidant proteins such as superoxide dismutases (SODs) finely regulate signal intensity and are capable of terminating the oxidative signal when needed. Thus, oxidative signals, such as T cell activation, are well-controlled and critical for cellular communication.
Collapse
Affiliation(s)
- Karsten Gülow
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, Immunology, and Infectious Diseases, University Hospital Regensburg, 93053 Regensburg, Germany; (D.T.); (P.H.); (S.S.); (A.K.); (M.M.); (C.K.)
| | | | | | | | | | | | | |
Collapse
|
50
|
Nishida S, Azetsu Y, Chatani M, Karakawa A, Otake K, Sugiki H, Sakai N, Maruoka Y, Myers M, Takami M. Tacrolimus, FK506, promotes bone formation in bone defect mouse model. J Oral Biosci 2024; 66:391-402. [PMID: 38360372 DOI: 10.1016/j.job.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 02/17/2024]
Abstract
OBJECTIVES Some studies have reported that tacrolimus (FK506), an immunosuppressant, may have positive effects on bone formation. However, the precise effects of FK506 on bone repair or osteoblasts remain inadequately elucidated, and limited research has explored the outcomes of its use in an in vivo mouse model. This study aims to examine the effects of FK506 on bone repair and osteoblast functions using bone defect and BMP-2-induced ectopic ossification mouse models, as well as cultured primary mouse osteoblasts treated with FK506. METHODS We established mouse models of femur bone defect and BMP-2-induced ectopic ossification to evaluate the effect of FK506 on new bone formation, respectively. Additionally, primary mouse osteoblasts were cultured with FK506 and examined for gene expressions related to osteoblast differentiation. RESULTS While FK506 promoted the repair of bone defect areas in the femur of the bone defect mouse model, it also led to widespread abnormal bone formation outside the intended area. Additionally, following the implantation of a collagen sponge containing BMP-2 into mouse muscle tissue, FK506 was found to promote ectopic ossification and enhance BMP-2-induced osteoblast differentiation in vitro. Our findings also revealed that FK506 increased the number of immature osteoblasts in the absence of BMP-2 without affecting osteoblast differentiation. Furthermore, direct effects were observed, reducing the ability of osteoblasts to support osteoclastogenesis. CONCLUSIONS These results indicate that FK506 increases new bone formation during bone repair and influences the proliferation of immature osteoblasts, as well as osteoblast-supported osteoclastogenesis.
Collapse
Affiliation(s)
- Satoko Nishida
- Department of Pharmacology, Graduate School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan; Department of Medical and Dental Cooperative Dentistry, Graduate School of Dentistry, Showa University, 2-1-1 Kitasenzoku, Ota, Tokyo, 145-8515, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Yuki Azetsu
- Department of Pharmacology, Graduate School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Masahiro Chatani
- Department of Pharmacology, Graduate School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Akiko Karakawa
- Department of Pharmacology, Graduate School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Kai Otake
- Department of Pharmacology, Graduate School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan; Department of Endodontology, Graduate School of Dentistry, Showa University, 2-1-1 Kitasenzoku, Ota, Tokyo, 145-8515, Japan
| | - Hidemitsu Sugiki
- Department of Pharmacology, School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Nobuhiro Sakai
- Department of Dental Education, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Yasubumi Maruoka
- Totsuka Kyoritsu Daini Hospital, 579-1 Totsuka, Yokohama, Kanagawa, 244-0817, Japan
| | - Mie Myers
- Department of Medical and Dental Cooperative Dentistry, School of Dentistry, Showa University, 2-1-1 Kitasenzoku, Ota, Tokyo, 145-8515, Japan
| | - Masamichi Takami
- Department of Pharmacology, Graduate School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan.
| |
Collapse
|