1
|
Zhang F, Li Y, Jiang L, Wang Y, Tang Y, Ouyang B, Song G, Li X, Quan X, Li M, Wang H, Pan Z, Chen K, Zhang P. Ectopic expression of Slc1a2 in the prefrontal cortex of sleep-deprived male mice counteracts the glutamate/GABA-glutamine dysfunction. BMC Biol 2025; 23:86. [PMID: 40140835 PMCID: PMC11948699 DOI: 10.1186/s12915-025-02161-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 02/14/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND The prefrontal cortex (PFC) plays a pronounced role in cognitive and emotional functions, which may be compromised by dismal sleep quality. This study intended to clarify the impact of Slc1a2 ectopic expression in the PFC on sleep deprivation (SD)-induced disturbances in the glutamate (Glu)/GABA-glutamine cycle and the role of astrocyte (AC)-neuron (Neu) communication. METHODS Single-cell RNA sequencing was adopted to illuminate cell-specific changes in the brainstem, cortex, and hypothalamus of mice under NS, SD, and post-SD conditions. Cell communication analysis was applied to study interactions between ACs and Neus, which altered after the SD. Slc1a2 was ectopically expressed in the PFC and subjected to SD, followed by electrophysiological, immunofluorescence staining, and [1H-13C]-nuclear magnetic resonance (NMR) assays to examine neural activity and metabolic status. Behavioral tests, including the open field, novel object recognition, and Y-maze, were conducted to examine cognitive functions and emotional states. RESULTS SD caused notable changes in cellular distribution and downregulation of metabolic and synaptic genes in affected brain regions. Cell communication studies highlighted a reduction in AC-Neu interactions, with corresponding metabolic disruptions in the Glu/GABA-glutamine cycle as depicted by [1H-13C]-NMR results. Behavior tests confirmed anxiety and cognitive deficits in SD mice, which were substantially alleviated by Slc1a2 ectopic expression in the PFC. CONCLUSIONS Slc1a2 ectopic expression in the PFC negates SD-induced GABA dysfunction through vital AC-Neu communication. This study sheds light on the mechanisms through which SD affects neural function and suggesting potential treatments for sleep-related disorders.
Collapse
Affiliation(s)
- Fengying Zhang
- Department of Neurology, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, No. 336, Dongfeng South Road, Zhuhui District, Hengyang, Hunan Province, 421001, China
- Department of Sleep Medical Centre, the Affiliated Nanhua Hospital,Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yao Li
- Department of Physiology, Jinzhou Medical University, Jinzhou, 121000, China
| | - Li Jiang
- Department of Neurology, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, No. 336, Dongfeng South Road, Zhuhui District, Hengyang, Hunan Province, 421001, China
- Department of Sleep Medical Centre, the Affiliated Nanhua Hospital,Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yingbin Wang
- Department of Neurosurgery, Central Hospital Affiliated to Shenyang Medical College, Shenyang, 110024, China
| | - Yonghong Tang
- Department of Neurology, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, No. 336, Dongfeng South Road, Zhuhui District, Hengyang, Hunan Province, 421001, China
- Department of Sleep Medical Centre, the Affiliated Nanhua Hospital,Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Bo Ouyang
- Department of Sleep Medical Centre, the Affiliated Nanhua Hospital,Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Department of Traditional Chinese Medicine, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Guojun Song
- Department of Radiology, The Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan, 421001, China
| | - Xuan Li
- Department of Docimasiology, Hengyang Medical School, The Affiliated Nanhua Hospital, University of South China, Hengyang, 421002, Hunan, China
| | - Xiajie Quan
- Department of Sleep Medical Centre, the Affiliated Nanhua Hospital,Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Department of Information, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Min Li
- Department of Neurology, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, No. 336, Dongfeng South Road, Zhuhui District, Hengyang, Hunan Province, 421001, China
- Department of Sleep Medical Centre, the Affiliated Nanhua Hospital,Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Hongying Wang
- Department of Respiratory and Critical Care Medicine, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zhao Pan
- Department of Neurology, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, No. 336, Dongfeng South Road, Zhuhui District, Hengyang, Hunan Province, 421001, China
- Department of Sleep Medical Centre, the Affiliated Nanhua Hospital,Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Keyan Chen
- Laboratory Animal Science of China Medical University, No.77 Puhe Road, Shenyang, Liaoning, 110122, China.
| | - Ping Zhang
- Department of Neurology, the Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, No. 336, Dongfeng South Road, Zhuhui District, Hengyang, Hunan Province, 421001, China.
- Department of Sleep Medical Centre, the Affiliated Nanhua Hospital,Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
2
|
Li C, Wang P, Dong Z, Cao W, Su Y, Zhang J, Zhao S, Wang Z, Lei Z, Shi L, Cheng R, Liu W. Single-cell transcriptomics analysis reveals that the tumor-infiltrating B cells determine the indolent fate of papillary thyroid carcinoma. J Exp Clin Cancer Res 2025; 44:91. [PMID: 40069827 PMCID: PMC11895268 DOI: 10.1186/s13046-025-03341-7] [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: 12/31/2024] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
OBJECTIVE Active surveillance (AS) offers a viable alternative to surgical intervention for the management of indolent papillary thyroid carcinoma (PTC), helping to minimize the incidence of unnecessary treatment. However, the broader adoption of AS is hindered by the need for more reliable diagnostic markers. This study aimed to identify the differences between indolent and progressive PTC and find new targets for biomarker development and therapeutic strategies. METHODS We used single-cell RNA sequencing (scRNA-seq) to analyze cellular differences in 10 early-stage PTC tumors. Findings were validated in an additional 25 tumors using cell co-culture, migration assays, immunofluorescence staining, flow cytometry, and analysis of data from The Cancer Genome Atlas (TCGA). RESULTS Tumor-infiltrating B cells (TIL-B), particularly germinal center B cells (GC-B), were more abundant in indolent PTC. These cells suppressed thyroid cell proliferation in both indolent and progressive cases, though indolent PTC had a higher capacity to recruit peripheral B cells. In indolent cases, TIL-B cells showed increased proliferation and formed clusters within tertiary lymphoid structures (TLS). PTPRC-CD22 interactions were identified as potential drivers of TIL-B cell proliferation. Markers linked to GC-B cells, such as LMO2, were highlighted as potential diagnostic and prognostic indicators for indolent PTC. CONCLUSION This study provides insights into the cellular landscape of early-stage PTC, revealing distinct tumor and immune microenvironment features in indolent and progressive cases. These findings advance the understanding of indolent PTC biology and support the development of reliable diagnostic and prognostic biomarkers.
Collapse
Affiliation(s)
- Chunmei Li
- State Key Laboratory for Conservation and Utilization of Bio-resources and School of Life Sciences, Yunnan University, Kunming, Yunnan, 650091, China
| | - Pei Wang
- Department of Radiation Oncology, Cancer Institute, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Zhizhong Dong
- Department of Thyroid Surgery, Clinical Research Center for Thyroid Diseases of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Weihan Cao
- Department of Ultrasound Imaging, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yanjun Su
- Department of Thyroid Surgery, Clinical Research Center for Thyroid Diseases of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jianming Zhang
- Department of Thyroid Surgery, Clinical Research Center for Thyroid Diseases of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Shuyan Zhao
- Department of Thyroid Surgery, Clinical Research Center for Thyroid Diseases of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhiyuan Wang
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zi Lei
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Li Shi
- Endocrine and Metabolic Diseases Clinical Medical Center of Yunnan, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Ruochuan Cheng
- Department of Thyroid Surgery, Clinical Research Center for Thyroid Diseases of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| | - Wen Liu
- Department of Thyroid Surgery, Clinical Research Center for Thyroid Diseases of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
3
|
Zhu Y, Su Y, Guo Y, Wang X, Zhang Z, Lu Y, Yang H, Pang H. Current state of cancer immunity cycle: new strategies and challenges of using precision hydrogels to treat breast cancer. Front Immunol 2025; 16:1535464. [PMID: 40124373 PMCID: PMC11926806 DOI: 10.3389/fimmu.2025.1535464] [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: 11/27/2024] [Accepted: 02/19/2025] [Indexed: 03/25/2025] Open
Abstract
The cancer-immunity cycle provides a framework for a series of events in anti-cancer immune responses, initiated by T cell-mediated tumor cell killing, which leads to antigen presentation and T cell stimulation. Current immunomodulatory therapies for breast cancer are often associated with short duration, poor targeting to sites of action, and severe side effects. Hydrogels, with their extracellular matrix-mimicking properties, tunable characteristics, and diverse bioactivities, have garnered significant attention for their ability to locally deliver immunomodulators and cells, providing an immunomodulatory microenvironment to recruit, activate, and expand host immune cells. This review focuses on the design considerations of hydrogel platforms, including polymer backbone, crosslinking mechanisms, physicochemical properties, and immunomodulatory components. The immunomodulatory effects and therapeutic outcomes of various hydrogel systems in breast cancer treatment and tissue regeneration are highlighted, encompassing hydrogel depots for immunomodulator delivery, hydrogel scaffolds for cell delivery, and immunomodulatory hydrogels dependent on inherent material properties. Finally, the challenges that persist in current systems and future directions for immunomodulatory hydrogels are discussed.
Collapse
Affiliation(s)
- Yingze Zhu
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yanlin Su
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yaxin Guo
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xinyue Wang
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Zhuoqi Zhang
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yige Lu
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hang Yang
- Department of Gastroenterology and Hepatology, Tianjin Second People’s Hospital, Tianjin, China
| | - Hui Pang
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
4
|
Xie D, Ma Y, Gao C, Pan S. Piezo1 activation on microglial cells exacerbates demyelination in sepsis by influencing the CCL25/GRP78 pathway. Int Immunopharmacol 2024; 142:113045. [PMID: 39236454 DOI: 10.1016/j.intimp.2024.113045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND In sepsis-associated encephalopathy (SAE), the activation of microglial cells and ensuing neuroinflammation are important in the underlying pathological mechanisms. Increasing evidence suggests that the protein Piezo1 functions as a significant regulator of neuroinflammation. However, the influence of Piezo1 on microglial cells in the context of SAE has not yet been determined. This study aims to investigate the role of Piezo1 in microglial cells in the context of SAE. METHODS By inducing cecal ligation and puncture (CLP), a mouse model of SAE was established, while the control group underwent a sham surgery in which the cecum was exposed without ligation and puncture. Piezo1 knockout mice were employed in this study. Morris water maze tests were conducted between Days 14 and 18 postop to assess both the motor activity and cognitive function. A proteomic analysis was conducted to assess the SAE-related pathways, whereas a Mendelian randomization analysis was conducted to identify the pathways associated with cognitive impairment. Dual-label immunofluorescence and flow cytometry were used to assess the secretion of inflammatory factors, microglial status, and oligodendrocyte development. Electron microscopy was used to evaluate axonal myelination. A western blot analysis was conducted to evaluate the influence of Piezo1 on oligodendrocyte ferroptosis. RESULTS The results of the bioinformatics analysis have revealed the significant involvement of CCL25 in the onset and progression of SAE-induced cognitive impairment. SAE leads to cognitive dysfunction by activating the microglial cells. The release of CCL25 by the activated microglia initiates the demyelination of oligodendrocytes in the hippocampus, resulting in ferroptosis and the disruption of hippocampal functional connectivity. Of note, the genetic knockout of the Piezo1 gene mitigates these changes. The treatment with siRNA targeting Piezo1 effectively reduces the secretion of inflammatory mediators CCL25 and IL-18 by inhibiting the p38 pathway, thus preventing the ferroptosis of oligodendrocytes through the modulation of the CCL25/GPR78 axis. CONCLUSION Piezo1 is involved in the activation of microglia and demyelinating oligodendrocytes in the animal models of SAE, resulting in cognitive impairment. Consequently, targeting Piezo1 suppression can be a promising approach for therapeutic interventions aimed at addressing cognitive dysfunction associated with SAE.
Collapse
Affiliation(s)
- Di Xie
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, 200092 Shanghai, China
| | - Yanli Ma
- Department of Pediatrics, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Hongkou District, 200434 Shanghai, China
| | - Chengjin Gao
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, 200092 Shanghai, China.
| | - Shuming Pan
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, 200092 Shanghai, China; Department of Emergency, Putuo District Central Hospital, Affiliated with Shanghai University of Traditional Chinese Medicine, Putuo District, 200062 Shanghai, China.
| |
Collapse
|
5
|
Zhang X, Guo L, Tian W, Yang Y, Yin Y, Qiu Y, Wang W, Li Y, Zhang G, Zhao X, Wang G, Lin Z, Yang M, Zhao W, Lu D. CD36+ Proinflammatory Macrophages Interact with ZCCHC12+ Tumor Cells in Papillary Thyroid Cancer Promoting Tumor Progression and Recurrence. Cancer Immunol Res 2024; 12:1621-1639. [PMID: 39178310 DOI: 10.1158/2326-6066.cir-23-1047] [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: 12/12/2023] [Revised: 04/28/2024] [Accepted: 08/21/2024] [Indexed: 08/25/2024]
Abstract
Local recurrence and distal metastasis negatively impact the survival and quality of life in patients with papillary thyroid cancer (PTC). Therefore, identifying potential biomarkers and therapeutic targets for PTC is clinically crucial. In this study, we performed a multiomics analysis that identified a subset of CD36+ proinflammatory macrophages within the tumor microenvironment of PTC. The recruitment of CD36+ macrophages to premalignant regions strongly correlated with unfavorable outcomes in PTC, and the presence of tumor-infiltrating CD36+ macrophages was determined to be a risk factor for recurrence. The CD36+ macrophages exhibited interactions with metabolically active ZCCHC12+ tumor cells. By secreting SPP1, the CD36+ macrophages activated the PI3K-AKT signaling pathway, thereby promoting proliferation of the cancer cells. Dysregulation of iodine metabolism was closely related to the acquisition of the pro-inflammatory phenotype in macrophages. Iodine supplementation inhibited the activation of proinflammatory signaling and impeded the development of CD36+ macrophages by enhancing DUSP2 expression. Overall, our findings shed light on the intricate cross-talk between CD36+ macrophages and ZCCHC12+ tumor cells, providing valuable insights for the treatment and prognosis of PTC.
Collapse
Affiliation(s)
- Xin Zhang
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University, Beijing, P.R. China
| | - Limei Guo
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University, Beijing, P.R. China
| | - Wenyu Tian
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University, Beijing, P.R. China
| | - Ying Yang
- Department of Blood Transfusion, Peking University People's Hospital, Beijing, P.R. China
| | - Yue Yin
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University, Beijing, P.R. China
| | - Yaruo Qiu
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University, Beijing, P.R. China
| | - Weixuan Wang
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University, Beijing, P.R. China
| | - Yang Li
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University, Beijing, P.R. China
| | - Guangze Zhang
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University, Beijing, P.R. China
| | - Xuyang Zhao
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University, Beijing, P.R. China
| | - Guangxi Wang
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University, Beijing, P.R. China
| | - Zhiqiang Lin
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University, Beijing, P.R. China
| | - Meng Yang
- Department of General Surgery, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Wei Zhao
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Dan Lu
- Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University, Beijing, P.R. China
| |
Collapse
|
6
|
Lai X, Luo J, Luo Y, Zheng Y, Yang H, Zou F. Targeting the autoreactive CD8 + T-cell receptor in type 1 diabetes: Insights from scRNA-seq for immunotherapy. Pharmacol Res 2024; 209:107433. [PMID: 39343113 DOI: 10.1016/j.phrs.2024.107433] [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: 05/15/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
Type 1 Diabetes (T1D) is an autoimmune disease characterized by the attack and destruction of Pancreatic islet beta cells by T cells. Understanding the role of T-cell receptor (TCR) in the development of T1D is of paramount importance. This study employs single-cell RNA sequencing (scRNA-seq) to delve into the mechanistic actions and potential therapeutic applications of autoreactive stem cell-like CD8 TCR in T1D. By retrieving T-cell data from non-obese diabetic (NOD) mice via the GEO database, it was revealed that CD8+ T cells are the predominant T-cell subset in the pancreatic tissue of T1D mice, along with the identification of T-cell marker genes closely associated with T1D. Moreover, the gene TRAJ23 exhibits a preference for T1D, and its knockout alleviates T1D symptoms and adverse reactions in NOD mice. Additionally, engineered TCR-T cells demonstrate significant cytotoxicity towards β cells in T1D.
Collapse
Affiliation(s)
- Xiaoyang Lai
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Junming Luo
- Department of Respiratory Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Yue Luo
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Yijing Zheng
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Huan Yang
- Department of Endocrinology, Jiujiang University Affiliated Hospital, Jiujiang, PR China
| | - Fang Zou
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, PR China.
| |
Collapse
|
7
|
Ma X, Sun Y, Li C, Wang M, Zang Q, Zhang X, Wang F, Niu Y, Hua J. Novel Insights Into DLAT's Role in Alzheimer's Disease-Related Copper Toxicity Through Microglial Exosome Dynamics. CNS Neurosci Ther 2024; 30:e70064. [PMID: 39428563 PMCID: PMC11491298 DOI: 10.1111/cns.70064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 08/10/2024] [Accepted: 09/03/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a complex neurodegenerative disorder, with recent research emphasizing the roles of microglia and their secreted extracellular vesicles in AD pathology. However, the involvement of specific molecular pathways contributing to neuronal death in the context of copper toxicity remains largely unexplored. OBJECTIVE This study investigates the interaction between pyruvate kinase M2 (PKM2) and dihydrolipoamide S-acetyltransferase (DLAT), particularly focusing on copper-induced neuronal death in Alzheimer's disease. METHODS Gene expression datasets were analyzed to identify key factors involved in AD-related copper toxicity. The role of DLAT was validated using 5xFAD transgenic mice, while in vitro experiments were conducted to assess the impact of microglial exosomes on neuronal PKM2 transfer and DLAT expression. The effects of inhibiting the PKM2 transfer via microglial exosomes on DLAT expression and copper-induced neuronal death were also evaluated. RESULTS DLAT was identified as a critical factor in the pathology of AD, particularly in copper toxicity. In 5xFAD mice, increased DLAT expression was linked to hippocampal damage and cognitive decline. In vitro, microglial exosomes were shown to facilitate the transfer of PKM2 to neurons, leading to upregulation of DLAT expression and increased copper-induced neuronal death. Inhibition of PKM2 transfer via exosomes resulted in a significant reduction in DLAT expression, mitigating neuronal death and slowing AD progression. CONCLUSION This study uncovers a novel pathway involving microglial exosomes and the PKM2-DLAT interaction in copper-induced neuronal death, providing potential therapeutic targets for Alzheimer's disease. Blocking PKM2 transfer could offer new strategies for reducing neuronal damage and slowing disease progression in AD.
Collapse
Affiliation(s)
- Xiang Ma
- Laboratory of Biochemistry and PharmacyTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Yusheng Sun
- Laboratory of Biochemistry and PharmacyTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Changchun Li
- Department of Chemistry and Chemical EngineeringTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Man Wang
- Laboratory of Biochemistry and PharmacyTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Qijiao Zang
- Laboratory of Biochemistry and PharmacyTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Xuxia Zhang
- Laboratory of Biochemistry and PharmacyTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Feng Wang
- Department of Chemistry and Chemical EngineeringTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Yulan Niu
- Department of Chemistry and Chemical EngineeringTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Jiai Hua
- Laboratory of Biochemistry and PharmacyTaiyuan Institute of TechnologyTaiyuanP. R. China
| |
Collapse
|
8
|
Qiu S, Cao L, Xiang D, Wang S, Wang D, Qian Y, Li X, Zhou X. Enhanced osteogenic differentiation in 3D hydrogel scaffold via macrophage mitochondrial transfer. J Nanobiotechnology 2024; 22:540. [PMID: 39237942 PMCID: PMC11375923 DOI: 10.1186/s12951-024-02757-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
To assess the efficacy of a novel 3D biomimetic hydrogel scaffold with immunomodulatory properties in promoting fracture healing. Immunomodulatory scaffolds were used in cell experiments, osteotomy mice treatment, and single-cell transcriptomic sequencing. In vitro, fluorescence tracing examined macrophage mitochondrial transfer and osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs). Scaffold efficacy was assessed through alkaline phosphatase (ALP), Alizarin Red S (ARS) staining, and in vivo experiments. The scaffold demonstrated excellent biocompatibility and antioxidant-immune regulation. Single-cell sequencing revealed a shift in macrophage distribution towards the M2 phenotype. In vitro experiments showed that macrophage mitochondria promoted BMSCs' osteogenic differentiation. In vivo experiments confirmed accelerated fracture healing. The GAD/Ag-pIO scaffold enhances osteogenic differentiation and fracture healing through immunomodulation and promotion of macrophage mitochondrial transfer.
Collapse
Affiliation(s)
- Shui Qiu
- Department of Orthopedics, First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning Province, China
| | - Lili Cao
- Department of Medical Oncology, First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, China
| | - Dingding Xiang
- School of Mechanical Engineering and Automation, Foshan Graduate School of Innovation, Northeastern University, Shenyang, 110819, China
| | - Shu Wang
- School of Mechanical Engineering and Automation, Foshan Graduate School of Innovation, Northeastern University, Shenyang, 110819, China
| | - Di Wang
- School of Mechanical Engineering and Automation, Foshan Graduate School of Innovation, Northeastern University, Shenyang, 110819, China
| | - Yiyi Qian
- School of Mechanical Engineering and Automation, Foshan Graduate School of Innovation, Northeastern University, Shenyang, 110819, China
| | - Xiaohua Li
- Department of Orthopedics, Zhongmeng Hospital, Arong Banner, Hulunbuir City, Inner, Mongolia
| | - Xiaoshu Zhou
- Department of Orthopedics, First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning Province, China.
| |
Collapse
|
9
|
Li M, Fu X, Zhou T, Han H. Biomarkers related to m6A and succinic acid metabolism in papillary thyroid carcinoma. BMC Med Genomics 2024; 17:199. [PMID: 39113023 PMCID: PMC11304613 DOI: 10.1186/s12920-024-01975-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 07/30/2024] [Indexed: 08/11/2024] Open
Abstract
BACKGROUND Studies have shown that m6A modification is related to the occurrence and development of papillary thyroid carcinoma (PTC). The disorder of succinic acid metabolism is associated with the occurrence and development of various tumors. However, there are few studies based on m6A and succinate metabolism-related genes (SMRGs) in PTC. METHODS The TCGA-Thyroid carcinoma (THCA), GSE33630, 1159 SMRGs, and 23 m6A regulatory factors were collected from the online databases. Subsequently, the differentially expressed genes (DEGs) were selected between PTC (Tumor) and Normal samples. The overlapping genes among the DEGs, m6A, and SMRGs were applied to screen the biomarkers. Using the 3 machine-learning algorithms, the biomarkers were determined based on the overlapping genes. Next, the biomarkers were evaluated by the ROC curve and expression analysis in TCGA-THCA and GSE33630. Then, the overall survival (OS) differences were compared between the high-and low-expression biomarkers. Finally, immune infiltration analysis, molecular regulatory network, and drug prediction were performed based on the biomarkers. RESULTS In TCGA-THCA, there were 2800 DEGs between and Normal samples, and then 7 overlapping genes were obtained. Importantly, ADK, TNFRSF10B, CYP7B1, FGFR2, and CPQ were determined as biomarkers with excellent diagnostic efficiency (AUC > 0.7). In PTC samples, ADK and TNFRSF10B were high-expressed while CYP7B1, FGFR2, and CPQ were low-expressed. Especially, the high-expression groups of ADK had a better prognosis, while the high-expression groups of CYP7B1, FGFR2, and CPQ had a worse prognosis. Afterward, immune infiltration analysis found that 16 immune cells had infiltration differences between the Tumor and Normal samples. Finally, transcription factor SP1 could regulate CYP7B1 and TNFRSF10B. Moreover, Navitoclax was a potential drug for PTC patients. CONCLUSION Overall, we described 5 biomarkers associated with adverse prognosis of PTC, including ADK, TNFRSF10B, CYP7B1, FGFR2, and CPQ. All these biomarkers were involved in succinate metabolism and m6A modification of RNA. This set of biomarkers should be explored further for their diagnostic value in PTC. Investigations into the mechanistic role of alteration of succinate metabolism and m6A modification of RNA pathways in the pathophysiology of PTC are warranted.
Collapse
Affiliation(s)
- Minyu Li
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiaodan Fu
- Department of Endocrinology, Affiliated Hangzhou First People's Hospital, School of medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Tianhan Zhou
- The Department of General Surgery, Hangzhou Hospital of Traditional Chinese Medicine Hangzhou TCM Hospital, Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Hui Han
- Department of Endocrinology, Affiliated Hangzhou First People's Hospital, School of medicine, Westlake University, Hangzhou, Zhejiang, China.
| |
Collapse
|
10
|
Martínez-Hernández R, Sánchez de la Blanca N, Sacristán-Gómez P, Serrano-Somavilla A, Muñoz De Nova JL, Sánchez Cabo F, Heyn H, Sampedro-Núñez M, Marazuela M. Unraveling the molecular architecture of autoimmune thyroid diseases at spatial resolution. Nat Commun 2024; 15:5895. [PMID: 39003267 PMCID: PMC11246508 DOI: 10.1038/s41467-024-50192-5] [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: 11/08/2023] [Accepted: 06/25/2024] [Indexed: 07/15/2024] Open
Abstract
Autoimmune thyroid diseases (AITD) such as Graves' disease (GD) or Hashimoto's thyroiditis (HT) are organ-specific diseases that involve complex interactions between distinct components of thyroid tissue. Here, we use spatial transcriptomics to explore the molecular architecture, heterogeneity and location of different cells present in the thyroid tissue, including thyroid follicular cells (TFCs), stromal cells such as fibroblasts, endothelial cells, and thyroid infiltrating lymphocytes. We identify damaged antigen-presenting TFCs with upregulated CD74 and MIF expression in thyroid samples from AITD patients. Furthermore, we discern two main fibroblast subpopulations in the connective tissue including ADIRF+ myofibroblasts, mainly enriched in GD, and inflammatory fibroblasts, enriched in HT patients. We also demonstrate an increase of fenestrated PLVAP+ vessels in AITD, especially in GD. Our data unveil stromal and thyroid epithelial cell subpopulations that could play a role in the pathogenesis of AITD.
Collapse
Affiliation(s)
- Rebeca Martínez-Hernández
- Department of Endocrinology and Nutrition Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER GCV14/ER/12), Madrid, Spain.
| | - Nuria Sánchez de la Blanca
- Department of Endocrinology and Nutrition Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER GCV14/ER/12), Madrid, Spain
| | - Pablo Sacristán-Gómez
- Department of Endocrinology and Nutrition Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER GCV14/ER/12), Madrid, Spain
| | - Ana Serrano-Somavilla
- Department of Endocrinology and Nutrition Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER GCV14/ER/12), Madrid, Spain
| | - José Luis Muñoz De Nova
- Department of General and Digestive Surgery, Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Fátima Sánchez Cabo
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Holger Heyn
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain
- Universitat de Barcelona (UB), Barcelona, Spain
| | - Miguel Sampedro-Núñez
- Department of Endocrinology and Nutrition Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER GCV14/ER/12), Madrid, Spain
| | - Mónica Marazuela
- Department of Endocrinology and Nutrition Hospital Universitario de la Princesa, Instituto de Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER GCV14/ER/12), Madrid, Spain.
| |
Collapse
|
11
|
Tong X, Qiao S, Dong Z, Zhao X, Du X, Niu W. Targeting CSF1R in myeloid-derived suppressor cells: insights into its immunomodulatory functions in colorectal cancer and therapeutic implications. J Nanobiotechnology 2024; 22:409. [PMID: 38992688 PMCID: PMC11238447 DOI: 10.1186/s12951-024-02584-4] [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: 01/21/2024] [Accepted: 05/26/2024] [Indexed: 07/13/2024] Open
Abstract
OBJECTIVE This study aimed to investigate the critical role of MDSCs in CRC immune suppression, focusing on the CSF1R and JAK/STAT3 signaling axis. Additionally, it assessed the therapeutic efficacy of LNCs@CSF1R siRNA and anti-PD-1 in combination. METHODS Single-cell transcriptome sequencing data from CRC and adjacent normal tissues identified MDSC-related differentially expressed genes. RNA-seq analysis comprehensively profiled MDSC gene expression in murine CRC tumors. LNCs@CSF1R siRNA nanocarriers effectively targeted and inhibited CSF1R. Flow cytometry quantified changes in MDSC surface markers post-CSF1R inhibition. RNA-seq and pathway enrichment analyses revealed the impact of CSF1R on MDSC metabolism and signaling. The effect of CSF1R inhibition on the JAK/STAT3 signaling axis was validated using Colivelin and metabolic assessments. Glucose and fatty acid uptake were measured via fluorescence-based flow cytometry. The efficacy of LNCs@CSF1R siRNA and anti-PD-1, alone and in combination, was evaluated in a murine CRC model with extensive tumor section analyses. RESULTS CSF1R played a significant role in MDSC-mediated immune suppression. LNCs@CSF1R siRNA nanocarriers effectively targeted MDSCs and inhibited CSF1R. CSF1R regulated MDSC fatty acid metabolism and immune suppression through the JAK/STAT3 signaling axis. Inhibition of CSF1R reduced STAT3 activation and target gene expression, which was rescued by Colivelin. Combined treatment with LNCs@CSF1R siRNA and anti-PD-1 significantly slowed tumor growth and reduced MDSC abundance within CRC tumors. CONCLUSION CSF1R via the JAK/STAT3 axis critically regulates MDSCs, particularly in fatty acid metabolism and immune suppression. Combined therapy with LNCs@CSF1R siRNA and anti-PD-1 enhances therapeutic efficacy in a murine CRC model, providing a strong foundation for future clinical applications.
Collapse
Affiliation(s)
- Xin Tong
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, P. R. China
| | - Shifeng Qiao
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, P. R. China
| | - Zhe Dong
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, P. R. China
| | - Xiaohui Zhao
- Department of Medical Oncology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, P. R. China
| | - Xiaxia Du
- Department of Rehabilitation, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, P. R. China
| | - Wei Niu
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning Province, 121000, P. R. China.
| |
Collapse
|
12
|
Wang X, Chen J, Li C, Liu Y, Chen S, Lv F, Lan K, He W, Zhu H, Xu L, Ma K, Guo H. Integrated bulk and single-cell RNA sequencing identifies an aneuploidy-based gene signature to predict sensitivity of lung adenocarcinoma to traditional chemotherapy drugs and patients' prognosis. PeerJ 2024; 12:e17545. [PMID: 38938612 PMCID: PMC11210463 DOI: 10.7717/peerj.17545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 05/19/2024] [Indexed: 06/29/2024] Open
Abstract
Background Patients with lung adenocarcinoma (LUAD) often develop a poor prognosis. Currently, researches on prognostic and immunotherapeutic capacity of aneuploidy-related genes in LUAD are limited. Methods Genes related to aneuploidy were screened based on bulk RNA sequencing data from public databases using Spearman method. Next, univariate Cox and Lasso regression analyses were performed to establish an aneuploidy-related riskscore (ARS) model. Results derived from bioinformatics analysis were further validated using cellular experiments. In addition, typical LUAD cells were identified by subtype clustering, followed by SCENIC and intercellular communication analyses. Finally, ESTIMATE, ssGSEA and CIBERSORT algorithms were employed to analyze the potential relationship between ARS and tumor immune environment. Results A five-gene ARS signature was developed. These genes were abnormally high-expressed in LUAD cell lines, and in particular the high expression of CKS1B promoted the proliferative, migratory and invasive phenotypes of LUAD cell lines. Low ARS group had longer overall survival time, higher degrees of inflammatory infiltration, and could benefit more from receiving immunotherapy. Patients in low ASR group responded more actively to traditional chemotherapy drugs (Erlotinib and Roscovitine). The scRNA-seq analysis annotated 17 cell subpopulations into seven cell clusters. Core transcription factors (TFs) such as CREB3L1 and CEBPD were enriched in high ARS cell group, while TFs such as BCLAF1 and UQCRB were enriched in low ARS cell group. CellChat analysis revealed that high ARS cell groups communicated with immune cells via SPP1 (ITGA4-ITGB1) and MK (MDK-NCl) signaling pathways. Conclusion In this research, integrative analysis based on the ARS model provided a potential direction for improving the diagnosis and treatment of LUAD.
Collapse
Affiliation(s)
- Xiaobin Wang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Military Medical University, Xi’an, China
| | - Jiakuan Chen
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Military Medical University, Xi’an, China
| | - Chaofan Li
- Department of Thoracic Surgery, The 986 Military Medical Hospital of the Air Force, Xi’an, China
| | - Yufei Liu
- Department of Thoracic Surgery, The 986 Military Medical Hospital of the Air Force, Xi’an, China
| | - Shiqun Chen
- Thoracic Surgery, Weinan Central Hospital, Weinan, China
| | - Feng Lv
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Military Medical University, Xi’an, China
| | - Ke Lan
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Military Medical University, Xi’an, China
| | - Wei He
- Department of Thoracic Surgery, The 986 Military Medical Hospital of the Air Force, Xi’an, China
| | - Hongsheng Zhu
- Thoracic Surgery, Shaanxi Chenggu County Hospital, Chenggu, China
| | - Liang Xu
- Thoracic Surgery, Shaanxi Chenggu County Hospital, Chenggu, China
| | - Kaiyuan Ma
- Thoracic Surgery, Shaanxi Chenggu County Hospital, Chenggu, China
| | - Haihua Guo
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Military Medical University, Xi’an, China
| |
Collapse
|
13
|
Yan S, Zhao J, Gao P, Li Z, Li Z, Liu X, Wang P. Diagnostic potential of NRG1 in benign nerve sheath tumors and its influence on the PI3K-Akt signaling and tumor immunity. Diagn Pathol 2024; 19:28. [PMID: 38331905 PMCID: PMC10851500 DOI: 10.1186/s13000-024-01438-9] [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: 11/06/2023] [Accepted: 01/04/2024] [Indexed: 02/10/2024] Open
Abstract
OBJECTIVE Benign nerve sheath tumors (BNSTs) present diagnostic challenges due to their heterogeneous nature. This study aimed to determine the significance of NRG1 as a novel diagnostic biomarker in BNST, emphasizing its involvement in the PI3K-Akt pathway and tumor immune regulation. METHODS Differential genes related to BNST were identified from the GEO database. Gene co-expression networks, protein-protein interaction networks, and LASSO regression were utilized to pinpoint key genes. The CIBERSORT algorithm assessed immune cell infiltration differences, and functional enrichment analyses explored BNST signaling pathways. Clinical samples helped establish PDX models, and in vitro cell lines to validate NRG1's role via the PI3K-Akt pathway. RESULTS Nine hundred eighty-two genes were upregulated, and 375 downregulated in BNST samples. WGCNA revealed the brown module with the most significant difference. Top hub genes included NRG1, which was also determined as a pivotal gene in disease characterization. Immune infiltration showed significant variances in neutrophils and M2 macrophages, with NRG1 playing a central role. Functional analyses confirmed NRG1's involvement in key pathways. Validation experiments using PDX models and cell lines further solidified NRG1's role in BNST. CONCLUSION NRG1 emerges as a potential diagnostic biomarker for BNST, influencing the PI3K-Akt pathway, and shaping the tumor immune microenvironment.
Collapse
Affiliation(s)
- Suwei Yan
- Department of Neurosurgery, The Third Hospital of Hebei Medical University, No. 139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, Hebei Province, P. R. China
| | - Jingnan Zhao
- Department of Neurosurgery, The Third Hospital of Hebei Medical University, No. 139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, Hebei Province, P. R. China
| | - Pengyang Gao
- Department of Neurosurgery, The Third Hospital of Hebei Medical University, No. 139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, Hebei Province, P. R. China
| | - Zhaoxu Li
- Department of Neurosurgery, The Third Hospital of Hebei Medical University, No. 139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, Hebei Province, P. R. China
| | - Zhao Li
- Department of Neurosurgery, The Third Hospital of Hebei Medical University, No. 139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, Hebei Province, P. R. China
| | - Xiaobing Liu
- Department of Neurosurgery, The Third Hospital of Hebei Medical University, No. 139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, Hebei Province, P. R. China
| | - Pengfei Wang
- Department of Neurosurgery, The Third Hospital of Hebei Medical University, No. 139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, Hebei Province, P. R. China.
| |
Collapse
|
14
|
Zhang W, Wang J, Huang D, Liu Z, Lu T, Cui C, Li Z. Single-cell sequencing reveals SATB2/NOTCH1 signaling promotes the progression of malignancy of epithelial cells from papillary thyroid cancer. Mol Carcinog 2024; 63:22-33. [PMID: 37877736 DOI: 10.1002/mc.23631] [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: 04/12/2023] [Revised: 08/01/2023] [Accepted: 08/22/2023] [Indexed: 10/26/2023]
Abstract
Although most papillary thyroid cancers (PTCs) are deemed to have a favorable clinical course and outcome, some develop an aggressive biological behavior at diagnosis or during treatment. Single-cell RNA sequencing (scRNA-seq), which is based on quantifying the features of individual cells to resolve tumor tissue heterogeneity, was used to uncover gene regulatory relationships and trace the transcriptional trajectories underlying the malignant transformation. In this study, we performed single-cell sequencing on samples from four PTC patients and one benign thyroid tumor patient. These included two papillary thyroid microcarcinoma cancers (PTMC) patients, two age-matched advanced PTC patients with invading surrounding tissues, and one patient undergoing surgical treatment due to a benign thyroid tumor. We constructed a new PTC RNA spectrum using single-cell sequencing. Single-cell sequencing analysis indicated that there was a highly invasive subgroup in the PTC epithelial cells, the expression of SATB2 (special AT-rich binding protein-2) was related to the prognosis and clinical progress of PTC, and SATB2 could promote the proliferation, migration, and invasion of PTC cells. We found that NOTCH1 was the key target gene of SATB2, and the activation of the SATB2/NOTCH1 pathway was one of the reasons for the high carcinogenicity of this subgroup.
Collapse
Affiliation(s)
- Wenqian Zhang
- Department of Head and Neck Surgery, Cancer Hospital of China Medical University, Shenyang, China
| | - Jing Wang
- Department of Head and Neck Surgery, Cancer Hospital of China Medical University, Shenyang, China
| | - Dongning Huang
- Department of Head and Neck Surgery, Cancer Hospital of China Medical University, Shenyang, China
| | - Zhu Liu
- Department of Head and Neck Surgery, Cancer Hospital of China Medical University, Shenyang, China
| | - Tie Lu
- Department of Head and Neck Surgery, Cancer Hospital of China Medical University, Shenyang, China
| | - Chen Cui
- Department of Head and Neck Surgery, Cancer Hospital of China Medical University, Shenyang, China
| | - Zhendong Li
- Department of Head and Neck Surgery, Cancer Hospital of China Medical University, Shenyang, China
| |
Collapse
|
15
|
Tan JK, Awuah WA, Roy S, Ferreira T, Ahluwalia A, Guggilapu S, Javed M, Asyura MMAZ, Adebusoye FT, Ramamoorthy K, Paoletti E, Abdul-Rahman T, Prykhodko O, Ovechkin D. Exploring the advances of single-cell RNA sequencing in thyroid cancer: a narrative review. Med Oncol 2023; 41:27. [PMID: 38129369 PMCID: PMC10739406 DOI: 10.1007/s12032-023-02260-x] [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: 08/04/2023] [Accepted: 11/16/2023] [Indexed: 12/23/2023]
Abstract
Thyroid cancer, a prevalent form of endocrine malignancy, has witnessed a substantial increase in occurrence in recent decades. To gain a comprehensive understanding of thyroid cancer at the single-cell level, this narrative review evaluates the applications of single-cell RNA sequencing (scRNA-seq) in thyroid cancer research. ScRNA-seq has revolutionised the identification and characterisation of distinct cell subpopulations, cell-to-cell communications, and receptor interactions, revealing unprecedented heterogeneity and shedding light on novel biomarkers for therapeutic discovery. These findings aid in the construction of predictive models on disease prognosis and therapeutic efficacy. Altogether, scRNA-seq has deepened our understanding of the tumour microenvironment immunologic insights, informing future studies in the development of effective personalised treatment for patients. Challenges and limitations of scRNA-seq, such as technical biases, financial barriers, and ethical concerns, are discussed. Advancements in computational methods, the advent of artificial intelligence (AI), machine learning (ML), and deep learning (DL), and the importance of single-cell data sharing and collaborative efforts are highlighted. Future directions of scRNA-seq in thyroid cancer research include investigating intra-tumoral heterogeneity, integrating with other omics technologies, exploring the non-coding RNA landscape, and studying rare subtypes. Overall, scRNA-seq has transformed thyroid cancer research and holds immense potential for advancing personalised therapies and improving patient outcomes. Efforts to make this technology more accessible and cost-effective will be crucial to ensuring its widespread utilisation in healthcare.
Collapse
Affiliation(s)
| | | | - Sakshi Roy
- School of Medicine, Queen's University Belfast, Belfast, UK
| | - Tomas Ferreira
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | | | - Saibaba Guggilapu
- Faculty of Medicine, Bangalore Medical College and Research Institute, Bengaluru, India
| | - Mahnoor Javed
- School of Medicine, The University of Nottingham, Nottingham, NG7 2UH, UK
| | | | | | | | - Emma Paoletti
- Faculty of Medicine, University of Manchester, Manchester, M13 9WJ, UK
| | | | - Olha Prykhodko
- Faculty of Medicine, Sumy State University, Sumy, Ukraine
| | - Denys Ovechkin
- Faculty of Medicine, Sumy State University, Sumy, Ukraine
| |
Collapse
|
16
|
Abstract
Thyroid cancer (TC) is the most common endocrine malignancy worldwide, and the incidence of TC has gradually increased in recent decades. Differentiated thyroid cancer (DTC) is the most common subtype and has a good prognosis. However, advanced DTC patients with recurrence, metastasis and iodine refractoriness, as well as more aggressive subtypes such as poorly differentiated thyroid cancer (PDTC) and anaplastic thyroid cancer (ATC), still pose a great challenge for clinical management. Therefore, it is necessary to continue to explore the inherent molecular heterogeneity of different TC subtypes and the global landscape of the tumor immune microenvironment (TIME) to find new potential therapeutic targets. Immunotherapy is a promising therapeutic strategy that can be used alone or in combination with drugs targeting tumor-driven genes. This article focuses on the genomic characteristics, tumor-associated immune cell infiltration and immune checkpoint expression of different subtypes of TC patients to provide guidance for immunotherapy.
Collapse
Affiliation(s)
- Yujia Tao
- School of Medical Laboratory, Weifang Medical University, Weifang, Shandong, China
- Department of Basic Medical Sciences, The 960th Hospital of the PLA, Jinan, Shandong, China
| | - Peng Li
- Department of Basic Medical Sciences, The 960th Hospital of the PLA, Jinan, Shandong, China
| | - Chao Feng
- Department of Basic Medical Sciences, The 960th Hospital of the PLA, Jinan, Shandong, China
| | - Yuan Cao
- Department of Basic Medical Sciences, The 960th Hospital of the PLA, Jinan, Shandong, China
| |
Collapse
|
17
|
Fagin JA, Krishnamoorthy GP, Landa I. Pathogenesis of cancers derived from thyroid follicular cells. Nat Rev Cancer 2023; 23:631-650. [PMID: 37438605 PMCID: PMC10763075 DOI: 10.1038/s41568-023-00598-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/08/2023] [Indexed: 07/14/2023]
Abstract
The genomic simplicity of differentiated cancers derived from thyroid follicular cells offers unique insights into how oncogenic drivers impact tumour phenotype. Essentially, the main oncoproteins in thyroid cancer activate nodes in the receptor tyrosine kinase-RAS-BRAF pathway, which constitutively induces MAPK signalling to varying degrees consistent with their specific biochemical mechanisms of action. The magnitude of the flux through the MAPK signalling pathway determines key elements of thyroid cancer biology, including differentiation state, invasive properties and the cellular composition of the tumour microenvironment. Progression of disease results from genomic lesions that drive immortalization, disrupt chromatin accessibility and cause cell cycle checkpoint dysfunction, in conjunction with a tumour microenvironment characterized by progressive immunosuppression. This Review charts the genomic trajectories of these common endocrine tumours, while connecting them to the biological states that they confer.
Collapse
Affiliation(s)
- James A Fagin
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Gnana P Krishnamoorthy
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Iñigo Landa
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
18
|
Abuduwaili M, Su A, Xing Z, Xia B, Wu Z, Fei Y, Zhu J, Chen Z. Clinical significance of extrathyroidal extension to major vessels in papillary thyroid carcinoma. J Endocrinol Invest 2022; 46:1155-1167. [PMID: 36427135 DOI: 10.1007/s40618-022-01966-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 11/13/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE Gross extrathyroidal extension (gETE) into major vessel is considered the most advanced stage of the locally advanced papillary thyroid cancer (PTC). Surgical intervention may not benefit some patients at this disease stage or even result in intraoperative death due to massive hemorrhage; however, it is still considered an effective strategy for most cases. The lack of description for this challenging invasion in PTC warrants detailed characterization of its pattern, risk factors, optimal surgical method, and prognostic value. METHODS In total, 3127 patients diagnosed as having PTC were enrolled and categorized into two the following groups, namely the major vessel invasion (MVI) group (n = 30) and the control group (n = 3097). Data regarding clinicopathological and demographic characteristics, vascular invasion sites, postoperative complications, locoregional recurrence, distant metastasis, and surgical strategies were collected. Predictive disease-free survival (DFS) was also compared between the two groups. RESULTS MVI was independently associated with invasion of the esophageal extension, age < 55 years, tumor size > 1 cm, lateral lymph node metastasis, and distant metastasis (P = 0.00; P = 0.01; 0.05; P = 0.00; P = 0.00, respectively). The difference in the predictive DFS between the two groups was significant (P = 0.00), and the difference remained significant even in patients with ETE when compared with patients without ETE (P = 0.00). Additionally, predictive DFS did not differ significantly between patients who received vessel repairment and those who received vessel resection (P = 0.28). CONCLUSIONS This study first characterized the gross MVI pattern exhibited by PTC and the risk factors for MVI. Additionally, it demonstrated the DFS of patients with PTC. Extensive gross MVI significantly worsened the biological characteristics of PTC. Regardless of the high risk and difficulty of the operation, patients still benefited from the surgical intervention, and vessel repairment may be the optimal surgical strategy.
Collapse
Affiliation(s)
- M Abuduwaili
- Center of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, China
| | - A Su
- Center of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, China.
| | - Z Xing
- Center of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, China
| | - B Xia
- Center of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, China
| | - Z Wu
- Center of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, China
| | - Y Fei
- Center of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, China
| | - J Zhu
- Center of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, China
| | - Z Chen
- Center of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, China
| |
Collapse
|