1
|
Zhao Y, An D, Bi L. Effect of Co-Administration of Midazolam and Dexmedetomidine on Haemodynamics and Stress Response in Elderly Patients with Non-Small Cell Lung Cancer. J INVEST SURG 2025; 38:2445587. [PMID: 39756799 DOI: 10.1080/08941939.2024.2445587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 10/11/2024] [Accepted: 12/16/2024] [Indexed: 01/07/2025]
Abstract
OBJECTIVE This study aimed to evaluate the effect of co-administration of midazolam and dexmedetomidine on hemodynamics and stress response in elderly patients with non-small cell lung cancer (NSCLC). METHODS In this prospective, randomized controlled trial, 154 elderly NSCLC patients scheduled for lobectomy in our oncology department from January 2019 to December 2021 were recruited. Patients were randomized 1:1 to receive either dexmedetomidine (control group) or dexmedetomidine plus midazolam (study group) for anesthesia during lobectomy via the random number table method, with 77 patients in each group. Perioperative indicators, hemodynamics, and stress reactions of the patients were recorded and compared between the two groups to investigate the efficacy of the two different anesthetic protocols. RESULTS No significant differences were observed between the two groups in terms of operative time, anesthesia time, and intraoperative bleeding volume (p > 0.05). Preoperative pain, pain at anesthesia recovery, and pain levels 7 days postoperatively were also comparable between the two groups. In the study group, the awakening time was 15 ± 2 min significantly shorter compared to the control group (25 ± 3 min). Cooperation within the first hour was significantly faster by 8.5 ± 0.5 min compared to 6.0 ± 1.0 min in the control group (p < 0.05). The cost of materials used was significantly higher in the study group, with an average of 300 ± 25 USD, compared to 200 ± 20 USD in the control group (p < 0.05). Additionally, the two groups showed no significant difference in the need for experience and surveillance (p > 0.05). Significantly lower visual analog scale (VAS) scores were found one day after the surgery in patients given dexmedetomidine plus midazolam than those anesthetized administered with dexmedetomidine only, suggesting an enhanced pain mitigation effect after incorporating midazolam for anesthetic induction. Patients treated with dexmedetomidine plus midazolam presented with a more stable hemodynamic status than those treated with dexmedetomidine only, as evidenced by the significantly lower variability of mean arterial pressure (MAP), oxygen saturation (SpO2), and heart rate (HR). Co-administration of dexmedetomidine plus midazolam for lobectomy anesthesia resulted in significantly lower serum cortisol (Cor) and norepinephrine (NE) concentrations in patients at anesthesia recovery than dexmedetomidine alone. However, this difference was not observed one day postoperatively. There was no statistically significant difference in the incidence of adverse reactions between the two groups. CONCLUSION The combination of midazolam with dexmedetomidine anesthesia in lobectomy improves the intraoperative hemodynamic status of elderly patients with NSCLC and mitigates their stress response. However, further research is required to explore the underlying mechanisms.
Collapse
Affiliation(s)
- Yanjun Zhao
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Dongjiao An
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Liang Bi
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Wang Y, Chen Y, Xiao Z, Shi Y, Fu C, Cao Y. Fecal microbiota transplantation modulates myeloid-derived suppressor cells and attenuates renal fibrosis in a murine model. Ren Fail 2025; 47:2480749. [PMID: 40141007 PMCID: PMC11951334 DOI: 10.1080/0886022x.2025.2480749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Renal fibrosis is a hallmark of progressive chronic kidney disease (CKD), with emerging evidence linking gut microbiota dysbiosis to disease progression. Myeloid-derived suppressor cells (MDSCs) have demonstrated renoprotective effects, yet the impact of fecal microbiota transplantation (FMT) on MDSC-mediated modulation of renal fibrosis remains unclear. METHODS C57BL/6J mice underwent unilateral ureteral obstruction (UUO) to induce renal fibrosis, followed by FMT administration via gavage. Flow cytometry was used to quantify granulocytic (G-MDSCs) and monocytic (M-MDSCs) MDSC populations in peripheral blood, kidney, and spleen. To elucidate the role of MDSCs in FMT-mediated effects, MDSCs were depleted or adoptively transferred in vivo. Renal fibrosis severity and inflammatory cytokine expression were subsequently analyzed. RESULTS FMT altered MDSC distribution, increasing M-MDSC accumulation in the blood and kidney. This was associated with downregulation of proinflammatory cytokines and attenuation of renal fibrosis. Adoptive MDSC transfer similarly produced anti-inflammatory and antifibrotic effects, reinforcing their therapeutic role in FMT-mediated renal protection. CONCLUSIONS FMT enhances M-MDSC-mediated immunomodulation, reducing inflammation and renal fibrosis in UUO-induced CKD. These findings suggest a potential therapeutic strategy targeting the gut-kidney axis in CKD management.
Collapse
Affiliation(s)
- Yajie Wang
- Department of Nephrology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
- Anesthesia Laboratory and Training Center of Wannan Medical College, Wuhu, Anhui, China
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution (Wannan Medical College), Wuhu, Anhui, China
| | - Yuye Chen
- Department of Nephrology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Zihao Xiao
- Department of Nephrology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Yuanhui Shi
- Department of Nephrology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Cong Fu
- Department of Cardiology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Yuhan Cao
- Department of Nephrology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| |
Collapse
|
3
|
Wang X, Chen M, Wu Y, Chen F, Shen T, Wang Z. RORα inhibits proliferation and chemoresistance through AKR1A1-induced glucose and lipid reprogramming in gastric cancer. Cell Signal 2025; 131:111741. [PMID: 40096932 DOI: 10.1016/j.cellsig.2025.111741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/22/2025] [Accepted: 03/10/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Abnormal glycolysis and lipid metabolism play important roles in the occurrence and development of gastric cancer (GC). Moreover, dysregulation of circadian genes is associated with metabolic reprogramming in the tumor microenvironment. This study aimed to determine the role of retinoic acid-related orphan receptor alpha (RORα) in glucose and lipid reprogramming in GC. METHODS The effects on cell proliferation and chemoresistance in vitro and in vivo were studied using gain- and loss-of-function experiments. Glycolytic activity and lipid synthesis were assessed using a Seahorse assay and reagent kits. Moreover, the regulatory mechanisms were explored using half-life, coimmunoprecipitation (Co-IP), chromatin immunoprecipitation (ChIP), luciferase reporter and immunofluorescence colocalization assays in GC cells. In addition, the relationships of RORα with E47 and AKR1A1 were analyzed using public databases and retrospective clinicopathological analyses. RESULTS RORα deletion promoted cell proliferation and fluorouracil (5-FU) chemoresistance by increasing glycolytic activity and lipid synthesis. In contrast, SR1078, an RORα activator, reversed these changes and had a synergistic inhibitory effect on cell proliferation in combination with 2-deoxygulose glucose (2-DG) or atorvastatin. Mechanistically, aldo-keto reductase family 1 member A1 (AKR1A1), is the key driver of RORα-mediated glucose and lipid reprogramming. Specifically, E47 is an AKR1A1 transcription factor, and its stability is affected by β-catenin. RORα deletion indirectly promoted E47 protein stability through the up-regulation of β-catenin, leading to increased AKR1A1 transcriptional activity. Moreover, RORα, E47 and AKR1A1 expression was dysregulated, and associated with clinicopathological parameters and prognosis in patients with GC. These expression patterns including RORα-low, E47-high and AKR1A1-high expression patterns alone or in combination were correlated with reduced responsiveness, poor prognosis, increased standard uptake value (SUV) levels and lipid droplet formation. CONCLUSIONS These findings reveal a novel mechanism by which RORα regulates glucose and lipid reprogramming and may be a promising target for GC treatment.
Collapse
Affiliation(s)
- Xiaoshan Wang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mengding Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yuwei Wu
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Feixu Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Tong Shen
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China.
| | - Zhengguang Wang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
4
|
Nedaeinia R, Dianat-Moghadam H, Movahednasab M, Khosroabadi Z, Keshavarz M, Amoozgar Z, Salehi R. Therapeutic and prognostic values of ferroptosis signature in glioblastoma. Int Immunopharmacol 2025; 155:114597. [PMID: 40239336 DOI: 10.1016/j.intimp.2025.114597] [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: 01/03/2025] [Revised: 03/15/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025]
Abstract
Ferroptosis is a regulated cell death process that results in decreased tumor growth and aggressiveness when targeted in various cancer cells. Studying the impact of ferroptosis in glioblastoma (GBM) will provide important knowledge about tumor biology and potential treatment strategies. The high metabolic activity resulting in ROS production, iron content and active lipid metabolism of glioblastoma cells make them particularly susceptible to ferroptosis. Single-cell RNA sequencing reveals the molecular signature of GBM and its tumor microenvironment, introducing ferroptosis-related biomarkers pathways and drug resistance mechanisms to enhance treatment outcomes for GBM patients. The relationship between ferroptosis and the immune landscape in GBM is complex and can have either positive or negative effects. These effects can be identified through single-cell RNA sequencing to develop targeted chemo-, radio- and immuno- therapies against glioma stem cells and tumor-supportive immune cells. Additionally, the implication of oncolytic virotherapy in combination with ferroptosis induction can lead to improved treatment of GBM in a clinical setting.
Collapse
Affiliation(s)
- Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hassan Dianat-Moghadam
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Maedeh Movahednasab
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Khosroabadi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohsen Keshavarz
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Zohreh Amoozgar
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Rasoul Salehi
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
5
|
Tong M, Tu Q, Wang L, Chen H, Wan X, Xu Z. Joint analysis of single-cell RNA sequencing and bulk transcriptome reveals the heterogeneity of the urea cycle of astrocytes in glioblastoma. Neurobiol Dis 2025; 208:106835. [PMID: 39938577 DOI: 10.1016/j.nbd.2025.106835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/22/2025] [Accepted: 02/06/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Glioblastoma (GB) is incurable with a dismal prognosis. Single-cell RNA sequencing (scRNA-seq) is a pivotal tool for studying tumor heterogeneity. The dysregulation of the urea cycle (UC) frequently occurs in tumors, but its characteristics in GB have not been illuminated. This study integrated scRNA-seq UC scores and bulk RNA-seq data to build a GB prognostic model. METHODS Samples from 3 pairs of GB patients were collected for scRNA-seq analysis. GB-mRNA expression data, clinical data, and SNV mutation data were sourced from the Cancer Genome Atlas (TCGA). GB-mRNA expression data and clinical data were downloaded from the Chinese Glioma Genome Atlas (CGGA). GB RNA-seq data and clinical data were obtained from Gene Expression Omnibus (GEO) database. The R package Seurat was applied for scRNA-seq data processing. UMAP and TSNE were used for dimensionality reduction. UCell enrichment method was employed to score each astrocyte. Monocle algorithm was applied for pseudotime trajectory analysis. CellChat R package was applied for cell communication analysis. Cell labeling was performed on the results of the nine subclusters of astrocytes. The GSE138794 dataset was used to validate the results of single-cell classification. For bulk RNA-seq, univariate Cox and LASSO analyses were undertaken to screen prognostic genes, while multivariate Cox regression analysis was applied to set up a prognostic model. The differences between high-risk (HR) and low-risk (LR) groups were studied in terms of immune infiltration, sensitivity to anti-tumor drugs, etc. We verified the effect of the marker gene on the function of GB cells at the cellular level. RESULTS The analysis of scRNA-seq data yielded 7 core cell types. Further clustering of the largest proportion of astrocytes resulted in 9 subclusters. UC score and pseudotime analysis revealed the heterogeneity and differentiation process among subclusters. Subcluster 8 was annotated as an immature astrocyte (marker: CXCL8), and this cell cluster had a higher UC score. The results were validated in the GSE138794 dataset. Combining UC scores, we performed univariate Cox and LASSO to select prognostic genes on bulk RNA-seq data. A prognostic model based on 5 feature genes (RGS4, CTSB, SERPINE2, ID1, and CALD1) was established through multivariate Cox analysis. In addition, patients in the HR group had higher immune infiltration and immune function. Final cell experiments demonstrated that 5 feature genes were highly expressed in GB cells. CALD1 promoted the malignant phenotype of GB cells. CONCLUSION We set up a novel prognostic model for predicting the survival of GB patients by integrating bulk RNA-seq and scRNA-seq data. The risk score was closely correlated with immune infiltration and drug sensitivity, pinpointing it as a promising independent prognostic factor.
Collapse
Affiliation(s)
- Minfeng Tong
- Department of Neurosurgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Qi Tu
- Department of Neurosurgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Lude Wang
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Huahui Chen
- Department of Neurosurgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Xing Wan
- Department of Neurosurgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Zhijian Xu
- Department of Neurosurgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, China.
| |
Collapse
|
6
|
Liu Y, Li X, Huo C, Hou L, Jia X, Xu R, Yang J, Wang X. Caveolae Modulate the Activity of LRRC8-Mediated VRAC by the Structural Membrane Protein Caveolin-1. Cell Biol Int 2025; 49:484-493. [PMID: 39953952 DOI: 10.1002/cbin.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 01/10/2025] [Accepted: 01/27/2025] [Indexed: 02/17/2025]
Abstract
The volume-regulated anion channel (VRAC) plays a critical role in cell volume regulation and other fundamental physiological processes. However, the mechanism of how VRAC is activated and modulated has not been completely clarified. Caveolin-1 (Cav-1), as an important ion channel binding protein, forms complexes with channel proteins and exchangers to regulate channel activity and function. The purpose of this study was to explore the importance and value of Cav-1 in cardiac VRAC activation and regulation. In the study, we proved that the membrane protein LRRC8A was detected in the same caveolae-enriched fractions, as the same as Caveolin-1 in ventricular myocytes. The intracellular Cl- concentration increased and the cell volume decreased dramatically after caveolae being destroyed in cardiomyocytes. Moreover, we found that ICl,vol decreased not only in LRRC8A silencing cardiomyocytes but also in Cav-1 silencing cardiomyocytes, which indicated that caveolin-1 may affect the function of VRAC. Then we further explore the physical relationship between LRRC8A and Cav-1 in cell membrane. We observed that the fluorescence label of LRRC8A was overlapping with Cav-1 in the cell plasma membrane and caveolin-1 co-immunoprecipitated with LRRC8A, which demonstrated that Cav-1 is the basis of VRAC channel activation by acting on LRRC8A. The whole study provides further evidence of the relevance of Cav-1 on the activation and modulation of endothelial LRRC8A-mediated VRAC.
Collapse
Affiliation(s)
- Yan Liu
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Xing Li
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Cong Huo
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Liming Hou
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Xin Jia
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Rong Xu
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Jie Yang
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Xiaoming Wang
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| |
Collapse
|
7
|
Suzuki Y. Ca 2+ microdomains in vascular smooth muscle cells: Roles in vascular tone regulation and hypertension. J Pharmacol Sci 2025; 158:59-67. [PMID: 40121058 DOI: 10.1016/j.jphs.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025] Open
Abstract
Vascular smooth muscle cells (VSMCs) modulate blood pressure by adjusting vascular contractility. Specific families of ion channels that are expressed in VSMCs regulate membrane potential and intracellular Ca2+ concentration ([Ca2+]cyt). Subsets of them are known to form molecular complexes with Ca2+-sensitive molecules via scaffolding proteins such as caveolin and junctophilin. This enables localized and molecular complex-specific signal transduction to regulate vascular contractility. This intracellular region is referred to as a Ca2+ microdomain. When hypertensive stimuli are applied to blood vessels, gene expression of ion channels and scaffold proteins in vascular cells changes dramatically, often leading to membrane depolarization and increased [Ca2+]cyt. As a result, blood vessels undergo functional remodeling characterized by enhanced contractility. In addition, the transcription of inflammatory genes in vascular cells is also upregulated. This induces leukocyte infiltration into the vascular wall and structural remodeling mediated by VSMC proliferation and extracellular matrix remodeling. This functional and structural remodeling perpetuates the hypertensive state, leading to progressive damage to systemic organs. This review summarizes recent findings on the mechanisms by which Ca2+ microdomains in VSMCs regulate contractility. In addition, the changes in Ca2+ microdomains due to hypertensive stimuli and their contributions to both functional and structural remodeling are summarized.
Collapse
Affiliation(s)
- Yoshiaki Suzuki
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan.
| |
Collapse
|
8
|
BharathwajChetty B, Kumar A, Deevi P, Abbas M, Alqahtani A, Liang L, Sethi G, Liu L, Kunnumakkara AB. Gut microbiota and their influence in brain cancer milieu. J Neuroinflammation 2025; 22:129. [PMID: 40312370 PMCID: PMC12046817 DOI: 10.1186/s12974-025-03434-2] [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: 11/21/2024] [Accepted: 04/01/2025] [Indexed: 05/03/2025] Open
Abstract
Microbial communities are not simply remnants of the past but dynamic entities that continuously evolve under the selective pressures of nature, reflecting the intricate and adaptive processes of evolution. The microbiota residing in the various regions of the human body has numerous roles in different physiological processes such as nutrition, metabolism, immune regulation, etc. In the zeal of achieving empirical insights into the ambit of the gut microbiome, the research over the years led to the revelation of reciprocal interaction between the gut microbiome and the cognitive functioning of the human body. Dysbiosis in the gut microbial composition disturbs the homeostatic cognitive functioning of the human body. This dysbiosis has been associated with various chronic diseases, including brain cancer, such as glioma, glioblastoma, etc. This review explores the mechanistic role of dysbiosis-mediated progression of brain cancers and their subtypes. Moreover, it demonstrates the regulatory role of microbial metabolites produced by the gut microbiota, such as short-chain fatty acids, amino acids, lipids, etc., in the tumour progression. Further, we also provide valuable insights into the microbiota mediating the efficiency of therapeutic regimens, thereby leveraging gut microbiota as potential biomarkers and targets for improved treatment outcomes.
Collapse
Affiliation(s)
- Bandari BharathwajChetty
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Aviral Kumar
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Pranav Deevi
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
- International Joint M. Tech Degree in Food Science and Technology, Department of Chemical Engineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, 61421, Saudi Arabia
| | - Athba Alqahtani
- Research Centre, King Fahad Medical City, Riyadh, 11525, Saudi Arabia
| | - Liping Liang
- Guangzhou Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Centre for Cancer Research, Yong Loo Lin Scool of Medicine, National University of Singapore, Singapore, 117699, Singapore.
| | - Le Liu
- Integrated Clinical Microecology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China.
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
- International Joint M. Tech Degree in Food Science and Technology, Department of Chemical Engineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
| |
Collapse
|
9
|
Rafea R, Siragusa M, Fleming I. The Ever-Expanding Influence of the Endothelial Nitric Oxide Synthase. Basic Clin Pharmacol Toxicol 2025; 136:e70029. [PMID: 40150952 PMCID: PMC11950718 DOI: 10.1111/bcpt.70029] [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: 01/17/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
Nitric oxide (NO) generated by the endothelial NO synthase (eNOS) plays an essential role in the maintenance of vascular homeostasis and the prevention of vascular inflammation. There are a myriad of mechanisms that regulate the activity of the enzyme that may prove to represent interesting therapeutic opportunities. In this regard, the kinases that phosphorylate the enzyme and regulate its activity in situations linked to vascular disease seem to be particularly promising. Although the actions of NO were initially linked mainly to the activation of the guanylyl cyclase and the generation of cyclic GMP in vascular smooth muscle cells and platelets, it is now clear that NO elicits the majority of its actions via its ability to modify redox-activated cysteine residues in a process referred to as S-nitrosylation. The more wide spread use of mass spectrometry to detect S-nitrosylated proteins has helped to identify just how large the NO sphere of influence is and just how many cellular processes are affected. It may be an old target, but the sheer impact of eNOS on vascular health really justifies a revaluation of therapeutic options to maintain and protect its activity in situations associated with a high risk of developing cardiovascular disease.
Collapse
Affiliation(s)
- Riham Rafea
- Institute for Vascular Signalling, Centre for Molecular MedicineGoethe UniversityFrankfurt am MainGermany
| | - Mauro Siragusa
- Institute for Vascular Signalling, Centre for Molecular MedicineGoethe UniversityFrankfurt am MainGermany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular MedicineGoethe UniversityFrankfurt am MainGermany
- Partner Site RheinMainGerman Center for Cardiovascular Research (DZHK)Frankfurt am MainGermany
| |
Collapse
|
10
|
Wang L, Wei L, Wei J, Zhu Y, Zhao H, Zhang Y, Zhang S, Zhang P, Xing X, Wu D, Wang F, Yang S, Li Y, Huang J, Zhao Y, Zhang Y. 3D printed porous PEEK scaffolds with stable and durable gelatin composite hydrogel coating loaded Yoda1 for in vivo osseointegration. Int J Biol Macromol 2025; 307:141577. [PMID: 40054797 DOI: 10.1016/j.ijbiomac.2025.141577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/17/2025] [Accepted: 02/26/2025] [Indexed: 04/15/2025]
Abstract
Polyetheretherketone (PEEK) has emerged as a promising material for bone substitution; however, its limited osseointegration hinders its clinical applications. This research employs a porous structural design alongside surface modification techniques to improve the osseointegration properties of PEEK. Porous PEEK scaffolds were fabricated via 3D printing technology, followed by the application of a stable coating of Yoda1-loaded methacrylated gelatin (GelMA) on their surfaces. After modification, the hydrophilicity of the PEEK scaffolds was considerably improved, and the GelMA coating facilitated the sustained release of Yoda1. In vitro experiments demonstrated that the modified surfaces promoted cell proliferation and adhesion, facilitated angiogenesis, and enhanced osteoblast differentiation and mineral deposition. Furthermore, porous PEEK scaffolds were implanted into the femoral condyles of SD rats for 6 weeks to evaluate in vivo osseointegration The results showed that the tailored three-dimensional porous structure, along with the Yoda1-loaded GelMA coating, promoted bone ingrowth and enhanced osseointegration within the scaffold. This study offers a viable strategy for enhancing PEEK osseointegration through a combination of structural design and surface modification, and introduces new avenues for the application of Yoda1 in bone tissue engineering.
Collapse
Affiliation(s)
- Lei Wang
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Luxing Wei
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education School of Mechanical Engineering Shandong University, Jinan, Shandong 250061, China
| | - Jianlu Wei
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan 250000, China
| | - Yanbin Zhu
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Haiyue Zhao
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Yiran Zhang
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Shuo Zhang
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Pengfei Zhang
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan 250000, China
| | - Xin Xing
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Dengying Wu
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China; Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Fengkun Wang
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Shuai Yang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China; Hebei Medical University, Shijiazhuang 050017, China
| | - Yonglong Li
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Jun Huang
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High Efficiency and Clean Mechanical Manufacture of Ministry of Education School of Mechanical Engineering Shandong University, Jinan, Shandong 250061, China.
| | - Yunpeng Zhao
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan 250000, China.
| | - Yingze Zhang
- School of Medicine, Nankai University, Tianjin 300071, China; Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China.
| |
Collapse
|
11
|
Han D, Ding L, Zheng X, Li S, Yan H, Liu J, Wang H. Proteomics and lipidomics of human umbilical cord mesenchymal stem cells exposed to ionizing radiation. Eur J Med Res 2025; 30:340. [PMID: 40296080 PMCID: PMC12036141 DOI: 10.1186/s40001-025-02578-4] [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: 03/08/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
OBJECTIVES Mesenchymal stem cell (MSC)-based therapies exhibit beneficial effects on various forms of tissue damage, including ionizing radiation-induced lesions. However, whether ionizing radiation affects the functions of human umbilical cord mesenchymal stem cells (hucMSCs) remains unclear. This study aimed to investigate the effect and possible mechanisms of ionizing radiation on the proliferation and differentiation of hucMSCs. METHODS The hucMSCs were divided into the 1 Gy group (exposure to a single dose (1 Gy) of X-ray radiation (1 Gy/min) for 14 days) and control (without radiation treatment) group. The proliferation, apoptosis, and adipogenic and osteogenic differentiation abilities of hucMSCs in the two groups were evaluated. Moreover, the lipidomics and proteomics analyses were conducted to explore crucial lipids and proteins by which ionizing radiation affected the functions of hucMSCs. In addition, the effects of BYSL on radiation-treated hucMSCs were explore, as well as the involved potential mechanisms. RESULTS X-ray radiation treatment inhibited proliferation, promoted apoptosis, and decreased adipogenic and osteogenic differentiation abilities of hucMSCs. Key lipids, such as triglyceride (TG) and phosphatidylcholine (PC), and hub proteins (BYSL, MRTO4, and RRP9) exhibited significant differences between the 1 Gy group and control group. Moreover, BYSL, MRTO4, and RRP9 were significantly correlated with TG and PC. BYSL overexpression evidently promoted the cell proliferation, adipogenic and osteogenic differentiation abilities of radiation-treated hucMSCs, as well as the protein expression levels of p-GSK-3β/GSK-3β and β-catenin, while suppressed cell apoptosis. However, the GSK-3β inhibitor (1-Az) treatment reversed the protein expression levels of p-GSK-3β/GSK-3β, β-catenin and BYSL, as well as the cell proliferation, apoptosis, adipogenic and osteogenic differentiation abilities of radiation-treated hucMSCs. CONCLUSIONS Our findings reveal that the proliferation and differentiation of hucMSCs are suppressed by radiation, which may be associated with the changes of key lipids (TG and PC) and proteins (BYSL, MRTO4, and RRP9). Furthermore, BYSL promotes adipogenic and osteogenic differentiation abilities of radiation-treated hucMSCs via GSK-3β/β-catenin pathway. These findings help explain the response of hucMSCs to radiation and have clinical implications for improving the outcomes of MSC-based therapies after radiotherapy.
Collapse
Affiliation(s)
- Dongmei Han
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China
| | - Li Ding
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China
| | - Xiaoli Zheng
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China
| | - Sheng Li
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China
| | - Hongmin Yan
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China
| | - Jing Liu
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China
| | - Hengxiang Wang
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China.
| |
Collapse
|
12
|
Rao J, Xia L, Li Q, Ma N, Li X, Li J, Zhu L, Zhao P, Zeng Y, Zhou S, Guo H, Lin S, Dong S, Lou S, Fan F, Wei J, Zhong JF, Gao L, Li SC, Zhang X. A 6-tsRNA signature for early detection, treatment response monitoring, and prognosis prediction in diffuse large B cell lymphoma. Blood Cancer J 2025; 15:79. [PMID: 40295511 PMCID: PMC12037784 DOI: 10.1038/s41408-025-01267-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: 09/30/2024] [Revised: 03/14/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) presents considerable clinical challenges due to its aggressive nature and diverse clinical progression. New molecular biomarkers are urgently needed for outcome prediction. We analyzed blood samples from DLBCL patients and healthy individuals using short, non-coding RNA sequencing. A classifier based on six tsRNAs was developed through random forest and primary component analysis. This classifier, established using Cox proportional hazards modeling with repeated 10-fold cross-validation on an internal cohort of 100 samples analyzed using RT-qPCR, effectively identified high-risk patients with significantly lower overall survival compared to low-risk patients (Hazard ratio: 6.657, 95%CI 2.827-15.68, P = 0.0006). Validation in an external cohort of 160 samples using RT-qPCR confirmed the classifier's robust performance. High-risk status was strongly associated with disease histological subtype, stage, and International Prognostic Index scores. Integration of the classifier into the IPI model enhanced the precision and consistency of prognostic predictions. A dynamic study revealed that patients experiencing a 1.06-fold decrease after one therapy cycle (early molecular response) exhibited better treatment outcomes and prognosis. Furthermore, the 6-tsRNA signature accurately differentiated healthy individuals from DLBCL (AUC 0.882, 95%CI 0.826-0.939). These findings underscore the potential of the identified 6-tsRNA profile as a biomarker for monitoring treatment effectiveness and predicting DLBCL outcomes.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Prognosis
- Male
- Female
- Middle Aged
- Aged
- Biomarkers, Tumor/genetics
- Adult
- Aged, 80 and over
- Gene Expression Profiling
- Early Detection of Cancer
Collapse
Affiliation(s)
- Jun Rao
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Lin Xia
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Qiong Li
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - NaYa Ma
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Xinlei Li
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Jiali Li
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Lidan Zhu
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Pan Zhao
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Yunjing Zeng
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Sha Zhou
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Huanping Guo
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Shijia Lin
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Song Dong
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Shifeng Lou
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fangyi Fan
- Department of Hematology, General Hospital of Chengdu Military Region, Chengdu, Chongqing, China
| | - Jin Wei
- Department of Hematology, North Sichuan Medical College, Nanchong, China
| | - Jiang F Zhong
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, CA, USA
| | - Li Gao
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China
| | - Shengwen Calvin Li
- CHOC Children's Research Institute, Children's Hospital of Orange County (CHOC®), part of Rady Children's Heath, Orange, CA, USA.
- Department of Neurology, University of California-Irvine School of Medicine, Orange, CA, USA.
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, China.
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China.
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Soochow, China.
- Jinfeng Laboratory, Chongqing, China.
| |
Collapse
|
13
|
Wang C, Luo S, Yan Y, Li J, Niu W, Hong T, Hao K, Sun X, Liu J, An R, Li J. Endothelial Piezo1 stimulates angiogenesis to offer protection against intestinal ischemia-reperfusion injury in mice. Mol Med 2025; 31:147. [PMID: 40263994 PMCID: PMC12016420 DOI: 10.1186/s10020-025-01197-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Accepted: 04/03/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Intestinal ischemia-reperfusion (I/R) injury, which occurs in the ileum and not only leads to intestinal tissue damage, but also may trigger systemic inflammatory responses, is a prevalent pathological condition that is typically associated with acute intestinal ischemia, surgical procedures, or trauma. However, the precise underlying pathogenic mechanisms have not yet been fully uncovered. In this study, we explored the specific roles and underlying mechanisms by which endothelial Piezo1 is involved in intestinal I/R injury. METHODS We evaluated the roles of Piezo1 using both in vivo mouse intestinal ischemia-reperfusion (I/R) injury and in vitro hypoxia-reoxygenation (H/R) models. The expression of Piezo1 was assessed using immunofluorescence and RT-qPCR. In vivo and in vitro experiments involving endothelial knockout and activation of Piezo1 with the specific agonist Yoda1 were conducted to observe the effects on angiogenesis and injury. RESULTS We found that in post-intestinal I/R mice, Piezo1 expression was markedly increased and was mainly abundant in ileum endothelial cells. Specific knockout of endothelial Piezo1 exhibited a more severe phenotype characterized by accelerating damage to the ileum structure, increasing inflammatory response, and inhibiting angiogenesis. Yoda1-mediated activation of Piezo1 significantly ameliorated intestinal I/R injury. Activation of Piezo1 induced by Yoda1 or H/R promoted angiogenesis in Human Umbilical Vein Endothelial Cells (HUVECs), which was inhibited by GsMTx4. Piezo1 mediated endothelial angiogenesis was linked to an increase of extracellular Ca2+ influx, which in turn enhanced hypoxia-inducible factor 1 alpha (HIF-1α) signaling pathway. CONCLUSIONS Our findings indicate that Piezo1 plays a crucial role in protecting against intestinal I/R injury by promoting angiogenesis in endothelial cells, possibly through the activation of the Ca2+/HIF-1α/VEGF signaling pathway. This suggests that targeting endothelial Piezo1 channels could be a therapeutic strategy for ileum I/R injury.
Collapse
Affiliation(s)
- Cuifen Wang
- Innovation Research Center, Shandong University of Chinese Medicine, Jinan, 250307, China
| | - Shangfei Luo
- Innovation Research Center, Shandong University of Chinese Medicine, Jinan, 250307, China
| | - Yameng Yan
- Innovation Research Center, Shandong University of Chinese Medicine, Jinan, 250307, China
| | - Jinze Li
- Innovation Research Center, Shandong University of Chinese Medicine, Jinan, 250307, China
| | - Weipin Niu
- Innovation Research Center, Shandong University of Chinese Medicine, Jinan, 250307, China
| | - Tianying Hong
- Innovation Research Center, Shandong University of Chinese Medicine, Jinan, 250307, China
| | - Kai Hao
- Innovation Research Center, Shandong University of Chinese Medicine, Jinan, 250307, China
| | - Xin Sun
- Innovation Research Center, Shandong University of Chinese Medicine, Jinan, 250307, China
| | - Jiali Liu
- Innovation Research Center, Shandong University of Chinese Medicine, Jinan, 250307, China
| | - Ran An
- Pacific College of Health and Science, 110 William St 19 th Floor, New York, NY, 10038, USA
| | - Jing Li
- Innovation Research Center, Shandong University of Chinese Medicine, Jinan, 250307, China.
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
14
|
Javier López Rivera J, Gomez-Lopera N, Moreno-Garcia DJ, Orduz-Rodriguez R, Combariza-Vallejo JF, Isaza-Ruget M. Plasma Cell Enrichment and New Genomic Approaches in Multiple Myeloma: A Scoping Review. J Appl Lab Med 2025:jfaf044. [PMID: 40248905 DOI: 10.1093/jalm/jfaf044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/12/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND Multiple myeloma (MM) is a genetically heterogeneous disease where specific genetic abnormalities have a significant impact on a patient's prognosis. Diagnostic and prognostic tools like fluorescence in situ hybridization (FISH), PCR, microarrays, and next-generation sequencing (NGS) have transformed MM management. However, the effectiveness of these techniques is often limited by the low concentration of plasma cells in bone marrow samples, which makes enrichment methods necessary. This review aims to clarify how these techniques enhance the detection of genetic abnormalities, reduce false-negative results, and facilitate more precise risk stratification for MM patients. CONTENT Following Preferred Reporting Items for Systematic reviews and Meta-Analyses Extension for Scoping Review (PRISMA-ScR) guidelines, the literature on plasma cell separation methods used in genetic studies of MM was systematically identified and mapped. Searches were conducted in the Medline and Embase databases using a structured strategy, supplemented by manual searches on Google Scholar. Of 399 publications evaluated, 69 met the inclusion criteria; 37% utilized FISH and 19% demonstrated an increasing use of NGS. Plasma cell enrichment significantly improved diagnostic accuracy, increasing the detection rates of genetic abnormalities from 61% in non-enriched samples to 95.5% in enriched samples. While FISH remains the gold standard, emerging technologies such as NGS offer superior sensitivity and the ability to identify critical genetic alterations to refine molecular subtypes. SUMMARY Clinically significant genetic alterations are detected more frequently with plasma cell enrichment techniques, contributing to improved prognosis and treatment strategies for MM patients.
Collapse
Affiliation(s)
- Juan Javier López Rivera
- Laboratorio Especializado en Biología Molecular, Clínica Colsanitas, Grupo Keralty, Bogotá, Colombia
- Grupo de Genética Médica, Clínica Universitaria Colombia, Clínica Colsanitas, Grupo Keralty, Bogotá, Colombia
| | - Natalia Gomez-Lopera
- Laboratorio Clínico y de Patología, Clínica Colsanitas, Grupo Keralty, Bogotá, Colombia
| | | | - Rocío Orduz-Rodriguez
- Laboratorio Clínico y de Patología, Clínica Colsanitas, Grupo Keralty, Bogotá, Colombia
| | - Juan F Combariza-Vallejo
- Servicio de Hematología, Clínica Universitaria Colombia, Clínica Colsanitas S.A., Grupo Keralty, Bogotá, Colombia
| | - Mario Isaza-Ruget
- Laboratorio Clínico y de Patología, Clínica Colsanitas, Grupo Keralty, Bogotá, Colombia
- Unidad de Investigación, Fundación Universitaria Sanitas, Grupo de investigación INPAC, Grupo Keralty, Bogotá, Colombia
| |
Collapse
|
15
|
Gao Y, Tillu VA, Wu Y, Rae J, Hall TE, Chen KE, Weeratunga S, Guo Q, Livingstone E, Tham WH, Parton RG, Collins BM. Nanobodies against Cavin1 reveal structural flexibility and regulated interactions of its N-terminal coiled-coil domain. J Cell Sci 2025; 138:jcs263756. [PMID: 40260863 DOI: 10.1242/jcs.263756] [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: 03/06/2025] [Indexed: 04/24/2025] Open
Abstract
Caveolae are abundant plasma membrane structures that regulate signalling, membrane homeostasis and mechanoprotection. Their formation is driven by caveolins and cavins and their coordinated interactions with lipids. Here, we developed nanobodies against the trimeric HR1 coiled-coil domain of Cavin1. We identified specific nanobodies that do not perturb Cavin1 membrane binding and localise to caveolae when expressed in cells. The crystal structure of a nanobody-Cavin 1 HR1 complex reveals a symmetric 3:3 architecture as validated by mutagenesis. In this structure, the C-terminal half of the HR1 domain is disordered, suggesting that the nanobody stabilises an open conformation of Cavin1, which has previously been identified as important for membrane interactions. A phosphomimic mutation in a threonine-serine pair proximal to this region reveals selective regulation of Cavin2 and Cavin3 association. These studies provide new insights into cavin domains required for assembly of multiprotein caveolar assemblies and describe new nanobody tools for structural and functional studies of caveolae.
Collapse
Affiliation(s)
- Ya Gao
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Vikas A Tillu
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Yeping Wu
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - James Rae
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Thomas E Hall
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Kai-En Chen
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Saroja Weeratunga
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Qian Guo
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Emma Livingstone
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Wai-Hong Tham
- Infectious Diseases and Immune Defence Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
- Centre for Microscopy and Microanalysis, The University of Queensland, St. Lucia, Queensland 4067, Australia
| | - Brett M Collins
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4067, Australia
| |
Collapse
|
16
|
Zhao B, Wu J, Zhang T, Han M, Zhang C, Rong X, Zhang R, Chen X, Peng F, Jin J, Liu S, Dong X, Zhao S. A spatial transcriptomics study of MES-like and mono/macro cells in gliomas. Sci Rep 2025; 15:12730. [PMID: 40222970 PMCID: PMC11994772 DOI: 10.1038/s41598-025-95277-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Gliomas, including both glioblastoma multiforme (GBM) and lower-grade glioma (LGG), present a substantial challenge in neuro-oncology because of genetic heterogeneity and unsatisfactory prognosis. This study aimed to conduct a comprehensive multi-omics analysis of gliomas using various bioinformatics approaches to identify potential therapeutic targets and prognostic markers. A comprehensive analysis was conducted on 1327 sequencing data samples alongside their relevant clinical information sourced from The Cancer Genome Atlas (TCGA) pertaining to glioblastoma (GBM), low-grade glioma (LGG), the Chinese Glioma Genome Atlas (CCGA) and University of California Santa Cruz Xena (UCSC Xena) datasets. These tools were employed for gene expression profiling, survival analysis, and cell communication mapping. Spatial transcriptomics revealed the localization of mesenchymal (MES)-like malignant tumors, and drug sensitivity analysis was performed to evaluate responses to quinpirole and meropenem. Additionally, the Tumor Immune Dysfunction and Exclusion (TIDE) framework was utilized to gauge the responsiveness to immunotherapy. The MES-like malignant and monocyte/macrophage (mono/macro) cell subsets showed high hallmark scores, playing key roles in the tumor microenvironment. MES-like malignant marker gene scores correlated with overall survival across datasets, whereas mono/macro marker gene scores were significant in the TCGA-LGG and CCGA datasets. Key interactions between these cell types were found, especially with CD14-ITGB2, LGALS1-CD69, and APOE-TREM2. The mono/macro cell subset demonstrated better immune therapy responsiveness, as indicated by lower TIDE scores. Spatial transcriptomics revealed that MES-like malignant tumors are predominantly localized in four distinct regions, with the marker genes CHI3L1 and ADM confirming these locations. Drug sensitivity analysis revealed differential responses of the MES-like malignant cell subset to quinpirole and meropenem. Our results offer fresh perspectives on the differential roles of MES-like malignant and monocyte/macrophage cell subsets in tumor progression and immune modulation, providing novel insights into glioma biology.
Collapse
Affiliation(s)
- Boyan Zhao
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China
| | - Jianing Wu
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
| | - Tiehui Zhang
- Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, 518104, Guangdong, China
| | - Mingyang Han
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
| | - Cheng Zhang
- University of Toronto Scarborough 1265 Military Trail, Scarborough, ON, M1C 1A4, Canada
| | - Xuan Rong
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
| | - Ruotian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xin Chen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Neurosurgery of Colleges and Universities in Heilongjiang Province, Harbin, 150001, Heilongjiang, China
| | - Fei Peng
- Department of Neurosurgery and Neurosurgical Disease Research Centre, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jin Jin
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China
| | - Shiya Liu
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China
| | - Xingli Dong
- Central Laboratory, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China.
| | - Shiguang Zhao
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China.
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China.
- Department of Neurosurgery, Shenzhen University General Hospital, 1088 Xueyuan Avenue, Nanshan District, Shenzhen, 518036, Guangdong, China.
| |
Collapse
|
17
|
Komemi O, Orbuch E, Jarchowsky-Dolberg O, Brin YS, Tartakover-Matalon S, Pasmanik-Chor M, Lishner M, Drucker L. Myeloma mesenchymal stem cells' bioenergetics afford a novel selective therapeutic target. Oncogenesis 2025; 14:9. [PMID: 40216736 PMCID: PMC11992228 DOI: 10.1038/s41389-025-00554-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 03/17/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
Bone-marrow mesenchymal stem cells (BM-MSCs) rely on glycolysis, yet their trafficked mitochondria benefit recipient cells' bioenergetics in regenerative and cancerous settings, most relevant to BM-resident multiple myeloma (MM) cells. Fission/fusion dynamics regulate mitochondria function. Proteomics demonstrates excessive mitochondrial processes in BM-MSCs from MM patients compared to normal donors (ND). Thus, we aimed to characterize BM-MSCs (ND, MM) mitochondrial fitness, bioenergetics and dynamics with a focus on therapeutics. MM-MSCs displayed compromised mitochondria evidenced by decreased mitochondrial membrane potential (ΔΨm) and elevated proton leak. This was accompanied by stimulation of stress-coping mechanisms: spare respiratory capacity (SRC), mitochondrial fusion and UPRmt. Interfering with BM-MSCs mitochondrial dynamics equilibrium demonstrated their significance to bioenergetics and fitness according to the source. While ND-MSCs depended on fission, reducing MM-MSCs fusion attenuated glycolysis, OXPHOS and mtROS. Interestingly, optimization of mtROS levels is central to ΔΨm preservation in MM-MSCs only. MM-MSCs also demonstrated STAT3 activation, which regulates their OXPHOS and SRC. Targeting MM-MSC' SRC with Venetoclax diminished their pro-MM support and sensitized co-cultured MM cells to Bortezomib. Overall, MM-MSCs distinct mitochondrial bioenergetics are integral to their robustness. Repurposing Venetoclax as anti-SRC treatment in combination with conventional anti-MM drugs presents a potential selective way to target MM-MSCs conferred drug resistance.
Collapse
Affiliation(s)
- Oded Komemi
- Oncogenetic Laboratory, Meir Medical Center, Kfar Saba, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Elina Orbuch
- Oncogenetic Laboratory, Meir Medical Center, Kfar Saba, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Osnat Jarchowsky-Dolberg
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Internal Medicine A, Meir Medical Center, Kfar Saba, Israel
- Hematology Unit, Meir Medical Center, Kfar Saba, Israel
| | | | - Shelly Tartakover-Matalon
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Autoimmunity Laboratory, Meir Medical Center, Kfar Saba, Israel
| | - Metsada Pasmanik-Chor
- Bioinformatics Unit, G.S.W., Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Michael Lishner
- Oncogenetic Laboratory, Meir Medical Center, Kfar Saba, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Hematology Unit, Meir Medical Center, Kfar Saba, Israel
| | - Liat Drucker
- Oncogenetic Laboratory, Meir Medical Center, Kfar Saba, Israel.
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
18
|
Xu C, Chen L, Liu G, Xu J, Lv W, Gao X, Xu P, Tang M, Wang Y, Zhao X, Nie G, Cheng K, Liu F. Tailoring an intravenously injectable oncolytic virus for augmenting radiotherapy. Cell Rep Med 2025:102078. [PMID: 40233744 DOI: 10.1016/j.xcrm.2025.102078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/19/2024] [Accepted: 03/18/2025] [Indexed: 04/17/2025]
Abstract
Oncolytic viruses (OVs) combined with radiotherapy (RT) show promise but are limited by challenges such as poor intravenous delivery and insufficient RT-induced DNA damage. In this study, an oncolytic adenovirus (AD) formulation, RadioOnco (AD@PSSP), is developed to improve delivery, infectivity, immune response, and RT efficacy. The multifunctional polyethylenimine (PEI)-selenium-polyethylene glycol (PEG) (PSSP) enhances intravenous delivery, shields the virus from rapid clearance, and enables targeted delivery to tumor sites after RT. The exposed PEI enhances the infectivity of AD through electrostatic interactions, thereby increasing DNA damage after RT by inhibiting the expression of DNA repair proteins, such as CHEK1 and CDK1. Furthermore, AD-PEI captures and delivers RT-induced tumor-released antigens to lymph nodes, activating robust anti-tumor immune responses. Animal model data demonstrate that RadioOnco overcomes RT resistance, targets distant metastases, and promotes long-term immunity, addressing metastasis and recurrence. In summary, this intravenously injectable OV enhances RT synergy through surface modification with multifunctional materials.
Collapse
Affiliation(s)
- Chen Xu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China; Phase I Clinical Trails Center, The First Hospital of China Medical University, Shenyang 110102, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Liting Chen
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China; Phase I Clinical Trails Center, The First Hospital of China Medical University, Shenyang 110102, China
| | - Guangna Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jiaqi Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wei Lv
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaoyu Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Peijun Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ming Tang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yaohe Wang
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China; IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Funan Liu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China; Phase I Clinical Trails Center, The First Hospital of China Medical University, Shenyang 110102, China.
| |
Collapse
|
19
|
Chintalaramulu N, Singh DP, Sapkota B, Raman D, Alahari S, Francis J. Caveolin-1: an ambiguous entity in breast cancer. Mol Cancer 2025; 24:109. [PMID: 40197489 PMCID: PMC11974173 DOI: 10.1186/s12943-025-02297-8] [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: 01/08/2025] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Breast cancer (BC) is the most frequently diagnosed cancer in women and the second leading cause of death from cancer among women. Metastasis is the major cause of BC-associated mortality. Accumulating evidence implicates Caveolin-1 (Cav-1), a structural protein of plasma membrane caveolae, in BC metastasis. Cav-1 exhibits a dual role, as both a tumor suppressor and promoter depending on the cellular context and BC subtype. This review highlights the role of Cav-1 in modulating glycolytic metabolism, tumor-stromal interactions, apoptosis, and senescence. Additionally, stromal Cav-1's expression is identified as a potential prognostic marker, offering insights into its contrasting roles in tumor suppression and progression. Furthermore, Cav-1's context-dependent effects are explored in BC subtypes including hormone receptor-positive, HER2-positive, and triple-negative BC (TNBC). The review further delves into the role of Cav-1 in regulating the metastatic cascade including extracellular matrix interactions, cell migration and invasion, and premetastatic niche formation. The later sections discuss the therapeutic targeting of Cav-1 by metabolic inhibitors such as betulinic acid and Cav-1 modulating compounds. While Cav-1 may be a potential biomarker and therapeutic target, its heterogeneous expression and context-specific activity necessitates further research to develop precise interventions. Future studies investigating the mechanistic role of Cav-1 in metastasis may pave the way for effective treatment of metastatic BC.
Collapse
Affiliation(s)
- Naveen Chintalaramulu
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | | | - Biplov Sapkota
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Dayanidhi Raman
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH, USA
| | | | - Joseph Francis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
20
|
Gentile R, Feudi D, Sallicandro L, Biagini A. Can the Tumor Microenvironment Alter Ion Channels? Unraveling Their Role in Cancer. Cancers (Basel) 2025; 17:1244. [PMID: 40227837 PMCID: PMC11988140 DOI: 10.3390/cancers17071244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 03/29/2025] [Accepted: 04/03/2025] [Indexed: 04/15/2025] Open
Abstract
Neoplastic cells are characterized by metabolic reprogramming, known as the Warburg effect, in which glucose metabolism is predominantly directed toward aerobic glycolysis, with reduced mitochondrial oxidative phosphorylation and increased lactate production even in the presence of oxygen. This phenomenon provides cancer cells with a proliferative advantage, allowing them to rapidly produce energy (in the form of ATP) and generate metabolic intermediates necessary for the biosynthesis of macromolecules essential for cell growth. It is important to understand the role of ion channels in the tumor context since they participate in various physiological processes and in the regulation of the tumor microenvironment. These changes may contribute to the development and transformation of cancer cells, as well as affect the communication between cells and the surrounding microenvironment, including impaired or altered expression and functionality of ion channels. Therefore, the aim of this review is to elucidate the impact of the tumor microenvironment on the electrical properties of the cellular membranes in several cancers as a possible therapeutic target.
Collapse
Affiliation(s)
- Rosaria Gentile
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06123 Perugia, Italy;
| | - Davide Feudi
- Department of Biostatistics, Epidemiology and Public Health, University of Padua, Via L. Loredan 18, 35131 Padova, Italy;
| | - Luana Sallicandro
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06123 Perugia, Italy;
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
| | - Andrea Biagini
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06123 Perugia, Italy;
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
| |
Collapse
|
21
|
Doktorova M, Daum S, Reagle TR, Cannon HI, Ebenhan J, Neudorf S, Han B, Sharma S, Kasson P, Levental KR, Bacia K, Kenworthy AK, Levental I. Caveolin assemblies displace one bilayer leaflet to organize and bend membranes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.28.610209. [PMID: 39257813 PMCID: PMC11383982 DOI: 10.1101/2024.08.28.610209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Caveolin is a monotopic integral membrane protein, widely expressed in metazoa and responsible for constructing enigmatic membrane invaginations known as caveolae. Recently, the high-resolution structure of a purified human caveolin assembly, the CAV1-8S complex, revealed a unique organization of 11 protomers arranged in a tightly packed, radially symmetric spiral disc. One face and the outer rim of this disc are hydrophobic, suggesting that the complex incorporates into membranes by displacing hundreds of lipids from one leaflet. The feasibility of this unique molecular architecture and its biophysical and functional consequences are currently unknown. Using Langmuir film balance measurements, we find that CAV1-8S is highly surface active, intercalating into lipid monolayers of various compositions. CAV1-8S can also incorporate into preformed bilayers, but only upon removal of phospholipids from the outer-facing leaflet. Atomistic and coarse-grained simulations of biomimetic bilayers support this 'leaflet replacement' model and also reveal that CAV1-8S accumulates 40-70 cholesterol molecules into a disordered monolayer between the complex and its distal lipid leaflet. We find that CAV1-8S preferentially associates with positively curved membrane surfaces due to its influence on the conformations of distal leaflet lipids, and that these effects laterally sort lipids. Large-scale simulations of multiple caveolin assemblies confirmed their association with large, positively curved membrane morphologies consistent with the shape of caveolae. Further, association with curved membranes regulates the exposure of caveolin residues implicated in protein-protein interactions. Altogether, the unique structure of CAV1-8S imparts unusual modes of membrane interaction with implications for membrane organization, morphology, and physiology.
Collapse
Affiliation(s)
- Milka Doktorova
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Sebastian Daum
- Department of Physical Chemistry, Martin Luther University Halle-Wittenberg, Germany
| | - Tyler R. Reagle
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA
| | - Hannah I. Cannon
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA
| | - Jan Ebenhan
- Department of Physical Chemistry, Martin Luther University Halle-Wittenberg, Germany
| | - Sarah Neudorf
- Department of Physical Chemistry, Martin Luther University Halle-Wittenberg, Germany
| | - Bing Han
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA
| | - Satyan Sharma
- Department of Cell and Molecular Biology, Uppsala University, Sweden
| | - Peter Kasson
- Department of Cell and Molecular Biology, Uppsala University, Sweden
- Departments of Chemistry and Biochemistry and Biomedical Engineering, Georgia Institute of Technology, USA
| | - Kandice R. Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA
| | - Kirsten Bacia
- Department of Physical Chemistry, Martin Luther University Halle-Wittenberg, Germany
| | - Anne K. Kenworthy
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, USA
| |
Collapse
|
22
|
Ibrahim MM, Azmi MN, Alhawarri MB, Kamal NNSNM, AbuMahmoud H. Synthesis, characterization and bioactivity of new pyridine-2(H)-one, nicotinonitrile, and furo[2,3-b]pyridine derivatives. Mol Divers 2025; 29:1569-1587. [PMID: 39009909 DOI: 10.1007/s11030-024-10934-5] [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: 05/05/2024] [Accepted: 07/09/2024] [Indexed: 07/17/2024]
Abstract
Pyridone heterocycles, such as furo[2,3-b]pyridines, have emerged as prominent scaffolds in medicinal chemistry due to their versatile pharmacological properties, including significant anticancer activity. In this study, we successfully synthesized new pyridine-2(H)-one, nicotinonitrile, and furo[2,3-b]pyridine derivatives from chalcones bearing 4-(benzyloxy)phenyl and dichlorothiophenyl subunits to explore their therapeutic potential against breast cancer. By employing a synthetic strategy involving Claisen-Schmidt condensation followed by sequential cyclizations and functional modifications, we synthesized and characterized four compounds (MI-S0, MI-S1, MI-S2, and MI-S3) using various spectroscopic methods, including FT-IR, 1H-NMR, 13C-NMR, DEPT, H,H- and C,H-COSY, and HRMS. The in vitro cytotoxic activity of these compounds was evaluated against two breast cancer cell lines, MCF-7 and MDA-MB-231, and compared with a noncancerous breast cell line, MCF-10A. All compounds exhibited potent cytotoxic activities with minimal selectivity toward normal cells. Molecular docking studies targeting the serine/threonine kinase AKT1, estrogen receptor alpha (ERα), and human epidermal growth factor receptor 2 (HER2) revealed strong binding affinities, suggesting a mechanism involving the disruption of key cellular signaling pathways. These findings underscore the potential of furo[2,3-b]pyridine derivatives as promising candidates for further development into anticancer agents, laying the groundwork for future investigations into their selective therapeutic efficacy and molecular mechanisms of action.
Collapse
Affiliation(s)
- Mohammad M Ibrahim
- Department of Chemistry, Faculty of Science, Al Al-Bayt University, P.O. BOX 130040, Al-Mafraq, 25113, Jordan.
| | - Mohamad Nurul Azmi
- School of Chemical Sciences, Universiti Sains Malaysia, 11800, Minden, Pulau Pinang, Malaysia
| | - Maram B Alhawarri
- Department of Pharmacy, Faculty of Pharmacy, Jadara University, P.O.Box 733, Irbid, 21110, Jordan
| | - Nik Nur Syazni Nik Mohamed Kamal
- Department of Toxicology, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200, Kepala Batas, Penang, Malaysia
| | - Hasan AbuMahmoud
- Department of Chemistry, Faculty of Science, Al Al-Bayt University, P.O. BOX 130040, Al-Mafraq, 25113, Jordan
| |
Collapse
|
23
|
Poluektov YM, Lopina OD, Strelkova MA, Kuleshova ID, Makarov AA, Petrushanko IY. Mechanisms mediating effects of cardiotonic steroids in mammalian blood cells. Front Pharmacol 2025; 16:1520927. [PMID: 40196366 PMCID: PMC11973394 DOI: 10.3389/fphar.2025.1520927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/21/2025] [Indexed: 04/09/2025] Open
Abstract
Cardiotonic steroids (CTSs) were known as steroidal plant compounds that exert cellular effects by the binding to Na,K-ATPase. Earlier, plant (exogenous) CTSs were used to treat chronic heart failure. By now, endogenous CTS have been identified in mammals, and their concentrations in the blood, normally in a subnanomolar range, are altered in numerous pathologies. This indicates their role as endogenous regulators of physiological processes. CTS transport occurs primarily in the blood, yet the CTS effects on blood cells remain poorly understood. This review summarizes the CTS effects on blood cells of animals and humans under normal and pathological conditions, and analyzes their action based on known mechanisms of action in mammalian cells. At high concentrations (greater than 10-9 M), CTS binding to Na,K-ATPase inhibits the enzyme, whereas lower concentrations of CTSs induce signaling cascades or activate the enzyme. All these mechanisms are shown to be present in blood cells. The particular CTS effect is determined by the CTS type, its concentration, the isoform composition of the catalytic α-subunit of Na,K-ATPase in the cell, and other cell features. It has been demonstrated that all blood cell types (erythrocytes, leukocytes, and platelets) expressed both ubiquitously distributed α1-isoform and tissue-specific α3-subunit, which exhibits a different ion and CTS affinity compared to α1. This results in a wide spectrum of blood cell responses to fluctuations in CTS levels in the blood. In particular, an increase in the level of endogenous CTSs by a more twofold is sufficient to induce a decline in the activity of erythrocyte Na,K-ATPase. The administration of exogenous CTSs is able to modulate the proinflammatory activity of leukocytes, which is attributed to the activation of signaling cascades, and to exert an influence on platelet activation. Hence, alterations of CTS levels in bloodstream significantly affect the functionality of blood cells, contributing to the organism's adaptive response. On top of this, a comparison of the effects of CTSs on human leukocytes and rodent leukocytes carrying the CTS-resistant α1-isoform often reveals opposite effects, thus indicating that rodents are an unsuitable model for studying CTS effects on these cells.
Collapse
Affiliation(s)
- Yuri M. Poluektov
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Olga D. Lopina
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Maria A. Strelkova
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Iuliia D. Kuleshova
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Alexander A. Makarov
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| | - Irina Yu. Petrushanko
- Engelhardt Institute of Molecular Biology Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
24
|
Stea DM, D’Alessio A. Caveolae: Metabolic Platforms at the Crossroads of Health and Disease. Int J Mol Sci 2025; 26:2918. [PMID: 40243482 PMCID: PMC11988808 DOI: 10.3390/ijms26072918] [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: 02/27/2025] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Caveolae are small flask-shaped invaginations of the plasma membrane enriched in cholesterol and sphingolipids. They play a critical role in various cellular processes, including signal transduction, endocytosis, and mechanotransduction. Caveolin proteins, specifically Cav-1, Cav-2, and Cav-3, in addition to their role as structural components of caveolae, have been found to regulate the activity of signaling molecules. A growing body of research has highlighted the pivotal role of caveolae and caveolins in maintaining cellular metabolic homeostasis. Indeed, studies have demonstrated that caveolins interact with the key components of insulin signaling, glucose uptake, and lipid metabolism, thereby influencing energy production and storage. The dysfunction of caveolae or the altered expression of caveolins has been associated with metabolic disorders, including obesity, type 2 diabetes, and ocular diseases. Remarkably, mutations in caveolin genes can disrupt cellular energy balance, promote oxidative stress, and exacerbate metabolic dysregulation. This review examines current research on the molecular mechanisms through which caveolae and caveolins regulate cellular metabolism, explores their involvement in the pathogenesis of metabolic disorders, and discusses potential therapeutic strategies targeting caveolin function and the stabilization of caveolae to restore metabolic homeostasis.
Collapse
Affiliation(s)
- Dante Maria Stea
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Alessio D’Alessio
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, 00168 Rome, Italy
| |
Collapse
|
25
|
Zhang Y, Li J, Peng D, Jiang L, Cheng S, Xiang H, Ni R, Li L, Long S, Li Z, Song L, Xie H, Liu Y. Integrating metabolomics and network pharmacology to explore the mechanism of Xiangshao Sanjie Oral Liquid in treating rats with mammary gland hyperplasia. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119493. [PMID: 39961425 DOI: 10.1016/j.jep.2025.119493] [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: 12/02/2024] [Revised: 01/24/2025] [Accepted: 02/11/2025] [Indexed: 02/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xiangshao Sanjie Oral Liquid (XSSJ) is a traditional Chinese medicine formulation used clinically for the treatment of mammary gland hyperplasia (MGH), yet its mechanism remains obscure. AIM OF THE STUDY The purpose of this study was to explore the therapeutic mechanism of XSSJ on MGH using a comprehensive strategy of plasma metabolomics and network pharmacology. MATERIALS AND METHODS The rat model of MGH was established, and the multiple indicators were employed for efficacy evaluation. Then, a metabolomics strategy was established to find plasma metabolites and metabolic pathways that may be important in inducing MGH. In addition, UPLC-Q-TOF-MS and network pharmacological analysis were used to identify the prototype compounds of XSSJ in rat plasma and target genes that may cause the effect. Finally, the results were integrated and verified by RT-qPCR and Western Blot (WB). RESULTS XSSJ has a therapeutic effect on MGH. 29 differential metabolites of XSSJ in the treatment of MGH were identified by metabolomics. After administration of XSSJ, 16 prototype compounds were found in the rat plasma, which were associated with 179 potential therapeutic targets. Comprehensive analysis revealed that XSSJ reversed the mRNA expression of EGFR, ESR1, AKT1, SRC and PTPN11 in MGH rats. In addition, different doses of XSSJ inhibited the expression of p-PI3K and p-AKT proteins. CONCLUSION This study combined metabolomics and network pharmacology to reveal the regulatory effect of XSSJ on MGH through PI3K/AKT pathway, which provided further support for the clinical application of XSSJ.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China; School of Medicine, Tibet University, Lhasa, China
| | - Jiaming Li
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Dan Peng
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Lining Jiang
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Shiqi Cheng
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China; Medical Research Institute, Southwest University, Chongqing, China
| | - Hong Xiang
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Rui Ni
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Li Li
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Shilin Long
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Ziwei Li
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Lin Song
- Department of Pharmacy, Children's Hospital of Chongqing Medical University, National Clinical Research Centre for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Clinical Pharmacy Key Specialty Construction Project, Chongqing, China
| | - Hongjun Xie
- School of Medicine, Tibet University, Lhasa, China
| | - Yao Liu
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China; School of Medicine, Tibet University, Lhasa, China.
| |
Collapse
|
26
|
Guan F, Wang R, Yi Z, Luo P, Liu W, Xie Y, Liu Z, Xia Z, Zhang H, Cheng Q. Tissue macrophages: origin, heterogenity, biological functions, diseases and therapeutic targets. Signal Transduct Target Ther 2025; 10:93. [PMID: 40055311 PMCID: PMC11889221 DOI: 10.1038/s41392-025-02124-y] [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] [Received: 07/21/2024] [Revised: 11/01/2024] [Accepted: 12/15/2024] [Indexed: 05/04/2025] Open
Abstract
Macrophages are immune cells belonging to the mononuclear phagocyte system. They play crucial roles in immune defense, surveillance, and homeostasis. This review systematically discusses the types of hematopoietic progenitors that give rise to macrophages, including primitive hematopoietic progenitors, erythro-myeloid progenitors, and hematopoietic stem cells. These progenitors have distinct genetic backgrounds and developmental processes. Accordingly, macrophages exhibit complex and diverse functions in the body, including phagocytosis and clearance of cellular debris, antigen presentation, and immune response, regulation of inflammation and cytokine production, tissue remodeling and repair, and multi-level regulatory signaling pathways/crosstalk involved in homeostasis and physiology. Besides, tumor-associated macrophages are a key component of the TME, exhibiting both anti-tumor and pro-tumor properties. Furthermore, the functional status of macrophages is closely linked to the development of various diseases, including cancer, autoimmune disorders, cardiovascular disease, neurodegenerative diseases, metabolic conditions, and trauma. Targeting macrophages has emerged as a promising therapeutic strategy in these contexts. Clinical trials of macrophage-based targeted drugs, macrophage-based immunotherapies, and nanoparticle-based therapy were comprehensively summarized. Potential challenges and future directions in targeting macrophages have also been discussed. Overall, our review highlights the significance of this versatile immune cell in human health and disease, which is expected to inform future research and clinical practice.
Collapse
Affiliation(s)
- Fan Guan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ruixuan Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wanyao Liu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yao Xie
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China.
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
27
|
Deng M, Yang R, Jiang J, Zhang J, He J, Miao J. The silent spread: exploring diverse metastatic pathways in high-grade serous ovarian cancer. Front Med (Lausanne) 2025; 12:1539024. [PMID: 40109727 PMCID: PMC11919666 DOI: 10.3389/fmed.2025.1539024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is a highly aggressive and deadly gynecological cancer, with metastasis being a key factor in its poor prognosis. Historically, HGSOC was thought to spread primarily through the peritoneal cavity, but recent research has revealed additional routes of metastasis, including the blood and lymphatic systems. This review discusses the complex processes of HGSOC metastasis, focusing on peritoneal immune suppression, stromal reprogramming, and the role of circulating tumor cells in blood-based spread. We also explore the clinical significance of lymphatic metastasis, particularly its impact on patient outcomes. Gaining insight into molecular and genetic drivers, such as BRCA mutations and interactions within the immune microenvironment, is essential for developing targeted treatments. Future studies should aim to enhance experimental models, identify early detection markers, and investigate novel therapeutic approaches to effectively address HGSOC metastasis and improve patient survival.
Collapse
Affiliation(s)
- Mengqi Deng
- Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Ruiye Yang
- Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Junyi Jiang
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Institute of Lifeomics, Beijing, China
| | - Jinxu Zhang
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Institute of Lifeomics, Beijing, China
| | - Junqi He
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
| | - Jinwei Miao
- Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Gunay A, Targum SD, Leow AD, Ajilore O, Rasenick MM. A simple platelet biomarker is associated with symptom severity in major depressive disorder. Mol Psychiatry 2025:10.1038/s41380-025-02941-1. [PMID: 40038544 DOI: 10.1038/s41380-025-02941-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 01/21/2025] [Accepted: 02/20/2025] [Indexed: 03/06/2025]
Abstract
Previous studies have shown that the heterotrimeric G protein, Gsalpha (Gsα), is ensconced predominantly in lipid rafts in acutely depressed subjects with major depressive disorder (MDD) in contrast to healthy controls, and that effective antidepressant treatment (ADT) facilitates translocation of Gsα from lipid rafts. The measurement of Gsα via prostaglandin E1 stimulation of adenylyl cyclase (PGE1 stimulation) has been proposed as a peripheral biomarker for assessing clinical status in MDD. We examined the Gsα biomarker in a new study. PGE1 stimulation was used to assess the coupling of Gsα with platelet adenylyl cyclase in depressed subjects in active treatment and healthy controls. The Quick Inventory of Depressive Symptomatology (QIDS-C16) measured thresholds of symptom severity at two study visits spaced 2 weeks apart. QIDS-C16 scores and PGE1 stimulated responses were stable between the two study visits. The QIDS-C16 was inversely correlated with PGE1 stimulated responses at each visit (rs = -0.33, rs = -0.60, respectively). MDD subjects with mild-moderate depressive symptoms (defined by QIDS-C16 ≥ 6) had significantly lower PGE1 stimulated responses than asymptomatic MDD subjects (QIDS-C16 < 6) or healthy controls (p = 0.001 and 0.002 respectively). MDD subjects with moderate depressive symptoms (QIDS-C16 ≥ 10) had the lowest PGE1 responses of all subjects (Fisher's exact = 0.012). These results support our earlier findings that a simple, high-throughput-capable platelet assay may be a useful biomarker to assess the clinical status of depressed subjects. Larger studies are needed to evaluate the utility of this biomarker for diagnosis and treatment response.
Collapse
Affiliation(s)
- Aksu Gunay
- Deparment of Psychiatry, University of Illinois College of Medicine, Chicago, IL, 60612, USA
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Steven D Targum
- Deparment of Psychiatry, University of Illinois College of Medicine, Chicago, IL, 60612, USA
- Pax Neuroscience, Glenview, IL, 60025, USA
| | - Alex D Leow
- Deparment of Psychiatry, University of Illinois College of Medicine, Chicago, IL, 60612, USA
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, 60612, USA
- Department of Computer Science, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Olusola Ajilore
- Deparment of Psychiatry, University of Illinois College of Medicine, Chicago, IL, 60612, USA
| | - Mark M Rasenick
- Deparment of Psychiatry, University of Illinois College of Medicine, Chicago, IL, 60612, USA.
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, 60612, USA.
- Pax Neuroscience, Glenview, IL, 60025, USA.
- Jesse Brown VAMC, Chicago, IL, USA.
| |
Collapse
|
29
|
He Y, Hong Q, Chen S, Zhou J, Qiu S. Reprogramming tumor-associated macrophages in gastric cancer: a pathway to enhanced immunotherapy. Front Immunol 2025; 16:1558091. [PMID: 40098971 PMCID: PMC11911521 DOI: 10.3389/fimmu.2025.1558091] [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: 01/09/2025] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Gastric cancer (GC) remains a significant global health concern due to its poor prognosis and limited therapeutic options, particularly in advanced stages. Tumor microenvironment (TME), particularly tumor-associated macrophages (TAMs), plays a key role in tumor progression, immune evasion, and therapy resistance. TAMs exhibit plasticity, shifting between pro-inflammatory M1 and immunosuppressive M2 phenotypes, with the latter predominating in GC and contributing to poor outcomes. Recent therapeutic advancements focus on targeting TAMs, including inhibiting M2 polarization, reprogramming TAMs to M1 phenotypes, and combining TAM-targeted approaches with immune checkpoint inhibitors. Innovations in nanotechnology, metabolic reprogramming, and targeting key pathways such as interleukin-6 and C-C motif ligand 2/C-C motif chemokine receptor 2 further enhance these strategies. However, challenges remain, including the spatial and functional heterogeneity of TAMs within the TME and the need for selective targeting to avoid disrupting immune homeostasis. Ongoing research on TAM origins, functions, and interactions within the TME is crucial for developing precise and effective therapies. These advances hold promise not only for improving outcomes in GC but also for addressing other cancers with similarly complex microenvironments.
Collapse
Affiliation(s)
| | | | | | | | - Shengliang Qiu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang
Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
30
|
Querio G, Geddo F, Antoniotti S, Femminò S, Gallo MP, Penna C, Pagliaro P. Stay connected: The myoendothelial junction proteins in vascular function and dysfunction. Vascul Pharmacol 2025; 158:107463. [PMID: 39814089 DOI: 10.1016/j.vph.2025.107463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/18/2025]
Abstract
The appropriate regulation of peripheral vascular tone is crucial for maintaining tissue perfusion. Myoendothelial junctions (MEJs), specialized connections between endothelial cells and vascular smooth muscle cells, are primarily located in peripheral resistance vessels. Therefore, these junctions, with their key membrane proteins, play a pivotal role in the physiological control of relaxation-contraction coupling in resistance arterioles, mainly mediated through endothelium-derived hyperpolarization (EDH). This review aims to illustrate the mechanisms involved in the initiation and propagation of EDH, emphasizing the role of membrane proteins involved in its generation (TRPV4, Piezo1, ASIC1a) and propagation (connexins, Notch). Finally, we discuss relevant studies on pathological events linked to EDH dysfunction and discuss novel approaches, including the effects of natural and dietary bioactive molecules, in modulating EDH-mediated vascular tone.
Collapse
Affiliation(s)
- Giulia Querio
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, 10043 Orbassano, Italy.
| | - Federica Geddo
- Department of Life Sciences and Systems Biology, University of Torino, Via Accademia Albertina 13, 10123 Torino, Italy.
| | - Susanna Antoniotti
- Department of Life Sciences and Systems Biology, University of Torino, Via Accademia Albertina 13, 10123 Torino, Italy.
| | - Saveria Femminò
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, 10043 Orbassano, Italy.
| | - Maria Pia Gallo
- Department of Life Sciences and Systems Biology, University of Torino, Via Accademia Albertina 13, 10123 Torino, Italy.
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, 10043 Orbassano, Italy; National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy.
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, Regione Gonzole 10, 10043 Orbassano, Italy.
| |
Collapse
|
31
|
Zhang Y, Jia X, Wang Y, Zheng Q. Caveolin-1-mediated LDL transcytosis across endothelial cells in atherosclerosis. Atherosclerosis 2025; 402:119113. [PMID: 39914325 DOI: 10.1016/j.atherosclerosis.2025.119113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/20/2024] [Accepted: 01/26/2025] [Indexed: 03/09/2025]
Abstract
Atherosclerosis is widely recognized as a chronic inflammatory disease of the arterial wall characterized by the progressive accumulation of lipids, inflammatory cells, and fibrous material in the subendothelial space of large arteries. The occurrence and pathogenesis of atherosclerosis are intricately linked to the deposition of low-density lipoprotein (LDL) in the arterial wall. LDL must cross the intact endothelium to reach the subendothelial space, with caveolin-1 assuming a crucial role in this process. Caveolin-1 is a 21-24 kDa membrane protein located in caveolae and highly expressed in endothelial cells. Previous investigations have demonstrated the pivotal role of caveolin-1 in fostering atherosclerosis through its modulation of membrane trafficking, cholesterol metabolism, and cellular signaling. However, how caveolin-1 regulates LDL transcytosis across endothelial cells in atherosclerosis remains unclear. We provide a comprehensive overview of recent research on the interplay between caveolin-1 and atherosclerosis, with a specific focus on elucidating the role of caveolin-1 in mediating LDL transcytosis across endothelial cells. This review furnishes theoretical foundations supporting the pivotal role of caveolin-1 in both the inception and progression of atherosclerosis. It underscores the prospective viability of caveolin-1 as a new therapeutic target for atherosclerosis and introduces novel perspectives for treatment strategies in the early stages of atherosclerosis.
Collapse
Affiliation(s)
- Yifei Zhang
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China
| | - Xiong Jia
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Yayu Wang
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Qijun Zheng
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China.
| |
Collapse
|
32
|
Xu Y, Li SC, Xiao J, Liu Q, Cherukuri D, Liu Y, Mirshahidi S, Xu J, Chen X, Homa D, Olea J, Wu K, Kelly KR, Sun F, Huang R, Wang X, Wen Q, Zhang X, Ghiuzeli CM, Chong E, Abdel-Azim H, Reeves ME, Baylink DJ, Cao H, Zhong JF. Exploring treatment-driven subclonal evolution of prognostic triple biomarkers: Dual gene fusions and chimeric RNA variants in novel subtypes of acute myeloid leukemia patients with KMT2A rearrangement. Drug Resist Updat 2025; 79:101199. [PMID: 39823827 DOI: 10.1016/j.drup.2024.101199] [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: 08/02/2024] [Revised: 12/24/2024] [Accepted: 12/24/2024] [Indexed: 01/20/2025]
Abstract
Chromosomal rearrangements (CR) initiate leukemogenesis in approximately 50 % of acute myeloid leukemia (AML) patients; however, limited targeted therapies exist due to a lack of accurate molecular and genetic biomarkers of refractory mechanisms during treatment. Here, we investigated the pathological landscape of treatment resistance and relapse in 16 CR-AML patients by monitoring cytogenetic, RNAseq, and genome-wide changes among newly diagnosed, refractory, and relapsed AML. First, in FISH-diagnosed KMT2A (MLL gene, 11q23)/AFDN (AF6, 6q27)-rearrangement, RNA-sequencing identified an unknown CCDC32 (15q15.1)/CBX3 (7p15.2) gene fusion in both newly diagnosed and relapsed samples, which is previously unknown in KMT2A/AFDN-rearranged AML patients. Second, the unreported CCDC32/CBX3 gene fusion significantly affected the expression of wild-type genes of both CCDC32 (essential for embryonic development) and CBX3 (an oncogene for solid tumors) during the relapse, as demonstrated by Quantitative PCR analyses. Third, we further confirmed the existence of triple biomarkers - KMT2A/AFDN (AF6, 6q27) rearrangement, the unknown CCDC32 (15q15.1)/CBX3 (7p15.2) gene fusion and chimeric RNA variants (treatment-resistant leukemic blasts harboring distinct breakpoints) in a 21-year-old male patient of rapid relapsed/refractory AML. Most intriguingly, in this work regarding 16 patients, patients 7 and 20 initially showed the KMT2A/AFDN gene fusion; upon relapse, patient 20 did not show this fusion. On the other hand, patient 7 retained the KMT2A/AFDN fusion at diagnosis and during the relapse, only identified by PCR and Sanger's Sequencing, not by cytogenetics. Interestingly, the chimeric CCDC32/CBX3 gene fusion persisted in the 21-year-old male patient over the diagnostic and relapse phases. Most intriguingly, the overexpression of CCDC32/CBX3 fusion gene in AML patient-specific MV4-11 cells confirms the functional validation, providing experimental evidence of the biological impact of the CCDC32/CBX3 fusion on AML pathogenesis and treatment resistance by promoting cell cycle progression, a mechanism through which AML evolves to become treatment-resistant. All these might exhort differential resistance to treatment. Thus, we found that prognostic and predictive triple biomarkers - KRAS mutated, dual fusions (KMT2A/AFDN, CCDC32/CBX3), and chimeric variants - might evolve with a potential oncogenic role of subclonal evolution for poor clinical outcomes.
Collapse
Affiliation(s)
- Yi Xu
- Loma Linda University Cancer Center, Loma Linda, CA 92354, United States; Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, United States; Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, United States.
| | - Shengwen Calvin Li
- Department of Neurology, University of California-Irvine School of Medicine, 200 S. Manchester Ave. Ste. 206, Orange, CA 92868, United States; CHOC Children's Research Institute, Children's Hospital of Orange County (CHOC®), Rady Children's Health, 1201 La Veta Ave, Orange, CA 92868-3874, United States.
| | - Jeffrey Xiao
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Qian Liu
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, 4505 S Maryland Pkwy, Las Vegas, NV 89154, United States; School of Life Sciences, College of Sciences, University of Nevada, 4505 S Maryland Pkwy, Las Vegas, NV 89154, United States
| | - Durga Cherukuri
- Department of Pathology & Laboratory Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Yan Liu
- Department of Pathology & Laboratory Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Saied Mirshahidi
- Loma Linda University Cancer Center, Loma Linda, CA 92354, United States; Biospecimen Laboratory, Loma Linda University Cancer Center, Department of Medicine and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Jane Xu
- Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92354, United States
| | - Xuelian Chen
- Department of Medicine/Division of Hematology, University of Southern California, Los Angeles, CA, United States
| | - Dadrastoussi Homa
- Department of Medicine/Division of Hematology, University of Southern California, Los Angeles, CA, United States
| | - Julian Olea
- Department of Medicine/Division of Hematology, University of Southern California, Los Angeles, CA, United States
| | - Kaijin Wu
- Department of Medicine/Division of Hematology, University of Southern California, Los Angeles, CA, United States
| | - Kevin R Kelly
- Department of Medicine/Division of Hematology, University of Southern California, Los Angeles, CA, United States
| | - Fengzhu Sun
- Quantitative and Computational Biology Department, University of Southern California, Los Angeles, CA, United States
| | - Ruihao Huang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Xiaoqi Wang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Qin Wen
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, China.
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Cristina M Ghiuzeli
- Division of Hematology, University of Washington School of Medicine, Seattle, WA, United States
| | - Esther Chong
- Loma Linda University Cancer Center, Loma Linda, CA 92354, United States; Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Hisham Abdel-Azim
- Loma Linda University Cancer Center, Loma Linda, CA 92354, United States; Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92354, United States; Division of Transplant and Cell Therapy/Hematological Malignancies, Departments of Pediatrics, Loma Linda University, Loma Linda, CA 92354, United States
| | - Mark E Reeves
- Loma Linda University Cancer Center, Loma Linda, CA 92354, United States; Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - David J Baylink
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Huynh Cao
- Loma Linda University Cancer Center, Loma Linda, CA 92354, United States; Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Jiang F Zhong
- Loma Linda University Cancer Center, Loma Linda, CA 92354, United States; Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92354, United States.
| |
Collapse
|
33
|
Li Z, Han L, Wang B, Wang P, Wang Y, Wang R, Lv X, Feng Y. The role of Piezo1 in bone marrow stem cells in response to elevated intraosseous pressure on regulating osteogenesis and angiogenesis of steroid-induced osteonecrosis of the femoral head. J Orthop Translat 2025; 51:278-289. [PMID: 40190343 PMCID: PMC11968285 DOI: 10.1016/j.jot.2025.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/18/2024] [Accepted: 01/08/2025] [Indexed: 04/09/2025] Open
Abstract
Objectives Steroid-induced osteonecrosis of the femoral head (SONFH) remains a significant global health issue, with an unclear pathogenesis. Elevated intraosseous pressure is considered a key initiating factor in SONFH development. Impaired osteogenesis and angiogenesis are believed to be critical in SONFH progression. Piezo1, a mechanosensitive cation channel, may sense changes in intraosseous pressure. In this study, we set out to explore the role of Piezo1 in SONFH and how to target Piezo1 to treat SONFH. Methods Femoral head tissue specimens were collected from patients with ONFH and femoral neck fracture. Histological staining, Western blotting, and RT-PCR analysis were conducted to investigate the relationship between elevated intraosseous pressure and SONFH in rat models. Immunofluorescence staining of femoral head tissues was performed to study the spatiotemporal relationship between elevated intraosseous pressure and angiogenesis, osteogenesis, and development of SONFH. Results In the early stages of SONFH, elevated intraosseous pressure increased angiogenesis and osteogenesis. However, as the pressure continued to rise, both processes were inhibited. Furthermore, Elevated intraosseous pressure activated the Piezo1 signaling pathway in bone marrow stem cells. Piezo1 activation led to increased intracellular calcium influx, thus enhancing osteogenesis and angiogenesis through CAM-NFAT1 signaling pathway. Conclusion In the early stages of SONFH, Piezo1 in BMSCs senses increased intraosseous pressure, promoting angiogenesis and osteogenesis. Targeting Piezo1 to promote the osteogenic and angiogenic potential of stem cells, which could curb further increases in pressure, contribute to early treatment of SONFH. The translational potential of this article Currently, many mechanisms of the impact of elevated intraosseous pressure on osteonecrosis of the femoral head are still in the basic theoretical research stage, and we hope to translate them into clinical applications as soon as possible. We discovered that targeting Piezo1 curb further increases in intraosseous pressure, alleviating the damaging effects of glucocorticoids on stem cells and blood vessels, which exerting great significance in treatment of early stage SONFH.
Collapse
Affiliation(s)
- Zilin Li
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lizhi Han
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, Anhui Key Laboratory of Tissue Transformation, Bengbu Medical University, Bengbu, 233000, Anhui Province, China
| | - Bo Wang
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Wang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuxi Wang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruoyu Wang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Lv
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Feng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
34
|
Silva B, Bragança J. Induced pluripotent stem cell-derived mesenchymal stem cells for modeling and treating metabolic associated fatty liver disease and metabolic associated steatohepatitis: Challenges and opportunities. World J Stem Cells 2025; 17:99331. [DOI: 10.4252/wjsc.v17.i2.99331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/21/2024] [Accepted: 01/14/2025] [Indexed: 02/24/2025] Open
Abstract
The potential of induced pluripotent stem cells (iPSCs) for modeling and treating metabolic associated fatty liver disease (MAFLD) and metabolic associated steatohepatitis (MASH) is emerging. MAFLD is a growing global health concern, currently with limited treatment options. While primary mesenchymal stem cells hold promise, iPSCs offer a versatile alternative due to their ability to differentiate into various cell types, including iPSC-derived mesenchymal stem cells. However, challenges remain, including optimizing differentiation protocols, ensuring cell safety, and addressing potential tumorigenicity risks. In addition, iPSCs offer the possibility to generate complex cellular models, including three-dimensional organoid models, which are closer representations of the human disease than animal models. Those models would also be valuable for drug discovery and personalized medicine approaches. Overall, iPSCs and their derivatives offer new perspectives for advancing MAFLD/MASH research and developing novel therapeutic strategies. Further research is needed to overcome current limitations and translate this potential into effective clinical applications.
Collapse
Affiliation(s)
- Bárbara Silva
- Algarve Biomedical Center-Research Institute, University of Algarve, Faro 8005-139, Portugal
- Algarve Biomedical Center, University of Algarve, Faro 8005-139, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro 8005-139, Portugal
- PhD Program in Biomedical Sciences, Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro 8005-139, Portugal
| | - José Bragança
- Algarve Biomedical Center, University of Algarve, Faro 8005-139, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro 8005-139, Portugal
- Faculty of Medicine and Biomedical Sciences, Algarve Biomedical Center-Research Institute, University of Algarve, Faro 8005-139, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, Lisbon 1000-001, Portugal
| |
Collapse
|
35
|
Toghraie FS, Bayat M, Hosseini MS, Ramezani A. Tumor-infiltrating myeloid cells; mechanisms, functional significance, and targeting in cancer therapy. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01051-y. [PMID: 39998754 DOI: 10.1007/s13402-025-01051-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 02/27/2025] Open
Abstract
Tumor-infiltrating myeloid cells (TIMs), which encompass tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), myeloid-derived suppressor cells (MDSCs), and tumor-associated dendritic cells (TADCs), are of great importance in tumor microenvironment (TME) and are integral to both pro- and anti-tumor immunity. Nevertheless, the phenotypic heterogeneity and functional plasticity of TIMs have posed challenges in fully understanding their complexity roles within the TME. Emerging evidence suggested that the presence of TIMs is frequently linked to prevention of cancer treatment and improvement of patient outcomes and survival. Given their pivotal function in the TME, TIMs have recently been recognized as critical targets for therapeutic approaches aimed at augmenting immunostimulatory myeloid cell populations while depleting or modifying those that are immunosuppressive. This review will explore the important properties of TIMs related to immunity, angiogenesis, and metastasis. We will also document the latest therapeutic strategies targeting TIMs in preclinical and clinical settings. Our objective is to illustrate the potential of TIMs as immunological targets that may improve the outcomes of existing cancer treatments.
Collapse
Affiliation(s)
- Fatemeh Sadat Toghraie
- Institute of Biotechnology, Faculty of the Environment and Natural Sciences, Brandenburg University of Technology, Cottbus, Germany
| | - Maryam Bayat
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsa Sadat Hosseini
- Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Amin Ramezani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
36
|
Sasamoto Y, Kiritoshi S, Lee CAA, Fukuda Y, Martin G, Ksander BR, Frank MH, Frank NY. Caveolin 1 and 2 enhance the proliferative capacity of BCAM-positive corneal progenitors. Sci Rep 2025; 15:6672. [PMID: 39994275 PMCID: PMC11850879 DOI: 10.1038/s41598-024-81283-4] [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: 06/21/2024] [Accepted: 11/25/2024] [Indexed: 02/26/2025] Open
Abstract
Caveolin (CAV) 1 and 2 are integral membrane proteins that constitute major components of small membrane pouches termed caveolae. While several functions have been described in other tissues, the roles of CAV1 and CAV2 in the ocular surface have remained unknown. In the current study, we investigated the expression and function of CAV1 and CAV2 in the human cornea. We found CAV1 and CAV2 to be preferentially expressed by proliferative Basal Cell Adhesion Molecule (BCAM)-positive progenitor cells along the entire limbal and corneal basal epithelial layer. Functional gene knockdown studies reveal that BCAM, BCAM co-expressed Laminin α5 (LAMA5) and Laminin α3 (LAMA3) regulate expression of CAV2. Mechanistically, we demonstrate that CAV1 and CAV2 contribute to enhanced BCAM-positive cell proliferation through regulation of Fibroblast Growth Factor Receptor 2 (FGFR2) cell surface expression. In aggregate, our study identifies specific expression of CAV1 and CAV2 in BCAM-positive corneal basal epithelial cells and uncovers a novel CAV1/CAV2-dependent mechanism of corneal progenitor cell proliferation, with potential implications for therapeutic enhancement of corneal regeneration.
Collapse
Affiliation(s)
- Yuzuru Sasamoto
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA.
- Transplant Research Program, Boston Children's Hospital, Boston, MA, USA.
- Department of Ophthalmology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA.
| | - Shoko Kiritoshi
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Yoshiko Fukuda
- Department of Ophthalmology, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Gabrielle Martin
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Transplant Research Program, Boston Children's Hospital, Boston, MA, USA
| | - Bruce R Ksander
- Massachusetts Eye & Ear Infirmary, Schepens Eye Research Institute, Boston, MA, USA
| | - Markus H Frank
- Transplant Research Program, Boston Children's Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Department of Dermatology, Harvard Skin Disease Research Center, Brigham and Women's Hospital, Boston, MA, USA
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| | - Natasha Y Frank
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
- Department of Medicine, VA Boston Healthcare System, Boston, MA, USA.
| |
Collapse
|
37
|
Zadorozny L, Du J, Supanekar N, Annamalai K, Yu Q, Wang M. Caveolin and oxidative stress in cardiac pathology. Front Physiol 2025; 16:1550647. [PMID: 40041164 PMCID: PMC11876135 DOI: 10.3389/fphys.2025.1550647] [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: 12/23/2024] [Accepted: 01/27/2025] [Indexed: 03/06/2025] Open
Abstract
Caveolins interact with signaling molecules within caveolae and subcellular membranes. Dysregulation of caveolin function and protein abundance contributes to cardiac pathophysiological processes, driving the development and progression of heart disease. Reactive oxygen species (ROS) play a critical role in maintaining cellular homeostasis and are key contributors to the pathophysiological mechanisms of cardiovascular disorders. Caveolins have been shown to modulate oxidative stress and regulate redox homeostasis. However, the specific roles of caveolins, particularly caveolin-1 and caveolin-3, in regulating ROS production during cardiac pathology remain unclear. This mini-review article highlights the correlation between caveolins and oxidative stress in maintaining cardiovascular health and modulating cardiac diseases, specifically in myocardial ischemia, heart failure, diabetes-induced metabolic cardiomyopathy, and septic cardiomyopathy. A deeper understanding of caveolin-mediated mechanisms may pave the way for innovative therapeutic approaches to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Lauren Zadorozny
- Center for Surgical Sciences, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Division of Cardiothoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jiayue Du
- Center for Surgical Sciences, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Division of Cardiothoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Neil Supanekar
- Center for Surgical Sciences, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Karthik Annamalai
- Center for Surgical Sciences, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Qing Yu
- Center for Surgical Sciences, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Division of Cardiothoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Meijing Wang
- Center for Surgical Sciences, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Division of Cardiothoracic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
38
|
Amatya B, Polzin JQM, Villar VAM, Yang J, Konkalmatt P, Wang X, Cadme RC, Xu P, Gildea JJ, Cuevas S, Armando I, Felder RA, Jose PA, Lee H. SNX19 Interacts with Caveolin-1 and Flotillin-1 to Regulate D 1R Endocytosis and Signaling. Biomedicines 2025; 13:481. [PMID: 40002894 PMCID: PMC11853350 DOI: 10.3390/biomedicines13020481] [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: 01/03/2025] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Sorting nexin 19 (SNX19) is important in the localization and trafficking of the dopamine D1 receptor (D1R) to lipid raft microdomains. However, the interaction between SNX19 and the lipid raft components caveolin-1 or flotillin-1 and, in particular, their roles in the cellular endocytosis and cell membrane trafficking of the D1R have not been determined. Methods: Caveolin-1 and flotillin-1 motifs were analyzed by in silico analysis; colocalization was observed by confocal immunofluorescence microscopy; protein-protein interaction was determined by co-immunoprecipitation. Results: In silico analysis revealed the presence of putative caveolin-1 and flotillin-1 binding motifs within SNX19. In mouse and human renal proximal tubule cells (RPTCs), SNX19 was localized mainly in lipid rafts. In mouse RPTCs transfected with wild-type (WT) Snx19, fenoldopam (FEN), a D1-like receptor agonist, increased the colocalization of SNX19 with caveolin-1 and flotillin-1. FEN also increased the co-immunoprecipitation of SNX19 with caveolin-1 and flotillin-1, effects that were prevented by SCH39166, a D1-like receptor antagonist. The FEN-mediated increase in the residence of SNX19 in lipid rafts and the colocalization of the D1R with caveolin-1 and flotilin-1 were attenuated by the deletion of a caveolin-1 (YHTVNRRYREF) (ΔCav1) or a flotillin-1 (EEGPGTETETGLPVS) (ΔFlot1) binding motif. The FEN-mediated increase in intracellular cAMP production was also impaired by the deletion of either the flotillin-1 or caveolin-1 binding motif. Nocodazole, a microtubule depolymerization inhibitor, interfered with the FEN-mediated increase in the colocalization between SNX19 and D1R. Conclusion: SNX19 contains caveolin-1 and flotillin-1 binding motifs, which play an important role in D1R endocytosis and signaling.
Collapse
Affiliation(s)
- Bibhas Amatya
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jacob Q. M. Polzin
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
| | - Van A. M. Villar
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 20 Penn Street, HSF II, Baltimore, MD 21201, USA;
| | - Jiang Yang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 20 Penn Street, HSF II, Baltimore, MD 21201, USA;
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Prasad Konkalmatt
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 20 Penn Street, HSF II, Baltimore, MD 21201, USA;
| | - Xiaoyan Wang
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 20 Penn Street, HSF II, Baltimore, MD 21201, USA;
- Department of Nephrology, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Raisha C. Cadme
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
| | - Peng Xu
- Department of Pathology, University of Virginia Health Sciences Center, Charlottesville, VA 22908, USA; (P.X.); (J.J.G.); (R.A.F.)
| | - John J. Gildea
- Department of Pathology, University of Virginia Health Sciences Center, Charlottesville, VA 22908, USA; (P.X.); (J.J.G.); (R.A.F.)
| | - Santiago Cuevas
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 20 Penn Street, HSF II, Baltimore, MD 21201, USA;
- Physiopathology of the Inflammation and Oxidative Stress Laboratory, Molecular Inflammation Group, Biomedical Research Institute of Murcia Pascual Parrilla (IMIB), 30120 Palmar, Spain
| | - Ines Armando
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 20 Penn Street, HSF II, Baltimore, MD 21201, USA;
| | - Robin A. Felder
- Department of Pathology, University of Virginia Health Sciences Center, Charlottesville, VA 22908, USA; (P.X.); (J.J.G.); (R.A.F.)
| | - Pedro A. Jose
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 20 Penn Street, HSF II, Baltimore, MD 21201, USA;
- Department of Pharmacology & Physiology, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA
| | - Hewang Lee
- Division of Kidney Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC 20052, USA; (B.A.); (J.Q.M.P.); (V.A.M.V.); (P.K.); (X.W.); (R.C.C.); (S.C.); (I.A.); (P.A.J.)
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 20 Penn Street, HSF II, Baltimore, MD 21201, USA;
| |
Collapse
|
39
|
Li S, Liu J, Peyton M, Lazaro O, McCabe SD, Huang X, Liu Y, Shi Z, Zhang Z, Walker BA, Johnson TS. Multiple Myeloma Insights from Single-Cell Analysis: Clonal Evolution, the Microenvironment, Therapy Evasion, and Clinical Implications. Cancers (Basel) 2025; 17:653. [PMID: 40002248 PMCID: PMC11852428 DOI: 10.3390/cancers17040653] [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: 01/10/2025] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Multiple myeloma (MM) is a complex and heterogeneous hematologic malignancy characterized by clonal evolution, genetic instability, and interactions with a supportive tumor microenvironment. These factors contribute to treatment resistance, disease progression, and significant variability in clinical outcomes among patients. This review explores the mechanisms underlying MM progression, including the genetic and epigenetic changes that drive clonal evolution, the role of the bone marrow microenvironment in supporting tumor growth and immune evasion, and the impact of genomic instability. We highlight the critical insights gained from single-cell technologies, such as single-cell transcriptomics, genomics, and multiomics, which have enabled a detailed understanding of MM heterogeneity at the cellular level, facilitating the identification of rare cell populations and mechanisms of drug resistance. Despite the promise of advanced technologies, MM remains an incurable disease and challenges remain in their clinical application, including high costs, data complexity, and the need for standardized bioinformatics and ethical considerations. This review emphasizes the importance of continued research and collaboration to address these challenges, ultimately aiming to enhance personalized treatment strategies and improve patient outcomes in MM.
Collapse
Affiliation(s)
- Sihong Li
- Indiana Bioscience Research Institute, Indianapolis, IN 46202, USA
- Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Jiahui Liu
- Indiana Bioscience Research Institute, Indianapolis, IN 46202, USA
- Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Madeline Peyton
- Indiana Bioscience Research Institute, Indianapolis, IN 46202, USA
- Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Regenstrief Institute, Indianapolis, IN 46202, USA
| | - Olivia Lazaro
- Indiana Bioscience Research Institute, Indianapolis, IN 46202, USA
| | - Sean D. McCabe
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Xiaoqing Huang
- Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA
| | - Yunlong Liu
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University, Indianapolis, IN 46202, USA
| | - Zanyu Shi
- Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA
| | - Zhiqi Zhang
- Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Brian A. Walker
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University, Indianapolis, IN 46202, USA
| | - Travis S. Johnson
- Indiana Bioscience Research Institute, Indianapolis, IN 46202, USA
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University, Indianapolis, IN 46202, USA
| |
Collapse
|
40
|
Than H, Fan X, Cheung AMS, Hwang WYK, Poon Z. Rapid disease progression of myelodysplastic syndrome is reflected in transcriptomic and functional abnormalities of bone marrow mesenchymal stromal cells. Stem Cells 2025; 43:sxae073. [PMID: 39541184 DOI: 10.1093/stmcls/sxae073] [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: 04/01/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Bone marrow (BM) mesenchymal stromal cells (MSCs) are important regulators of hematopoietic stem and progenitor cells (HSPCs). When transformed into a dysplastic phenotype, MSCs contribute to hematopoietic diseases such as myelodysplastic syndromes (MDS), but it remains unclear if there are specific properties in MDS-MSCs that contribute to the disease course. To understand this, we investigated MDS-MSCs from fast (MDSfast) vs slow (MDSslow) progressing disease groups and discovered differences between these groups. MDSfast-MSCs secrete more inflammatory factors, support myeloid-skewed differentiation of HSPCs, and importantly, show poorer response to hypomethylation as a key differentiator in GSEA analysis. When exposed to long-term in vivo stimulation with primary MDSfast-MSCs-based scaffolds, healthy donor (HD) HSPCs show elevated NF-κB expression, similar to leukemic HSPCs in MDS. Those "MDSfast-MSCs-primed" HD-HSPCs continue to show enhanced engraftment rates in secondary MDS-MSC-based scaffolds, providing evidence for the microenvironmental selection pressures in MDS toward leukemic HSPCs. Together, our data point toward a degree of co-development between MSCs and HSPCs during the progression of MDS, where changes in MDS-MSCs take place mainly at the transcriptomic and functional levels. These unique differences in MDS-MSCs can be utilized to improve disease prognostication and implement targeted therapy for unmet clinical needs.
Collapse
Affiliation(s)
- Hein Than
- Department of Hematology, Singapore General Hospital, Singapore 169608
- Duke-NUS Medical School, National University of Singapore, Singapore 169857
- Department of Hematology, National Cancer Center, Singapore 168583
| | - Xiubo Fan
- Duke-NUS Medical School, National University of Singapore, Singapore 169857
- Department of Clinical Translational Research, Singapore General Hospital, Singapore 169608
| | - Alice M S Cheung
- Department of Hematology, Singapore General Hospital, Singapore 169608
- Duke-NUS Medical School, National University of Singapore, Singapore 169857
| | - William Y K Hwang
- Department of Hematology, Singapore General Hospital, Singapore 169608
- Duke-NUS Medical School, National University of Singapore, Singapore 169857
- Department of Hematology, National Cancer Center, Singapore 168583
| | - Zhiyong Poon
- Duke-NUS Medical School, National University of Singapore, Singapore 169857
- Department of Clinical Translational Research, Singapore General Hospital, Singapore 169608
| |
Collapse
|
41
|
Benarroch E. What Are the Functions of Caveolins and Their Role in Neurologic Disorders? Neurology 2025; 104:e213341. [PMID: 39805058 DOI: 10.1212/wnl.0000000000213341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 01/16/2025] Open
|
42
|
Zhang J, Zhang L, Sun Z, Li Z, Zou X, Sun S, Zhu L, Xi K, Liu Z, Deng G. Nano-alkaline ion-excited NETs ablative eye drops promote ocular surface recovery. J Control Release 2025; 378:864-879. [PMID: 39740695 DOI: 10.1016/j.jconrel.2024.12.071] [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: 10/14/2024] [Revised: 12/13/2024] [Accepted: 12/27/2024] [Indexed: 01/02/2025]
Abstract
Neutrophil extracellular traps (NETs) promote neovascularization during the acute phase after ocular chemical injury, while the local inflammatory acidic environment delays post-injury repair. Currently, the mechanism of NETs promoting neovascularization has not been fully elucidated, and there is a lack of therapeutic strategies to effectively improve the local microenvironment for corneal repair. In this study, we validated the NETs-M2-angiogenic pathway after injury. Using transcriptomics sequencing and liquid-phase microarray assays, the intrinsic immune cascade mechanism of NETs inducing macrophage M2 polarization and releasing VEGF via PI3K/AKT was identified. Based on this pathology and the physiological need to improve the local inflammatory acidic environment and promote corneal repair, we organically integrated the alkaline ion-rich bioglass with the highly transmissive and highly adhesive filipin protein, and constructed NETs ablative gel eye drops (DMS) that can release DNase I and alkaline ions in a sustained manner. The eye drops restricted the inflammatory interaction of NETs with macrophages from the source, adhered to the corneal surface and continuously released alkaline ions to improve the local acidic inflammatory environment, providing a favorable immune microenvironment for corneal recovery. We established a cell co-culture system and a corneal alkali burn model to further validate the role of DMS in modulating the intrinsic immune cascade of neovascularization for corneal repair and the related mechanisms. In conclusion, based on the biological mechanism of NETs-M2-VEGF after corneal chemical injury, the present study designed eye drops for dual regulation of intrinsic immunity and the inflammatory acid environment, which not only further supplemented and improved the pathophysiological mechanism of corneal neovascularization after chemical injury, but also provided a new way of thinking about corneal regeneration after injury.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Ophthalmology, Changzhou Third Peopls's Hospital, Changzhou Clinical College of Xuzhou Medical University, 300 Lanlin North road, Changzhou, Jiangsu 213000, China
| | - Lichen Zhang
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 708 Renmin Road, SuZhou, Jiangsu 215000, China
| | - Zhuo Sun
- Department of Ophthalmology, Changzhou Third Peopls's Hospital, Changzhou Clinical College of Xuzhou Medical University, 300 Lanlin North road, Changzhou, Jiangsu 213000, China
| | - Ziang Li
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 708 Renmin Road, SuZhou, Jiangsu 215000, China
| | - Xi Zou
- Department of Ophthalmology, Changzhou Third Peopls's Hospital, Changzhou Clinical College of Xuzhou Medical University, 300 Lanlin North road, Changzhou, Jiangsu 213000, China
| | - Shanshan Sun
- Department of Ophthalmology, Changzhou Third Peopls's Hospital, Changzhou Clinical College of Xuzhou Medical University, 300 Lanlin North road, Changzhou, Jiangsu 213000, China
| | - Lin Zhu
- Department of Ophthalmology, Changzhou Third Peopls's Hospital, Changzhou Clinical College of Xuzhou Medical University, 300 Lanlin North road, Changzhou, Jiangsu 213000, China
| | - Kun Xi
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 708 Renmin Road, SuZhou, Jiangsu 215000, China.
| | - Zhinan Liu
- Department of Ophthalmology, Changzhou Third Peopls's Hospital, Changzhou Clinical College of Xuzhou Medical University, 300 Lanlin North road, Changzhou, Jiangsu 213000, China.
| | - Guohua Deng
- Department of Ophthalmology, Changzhou Third Peopls's Hospital, Changzhou Clinical College of Xuzhou Medical University, 300 Lanlin North road, Changzhou, Jiangsu 213000, China.
| |
Collapse
|
43
|
Han N, Yu N, Yu L. The mRNA Stability of PIEZO1, Regulated by Methyltransferase-Like 3 via N 6-Methylation of Adenosine Modification in a YT521-B Homology Domain Family 2-Dependent Manner, Facilitates the Progression of Diabetic Retinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:265-280. [PMID: 39476953 DOI: 10.1016/j.ajpath.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 09/29/2024] [Accepted: 10/10/2024] [Indexed: 11/09/2024]
Abstract
Diabetic retinopathy (DR) is the major ocular complication of diabetes caused by chronic hyperglycemia, which leads to incurable blindness. Currently, the effectiveness of therapeutic interventions is limited. This study aimed to investigate the function of piezo-type mechanosensitive ion channel component 1 (PIEZO1) and its potential regulatory mechanism in DR progression. PIEZO1 expression was up-regulated in the retinal tissues of streptozotocin-induced diabetic mice and high-glucose (HG)-triggered Müller cells. Functionally, the knockdown of PIEZO1 improved the abnormal retinal function of diabetic mice and impeded inflammatory cytokine secretion and gliosis of Müller cells under HG conditions. Mechanistic investigations using RNA immunoprecipitation-real-time quantitative PCR, methylation RNA immunoprecipitation-real-time quantitative PCR, and luciferase reporter assays demonstrated that PIEZO1 was a downstream target of methyltransferase-like 3 (METTL3). METTL3-mediated N6-methyladenosine (m6A) modification within the coding sequence of PIEZO1 mRNA significantly shortened its half-life. In HG-stimulated cells, there was a negative regulatory relationship between PIEZO1 and YTH (YT521-B homology) domain family 2 (YTHDF2), a recognized m6A reader. The loss of YTHDF2 resulted in an extended half-life of PIEZO1 in cells with overexpression of METTL3, indicating that the effect of METTL3 on the mRNA stability of PIEZO1 was dependent on YTHDF2. Taken together, this study demonstrated the protective role of the PIEZO1 silencing in DR development, and that the degradation of PIEZO1 mRNA is accelerated by METTL3/YTHDF2-mediated m6A modification.
Collapse
Affiliation(s)
- Ning Han
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Na Yu
- Department of Blood Transfusion, The Second Hospital of Jilin University, Changchun, China
| | - Li Yu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
44
|
Su Y, Yin X. The Molecular Mechanism of Macrophages in Response to Mechanical Stress. Ann Biomed Eng 2025; 53:318-330. [PMID: 39354279 DOI: 10.1007/s10439-024-03616-8] [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: 05/10/2024] [Accepted: 09/03/2024] [Indexed: 10/03/2024]
Abstract
Macrophages, a type of functionally diversified immune cell involved in the progression of many physiologies and pathologies, could be mechanically activated. The physical properties of biomaterials including stiffness and topography have been recognized as exerting a considerable influence on macrophage behaviors, such as adhesion, migration, proliferation, and polarization. Recent articles and reviews on the physical and mechanical cues that regulate the macrophage's behavior are available; however, the underlying mechanism still deserves further investigation. Here, we summarized the molecular mechanism of macrophage behavior through three parts, as follows: (1) mechanosensing on the cell membrane, (2) mechanotransmission by the cytoskeleton, (3) mechanotransduction in the nucleus. Finally, the present challenges in understanding the mechanism were also noted. In this review, we clarified the associated mechanism of the macrophage mechanotransduction pathway which could provide mechanistic insights into the development of treatment for diseases like bone-related diseases as molecular targets become possible.
Collapse
Affiliation(s)
- Yuntong Su
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xing Yin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
45
|
Xiang Y, Sun G, Tian L, Xiang P, Xie C. Single-cell sequencing reveals the mechanisms of multiple myeloma progression: clarity or confusion? Ann Hematol 2025; 104:895-912. [PMID: 39918600 PMCID: PMC11971202 DOI: 10.1007/s00277-025-06241-0] [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: 12/20/2024] [Accepted: 01/30/2025] [Indexed: 04/05/2025]
Abstract
Multiple myeloma (MM), the second most common hematologic malignancy, is characterized by the clonal expansion of myeloma cells and accumulation of genetic lesions. MM progression is accompanied by increased aggressiveness and drug resistance. Even the goal of "cure" remains hard to reach for most patients, advances in diagnosis and treatment have allowed some to achieve durable remissions and transition to plateau phase. Single-cell sequencing, with its powerful ability to analyze cellular heterogeneity and molecular patterns at ground-breaking resolution, is informative for deciphering tumors and their microenvironment. In this review, we summarize the new insights of studies facilitated by emerging single-cell sequencing into clonal evolution, myeloma-supported microenvironment transformation, epigenetic changes, and novel prognostic and therapeutic strategies for MM, revealing the key mechanisms underlying MM progression and the direction of future efforts. With the continuous expansion of the research scope and optimization of related technologies, single-cell sequencing is expected to revolutionize our understanding of the biology and evolutionary dynamics of MM and contribute to the radical and precise improvement of treatment.
Collapse
Affiliation(s)
- Yunhui Xiang
- Department of Laboratory Medicine and Key Laboratory of Port Epidemic Surveillance in Sichuan Province, Sichuan International Travel and Healthcare Center (Chengdu Customs District Port Clinic), Chengdu, 610042, China
| | - Guokang Sun
- Department of Laboratory Medicine, West China School of Public Health and West China Fourth Hospital of Sichuan University, Chengdu, 610041, China
| | - Lvbo Tian
- Department of Laboratory Medicine and Key Laboratory of Port Epidemic Surveillance in Sichuan Province, Sichuan International Travel and Healthcare Center (Chengdu Customs District Port Clinic), Chengdu, 610042, China
| | - Pinpin Xiang
- Department of Laboratory Medicine, Xiping Community Healthcare Center of Longquanyi District, Chengdu, 610107, China
| | - Chunbao Xie
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital & University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
46
|
Li Y, Li W, Liu Y, Peng Y, Tang J, Li X. Efficacy and safety of anlotinib combined with vinorelbine as second‑line treatment for elderly patients with advanced squamous cell lung carcinoma: A retrospective cohort. Mol Clin Oncol 2025; 22:21. [PMID: 39776939 PMCID: PMC11706458 DOI: 10.3892/mco.2024.2816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/06/2024] [Indexed: 01/05/2025] Open
Abstract
The aim of the present study was to investigate the efficacy and safety of anlotinib combined with vinorelbine (NVB) as a second-line treatment for elderly patients with advanced squamous cell lung carcinoma (SqCLC). The present retrospective analysis included 48 elderly patients (aged ≥65 years) diagnosed with advanced SqCLC who received anlotinib in combination with NVB as a second-line therapy between January 2021 and December 2023. The primary endpoints assessed were overall survival (OS), progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR) and safety profile. The median PFS and OS for the cohort was found to be 5.0 and 9.5 months, respectively. By contrast, the ORR and DCR were found to be 29.17 and 70.83%. Further subgroup analysis indicated that patients who experienced specific adverse events (AEs), such as hypertension, proteinuria and hand-foot syndrome during treatment, generally had superior efficacy compared with those who did not experience these AEs (mPFS, 6.0 vs. 4.0 months; mOS, 11.0 vs. 8.5 months). In addition, apart from promising efficacy, patients who experienced common AEs also experienced decreased appetite (35.42%), fatigue (29.17%), hypertension (25%) and hand-foot syndrome (27.08%). Grade 3 or higher AEs occurred in <30% of patients, the majority of which was alleviated through corresponding support care. These results suggest that the combination of anlotinib and NVB as second-line therapy for elderly patients with advanced SqCLC demonstrated promising efficacy and a manageable safety profile. Such regimen may be a viable treatment option for this patient population. However, further prospective studies are required to validate these findings and optimize the dosing schedule for improved therapeutic outcomes.
Collapse
Affiliation(s)
- Yonghong Li
- The Department of Oncology, The First People's Hospital of Tianmen, Tianmen, Hubei 431700, P.R. China
| | - Wei Li
- The Department of Oncology, The First People's Hospital of Tianmen, Tianmen, Hubei 431700, P.R. China
| | - Yirui Liu
- Department of Nursing, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, P.R. China
| | - Yi Peng
- Department of Radiotherapy, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, P.R. China
| | - Jing Tang
- Department of Lymphoma, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, P.R. China
| | - Xiaobing Li
- Department of Thoracic Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, P.R. China
| |
Collapse
|
47
|
Benavent N, Cañete A, Moreno L, Gros L, Verdú-Amorós J, Salinas JA, Navarro S, Álvaro T, Carbonell-Asins JA, Noguera R. Risk of developing neuroblastoma influenced by maternal stressful life events during pregnancy and congenital pathologies. Pediatr Blood Cancer 2025; 72:e31402. [PMID: 39618320 DOI: 10.1002/pbc.31402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 12/14/2024]
Abstract
OBJECTIVE A retrospective multicenter study to investigate the potential association between descriptive information related to pregnancy history and perinatal features and the risk of neuroblastoma (NB) in children. STUDY DESIGN Data from 56 mothers during 105 pregnancies (56 cases of NB, 49 control siblings) were collected through face-to-face or telephone interviews with mothers of children diagnosed with NB, along with information extracted from Health System databases. Descriptive information related to (a) pregnancy history as maternal stressful life events with perceived distress during pregnancy, weight gain, alcohol and tobacco consumption, mode of delivery and gestational age; and (b) perinatal features as congenital pathologies, weight at birth and type of feeding were examined to identify potential risk factors for NB. RESULTS Stressful life events during pregnancy and certain congenital pathologies were independently associated with NB risk. No significant associations were found between other features. Breastfeeding rates were similar between cases and controls. CONCLUSION Our results underscore the importance of providing support and care to pregnant women to reduce potential stressors. Further research is needed to better understand the influence of dysbiosis and mitochondrial-nuclear communication impairment as underlying mechanisms of maternal stress during pregnancy and presence of congenital pathologies in order to confirm them as potential risk factors for NB.
Collapse
Affiliation(s)
- Nuria Benavent
- Pathology Department, Medical School, University of Valencia, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Adela Cañete
- Pediatric Oncology Unit, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Lucas Moreno
- Pediatric Oncology Unit, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Luis Gros
- Pediatric Oncology Unit, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Jaime Verdú-Amorós
- Pediatric Oncology Unit, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Jose Antonio Salinas
- Pediatric Oncology Unit, Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | - Samuel Navarro
- Pathology Department, Medical School, University of Valencia, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
- Centro de investigación biomédica en red de cáncer (CIBERONC), Madrid, Spain
| | - Tomas Álvaro
- Centro de investigación biomédica en red de cáncer (CIBERONC), Madrid, Spain
- Department of Pathology, Hospital de Tortosa Verge de la Cinta, Madrid, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tortosa, Spain
| | | | - Rosa Noguera
- Pathology Department, Medical School, University of Valencia, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
- Centro de investigación biomédica en red de cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|
48
|
Segal D, Wang X, Mazloom-Farisbaf H, Rajendran D, Butler E, Chen B, Chang BJ, Ahuja K, Perny A, Bhatt K, Reed DK, Castrillon DH, Lee J, Jeffery E, Wang L, Nguyen K, Williams NS, Skapek SX, Rajaram S, Fiolka R, Jaqaman K, Hon G, Amatruda JF, Danuser G. Caveolin-1 regulates context-dependent signaling and survival in Ewing sarcoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.23.614468. [PMID: 39713413 PMCID: PMC11661136 DOI: 10.1101/2024.09.23.614468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Plasticity is a hallmark function of cancer cells, but many of the underlying mechanisms have yet to be discovered. In this study, we identify Caveolin-1, a scaffolding protein that organizes plasma membrane domains, as a context-dependent regulator of survival signaling in Ewing sarcoma (EwS). Single cell analyses reveal a distinct subpopulation of EwS cells, which highly express the surface marker CD99 as well as Caveolin-1. CD99 High cells exhibit distinct morphology, gene expression, and enhanced survival capabilities compared to CD99 Low cells, both under chemotherapeutic challenge and in vivo. Mechanistically, we show that elevated Caveolin-1 expression in CD99 High cells orchestrates PI3K/AKT survival signaling by modulating the spatial organization of PI3K activity at the cell surface. Notably, CD99 itself is not directly involved in this pathway, making it a useful independent marker for identifying these subpopulations. We propose a model where the CD99 High state establishes a Cav-1-driven signaling network to support cell survival that is distinct from the survival mechanisms of CD99 Low cells. This work reveals a dynamic state transition in EwS cells and highlights Caveolin-1 as a key driver of context-specific survival signaling.
Collapse
Affiliation(s)
- Dagan Segal
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
| | - Xiaoyu Wang
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX
| | | | - Divya Rajendran
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
| | - Erin Butler
- Department of Pediatrics, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bingying Chen
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
| | - Bo-Jui Chang
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
| | - Khushi Ahuja
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
| | - Averi Perny
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
| | - Kushal Bhatt
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
| | - Dana Kim Reed
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
| | | | - Jeon Lee
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
| | - Elise Jeffery
- Children's Medical Center Research Institute, UT Southwestern Medical Center, Dallas, TX
| | - Lei Wang
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, UT Southwestern Medical Center, Dallas, TX
| | - Khai Nguyen
- Department of Biophysics, UT Southwestern Medical Center, Dallas, TX
| | - Noelle S Williams
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX
| | - Stephen X Skapek
- Department of Pediatrics, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Satwik Rajaram
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
| | - Reto Fiolka
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX
| | - Khuloud Jaqaman
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
- Department of Biophysics, UT Southwestern Medical Center, Dallas, TX
| | - Gary Hon
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, UT Southwestern Medical Center, Dallas, TX
| | - James F Amatruda
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles; Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
| |
Collapse
|
49
|
Yiu SPT, Liao Y, Yan J, Weekes MP, Gewurz BE. Epstein-Barr virus BALF0/1 subverts the Caveolin and ERAD pathways to target B cell receptor complexes for degradation. Proc Natl Acad Sci U S A 2025; 122:e2400167122. [PMID: 39847318 PMCID: PMC11789056 DOI: 10.1073/pnas.2400167122] [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: 01/10/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
Epstein-Barr virus (EBV) establishes persistent infection, causes infectious mononucleosis, is a major trigger for multiple sclerosis and contributes to multiple cancers. Yet, knowledge remains incomplete about how the virus remodels host B cells to support lytic replication. We previously identified that EBV lytic replication results in selective depletion of plasma membrane (PM) B cell receptor (BCR) complexes, composed of immunoglobulin and the CD79A and CD79B signaling chains. Here, we used proteomic and biochemical approaches to identify that the EBV early lytic protein BALF0/1 is responsible for EBV lytic cycle BCR degradation. Mechanistically, an immunoglobulin heavy chain (HC) cytoplasmic tail KVK motif was required for ubiquitin-mediated BCR degradation, while CD79A and CD79B were dispensable. BALF0/1 subverted caveolin-mediated endocytosis to internalize PM BCR complexes and to deliver them to the endoplasmic reticulum. BALF0/1 stimulated immunoglobulin HC cytoplasmic tail ubiquitination, which together with the ATPase valosin-containing protein/p97 drove ER-associated degradation of BCR complexes by cytoplasmic proteasomes. BALF0/1 knockout reduced the viral load of secreted EBV particles from B cells that expressed a monoclonal antibody against EBV glycoprotein 350 but not a control anti-influenza hemagglutinin antibody and increased viral particle immunoglobulin incorporation. Consistent with downmodulation of PM BCR, BALF0/1 overexpression reduced viability of a diffuse large B cell lymphoma cell line whose survival is dependent upon BCR signaling. Collectively, our results suggest that EBV BALF0/1 downmodulates immunoglobulin upon lytic reactivation to block BCR signaling and support virion release, but await the development of suitable models to test its roles in EBV reactivation in vivo.
Collapse
Affiliation(s)
- Stephanie Pei Tung Yiu
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Graduate Program in Virology, Boston, MA02115
- Center for Integrated Solutions to Infectious Diseases, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA02142
- Department of Microbiology, Harvard Medical School, Boston, MA02115
| | - Yifei Liao
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Center for Integrated Solutions to Infectious Diseases, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA02142
- Department of Microbiology, Harvard Medical School, Boston, MA02115
| | - Jinjie Yan
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Center for Integrated Solutions to Infectious Diseases, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA02142
- Department of Microbiology, Harvard Medical School, Boston, MA02115
| | - Michael P. Weekes
- Cambridge Institute for Medical Research, University of Cambridge, CambridgeCB2 0XY, United Kingdom
| | - Benjamin E. Gewurz
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Graduate Program in Virology, Boston, MA02115
- Center for Integrated Solutions to Infectious Diseases, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA02142
- Department of Microbiology, Harvard Medical School, Boston, MA02115
| |
Collapse
|
50
|
Summer M, Riaz S, Ali S, Noor Q, Ashraf R, Khan RRM. Understanding the Dual Role of Macrophages in Tumor Growth and Therapy: A Mechanistic Review. Chem Biodivers 2025:e202402976. [PMID: 39869825 DOI: 10.1002/cbdv.202402976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/29/2025]
Abstract
Macrophages are heterogeneous cells that are the mediators of tissue homeostasis. These immune cells originated from monocytes and are classified into two basic categories, M1 and M2 macrophages. M1 macrophages exhibit anti-tumorous inflammatory reactions due to the behavior of phagocytosis. M2 macrophages or tumor-associated macrophages (TAMs) are the most abundant immune cells in the tumor microenvironment (TME) and have a basic role in tumor progression by interacting with other immune cells in TME. By the expression of various cytokines, chemokines, and growth factors, TAMs lead to strengthening tumor cell proliferation, angiogenesis, and suppression of the immune system which further support invasion and metastasis. This review discusses recent and updated mechanisms regarding tumor progression by M2 macrophages. Moreover, the current therapeutic approaches targeting TAMs, their advantages, and limitations are also summarized, and further treatment approaches are outlined along with an elaboration of the tumor regression role of macrophages. This comprehensive review article possibly helps to understand the mechanisms underlying the tumor progression and regression role of macrophages in a comparative way from a basic level to the advanced one.
Collapse
Affiliation(s)
- Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Saima Riaz
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Shaukat Ali
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Qudsia Noor
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Rimsha Ashraf
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Rana Rashad Mahmood Khan
- Faculty of Chemistry and Life Sciences, Department of Chemistry, Government College University Lahore, Lahore, Pakistan
| |
Collapse
|