1
|
Shan Y, Hu H, Yang A, Zhao W, Chu Y. An integrative approach to identifying NPC1 as a susceptibility gene for gestational diabetes mellitus. J Matern Fetal Neonatal Med 2025; 38:2445665. [PMID: 39746811 DOI: 10.1080/14767058.2024.2445665] [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: 04/24/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025]
Abstract
OBJECTIVE The objective of this study was to identify a novel gene and its potential mechanisms associated with susceptibility to gestational diabetes mellitus (GDM) through an integrative approach. METHODS We analyzed data from genome-wide association studies (GWAS) of GDM in the FinnGen R11 dataset (16,802 GDM cases and 237,816 controls) and Genotype Tissue Expression v8 expression quantitative trait locus data. We used summary-data-based Mendelian randomization to determine associations between transcript levels and phenotypes, transcriptome-wide association studies to provide insights into gene-trait associations, multi-marker analysis of genomic annotation to perform gene-based analysis, genome-wide complex trait analysis-multivariate set-based association test-combo to determine gene prioritization, and polygenic priority scores to prioritize the causal genes to screen candidate genes. Subsequent Mendelian randomization analysis was performed to infer causality between the candidate genes and GDM and phenome-wide association study (PheWAS) analysis was used to explore the associations between selected genes and other characteristics. Furthermore, to gain a deeper understanding of the functional implications of these susceptibility genes, GeneMANIA analysis was used to determine the fundamental biological functions of the therapeutic targets and protein-protein interaction network analysis was performed to identify intracellular protein interactions. RESULTS We identified two novel susceptibility genes associated with GDM: NPC1 and KIAA1191. Magnetic resonance imaging revealed a strong correlation between NPC1 expression levels and a lower incidence of GDM (odds ratio: 0.922, 95% confidence interval: 0.866-0.981, p = 0.011). PheWAS at the gene level indicated that NPC1 was not associated with any other trait. The biological significance of this gene was evidenced by its strong association with sterol metabolism. CONCLUSION Our study identified NPC1 as a novel gene whose predicted expression level is linked to a reduced risk of GDM, providing new insights into the genetic framework of this disease.
Collapse
Affiliation(s)
- Yuping Shan
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hong Hu
- Clinical medicine, Nantong University, Nantong, China
| | - Anning Yang
- Department of Obstetrics and Gynecology, Qingdao Eighth People's Hospital, Qingdao, China
| | - Wendi Zhao
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yijing Chu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
2
|
Park SJ, Kim KW, Lee EJ. Gut-brain axis and environmental factors in Parkinson's disease: bidirectional link between disease onset and progression. Neural Regen Res 2025; 20:3416-3429. [PMID: 39688568 PMCID: PMC11974660 DOI: 10.4103/nrr.nrr-d-24-00994] [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: 08/28/2024] [Revised: 10/21/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Parkinson's disease has long been considered a disorder that primarily affects the brain, as it is defined by the dopaminergic neurodegeneration in the substantia nigra and the brain accumulation of Lewy bodies containing α-synuclein protein. In recent decades, however, accumulating research has revealed that Parkinson's disease also involves the gut and uncovered an intimate and important bidirectional link between the brain and the gut, called the "gut-brain axis." Numerous clinical studies demonstrate that gut dysfunction frequently precedes motor symptoms in Parkinson's disease patients, with findings including impaired intestinal permeability, heightened inflammation, and distinct gut microbiome profiles and metabolites. Furthermore, α-synuclein deposition has been consistently observed in the gut of Parkinson's disease patients, suggesting a potential role in disease initiation. Importantly, individuals with vagotomy have a reduced Parkinson's disease risk. From these observations, researchers have hypothesized that α-synuclein accumulation may initiate in the gut and subsequently propagate to the central dopaminergic neurons through the gut-brain axis, leading to Parkinson's disease. This review comprehensively examines the gut's involvement in Parkinson's disease, focusing on the concept of a gut-origin for the disease. We also examine the interplay between altered gut-related factors and the accumulation of pathological α-synuclein in the gut of Parkinson's disease patients. Given the accessibility of the gut to both dietary and pharmacological interventions, targeting gut-localized α-synuclein represents a promising avenue for developing effective Parkinson's disease therapies.
Collapse
Affiliation(s)
- Soo Jung Park
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| | - Kyung Won Kim
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon, South Korea
| | - Eun Jeong Lee
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| |
Collapse
|
3
|
Yao C, Ma Q, Wang H, Wu B, Dai H, Xu J, Bai J, Xu F, Dube A, Wang C. Targeting myeloid cells with platelet-derived extracellular vesicles to overcome resistance of immune checkpoint blockade therapy. Biomaterials 2025; 321:123336. [PMID: 40233711 DOI: 10.1016/j.biomaterials.2025.123336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/17/2025]
Abstract
Immune checkpoint blockade (ICB) therapy is designed to boost antitumor immune responses, yet it may unintentionally alter the chemokine profile, which can attract suppressive myeloid cells to the tumor, leading to acquired immune resistance. To address this, we developed a platform that targets myeloid cells post-ICB therapy using platelet-derived extracellular vesicles (PEVs). Unlike free drug administration, this system selectively targets anti-PD-L1-treated tumors through the CXCL-CXCR2 axis, effectively redirecting myeloid cells and overcoming ICB resistance. Consequently, mice exhibited robust responses to subsequent ICB therapy cycles, resulting in significantly enhanced tumor clearance and prolonged survival. The PEVs' targeting capability was also effective in tumors treated with chemotherapy and radiotherapy, suggesting a wide range of potential applications. In summary, PEVs offer a versatile platform for targeted immunomodulation to counteract acquired immune resistance during ICB therapy.
Collapse
Affiliation(s)
- Chenlu Yao
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Qingle Ma
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Heng Wang
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Bingbing Wu
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Huaxing Dai
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jialu Xu
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jinyu Bai
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, China
| | - Fang Xu
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Admire Dube
- School of Pharmacy, University of the Western Cape, Bellville, 7535, South Africa
| | - Chao Wang
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
4
|
He H, Wen Y, Huo Q, Yu H, Liu J, Jin W, Wang Z, Wang H, Zhao Z, Cai Z. Cooperative progression of colitis and leukemia modulated by clonal hematopoiesis via PTX3/IL-1β pro-inflammatory signaling. Genes Dis 2025; 12:101397. [PMID: 40230417 PMCID: PMC11995054 DOI: 10.1016/j.gendis.2024.101397] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/30/2024] [Accepted: 07/28/2024] [Indexed: 04/16/2025] Open
Affiliation(s)
- Hang He
- National Key Laboratory of Experimental Hematology, Tianjin 300070, China
- Tianjin Key Laboratory of Inflammatory Biology, Tianjin 300070, China
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China
| | - Yuchen Wen
- National Key Laboratory of Experimental Hematology, Tianjin 300070, China
- Tianjin Key Laboratory of Inflammatory Biology, Tianjin 300070, China
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China
| | - Qingran Huo
- National Key Laboratory of Experimental Hematology, Tianjin 300070, China
- Tianjin Key Laboratory of Inflammatory Biology, Tianjin 300070, China
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China
| | - Hanzhi Yu
- National Key Laboratory of Experimental Hematology, Tianjin 300070, China
- Tianjin Key Laboratory of Inflammatory Biology, Tianjin 300070, China
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China
| | - Jingjing Liu
- National Key Laboratory of Experimental Hematology, Tianjin 300070, China
- Tianjin Key Laboratory of Inflammatory Biology, Tianjin 300070, China
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China
| | - Wenyan Jin
- National Key Laboratory of Experimental Hematology, Tianjin 300070, China
- Tianjin Key Laboratory of Inflammatory Biology, Tianjin 300070, China
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China
| | - Zhiqin Wang
- National Key Laboratory of Experimental Hematology, Tianjin 300070, China
- Tianjin Key Laboratory of Inflammatory Biology, Tianjin 300070, China
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China
| | - Huaquan Wang
- Department of Hematology, Tianjin Medical University Tianjin General Hospital, Tianjin 300070, China
| | - Zhigang Zhao
- Department of Medical Oncology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300070, China
| | - Zhigang Cai
- National Key Laboratory of Experimental Hematology, Tianjin 300070, China
- Tianjin Key Laboratory of Inflammatory Biology, Tianjin 300070, China
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology, School of Basic Medical Science, Tianjin Medical University, Tianjin 300070, China
- Department of Hematology, Tianjin Medical University Tianjin General Hospital, Tianjin 300070, China
- Department of Rheumatology and Immunology, Tianjin Medical University Tianjin General Hospital, Tianjin 300070, China
| |
Collapse
|
5
|
Churchill MJ, Pandeya A, Bauer R, Christopher T, Krug S, Honodel R, Smita S, Warner L, Mooney BM, Gibson AR, Mitchell PS, Tait Wojno ED, Rauch I. Enteric tuft cell inflammasome activation drives NKp46+ILC3 IL22 via PGD2 and inhibits Salmonella. J Exp Med 2025; 222:e20230803. [PMID: 40079814 PMCID: PMC11905811 DOI: 10.1084/jem.20230803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 12/12/2024] [Accepted: 02/10/2025] [Indexed: 03/15/2025] Open
Abstract
To distinguish pathogens from commensals, the intestinal epithelium employs cytosolic innate immune sensors. Activation of the NAIP-NLRC4 inflammasome initiates extrusion of infected intestinal epithelial cells (IEC) upon cytosolic bacterial sensing. We previously reported that activation of the inflammasome in tuft cells, which are primarily known for their role in parasitic infections, leads to the release of prostaglandin D2 (PGD2). We observe that NAIP-NLRC4 inflammasome activation in tuft cells leads to an antibacterial response with increased IL-22 and antimicrobial protein levels within the small intestine, which is dependent on PGD2 signaling. A NKp46+ subset of ILC3 expresses the PGD2 receptor CRTH2 and is the source of the increased IL-22. Inflammasome activation in tuft cells also leads to better control of Salmonella Typhimurium in the distal small intestine. However, tuft cells in the cecum and colon are dispensable for antibacterial immunity. These data support that intestinal tuft cells can also induce antibacterial responses, possibly in a tissue-specific manner.
Collapse
Affiliation(s)
- Madeline J Churchill
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Ankit Pandeya
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Renate Bauer
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
| | - Tighe Christopher
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Stefanie Krug
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Roslyn Honodel
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Shuchi Smita
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Lindsey Warner
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Bridget M Mooney
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Alexis R Gibson
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Patrick S Mitchell
- Department of Microbiology, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington , Seattle, WA, USA
| | - Elia D Tait Wojno
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Isabella Rauch
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
6
|
Tanaka M, Lum L, Hu KH, Chaudhary P, Hughes S, Ledezma-Soto C, Samad B, Superville D, Ng K, Chumber A, Benson C, Adams ZN, Kersten K, Aguilar OA, Fong L, Combes AJ, Krummel MF, Reeves MQ. Tumor cell heterogeneity drives spatial organization of the intratumoral immune response. J Exp Med 2025; 222:e20242282. [PMID: 40167599 PMCID: PMC11960709 DOI: 10.1084/jem.20242282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Intratumoral heterogeneity (ITH)-defined as genetic and cellular diversity within a tumor-is linked to failure of immunotherapy and an inferior anti-tumor immune response. We modeled heterogeneous tumors comprised of "hot" and "cold" tumor populations (giving rise to T cell-rich and T cell-poor tumors, respectively) and introduced fluorescent labels to enable precise spatial tracking. We found the cold tumor cell population exerted a "dominant cold" effect in mixed tumors. Strikingly, spatial analysis revealed that the tumor cells themselves created distinct local microenvironments within heterogeneous tumors: regions occupied by cold tumor cells showed pronounced immunosuppression, harboring increased CD206Hi macrophages and diminished local T cell function. This inferior T cell activity in cold regions persisted even after immunotherapy and mechanistically was mediated by CX3CL1 produced by the cold tumor cells. An immune cold tumor population within a heterogeneous tumor thus impairs tumor immunity on both a tumor-wide and a highly localized spatial scale.
Collapse
Affiliation(s)
- Miho Tanaka
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Lotus Lum
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Kenneth H. Hu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The James P. Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Piyush Chaudhary
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Savannah Hughes
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Cecilia Ledezma-Soto
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Bushra Samad
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
| | - Daphne Superville
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| | - Kenneth Ng
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Arun Chumber
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Ciara Benson
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Zoe N. Adams
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Kelly Kersten
- Cancer Metabolism and Microenvironment Program, NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, LA Jolla, CA, USA
| | - Oscar A. Aguilar
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
- ImmunoX Initiative, University of California San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California San Francisco, San Francisco, CA, USA
| | - Lawrence Fong
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Alexis J. Combes
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
- ImmunoX Initiative, University of California San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Matthew F. Krummel
- ImmunoX Initiative, University of California San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Melissa Q. Reeves
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
7
|
Zhan Y, Deng Q, Jia Y, Chen Z, Zhao X, Ling Y, Qiu Y, Wang X, Wang F, He M, Huang W, Shen J, Wen S. Pdia3 deficiency exacerbates intestinal injury by disrupting goblet and Paneth cell function during ischemia/reperfusion. Cell Signal 2025; 130:111682. [PMID: 39988288 DOI: 10.1016/j.cellsig.2025.111682] [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: 12/24/2024] [Revised: 01/27/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025]
Abstract
Intestinal ischemia/reperfusion (I/R) injury is a severe medical condition associated with high mortality rates due to its disruption of intestinal homeostasis and impairment of mucosal defenses. The intestinal epithelium, particularly goblet and Paneth cells, plays a critical role in maintaining gut barrier integrity. Protein disulfide isomerase A3 (PDIA3) is involved in protein folding within intestinal epithelial cells (IECs) and has been linked to the stress response during I/R injury. This study aims to explore the role of PDIA3 in preserving intestinal integrity and immune function during I/R injury. Our study employed both human and mouse models to investigate PDIA3's expression and function. The correlation between PDIA3 expression and disease severity was analyzed using statistical tests, including Pearson's correlation coefficient. An intestinal I/R model was established in intestinal epithelium-specific conditional knockout mice lacking the Pdia3 gene. Single-cell RNA sequencing, immunohistochemistry, and transcriptomic analysis were used to assess PDIA3 expression in various intestinal cell types and to evaluate its role in epithelial differentiation and immune responses. PDIA3 was found to be highly expressed in healthy IECs, especially in goblet and Paneth cells. Its expression was reduced in patients with mesenteric artery ischemia and Pdia3-deficient mice, leading to severe intestinal damage, including impaired goblet and Paneth cell function, reduced antimicrobial peptide production, and altered gut microbiota. Treatment with recombinant defensin α1, an antimicrobial peptide secreted by Paneth cells, significantly alleviated the adverse effects of Pdia3 deficiency, restoring gut microbiota balance and reducing inflammation in the intestinal I/R injury mice. Taken together, our findings suggest that Pdia3 plays a vital role in maintaining intestinal barrier function and immune defense. Its deficiency exacerbates I/R-induced intestinal damage by impairing epithelial differentiation, mucus production, and antimicrobial peptide secretion. Targeting Pdia3 and associated pathways offers promising therapeutic strategies for mitigating I/R injury and restoring intestinal homeostasis.
Collapse
Affiliation(s)
- Yaqing Zhan
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Qiwen Deng
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yifan Jia
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhaorong Chen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Xu Zhao
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yihong Ling
- State Key Laboratory of Oncology in South, China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuxin Qiu
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiwen Wang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Fan Wang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Muchen He
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wenqi Huang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Jiantong Shen
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Shihong Wen
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Department of Anesthesiology, Guangxi Hospital Division of the First Affiliated Hospital of Sun Yat-sen University, Nanning, China.
| |
Collapse
|
8
|
Guequen A, Tapia-Balladares B, Apablaza T, Guidone D, Cárcamo-Lemus N, Villanueva S, Sandoval PY, Galietta LJV, Flores CA. Sodium-Coupled Monocarboxylate Absorption in the Airway Epithelium Is Facilitated by the SLC5A8 Co-Transporter. Acta Physiol (Oxf) 2025; 241:e70051. [PMID: 40326639 DOI: 10.1111/apha.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/27/2025] [Accepted: 04/17/2025] [Indexed: 05/07/2025]
Abstract
AIM Amino acids, sugars, short-chain fatty acids (SCFA), vitamins, and other small molecules compose the extracellular metabolome on the airway lumen surface, but how the airway epithelium deals with these molecules has not been deeply studied. Due to the broad spectrum of metabolites transported by SLC5A8 and SLC5A12, we aim to determine if they are functionally expressed and participate in the absorption of Na+, short-chain fatty acids, and monocarboxylates in mouse and human airway epithelium. METHODS Tracheas isolated from male or female mice and human bronchial epithelial cells (HBECs) were used for electrophysiological studies in the Ussing chamber and to detect members of the SLC16 family by RT-PCR and bulk RNAseq. Additionally, cell lines expressing the human and murine SLC5A8 transporter were employed for uptake studies using a fluorescent lactate probe. RESULTS We showed for the first time that human and murine airway epithelium express a functional SLC5A8 transporter, facilitating the absorption of glucose metabolites and SCFAs. The Na+-coupled monocarboxylate transport was not additive with ENaC-mediated Na+ absorption in mouse trachea. We observed that valproate acts as an inhibitor of the murine but not of the human SLC5A8 transporter. CONCLUSIONS Our results demonstrate that several metabolites derived from bacterial and cellular metabolism can be transported from the airway lumen into the epithelial cells, participating in a homeostatic relation of the tissue with its environment.
Collapse
Affiliation(s)
- Anita Guequen
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Universidad Austral de Chile, Valdivia, Chile
| | | | - Tábata Apablaza
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Estudiante Programa de Doctorado en Enfermedades Crónicas, Facultad de Medicina, Universidad San Sebastián, Valdivia, Chile
| | - Daniela Guidone
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | | | - Sandra Villanueva
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Facultad de Medicina, Universidad San Sebastián, Valdivia, Chile
| | - Pamela Y Sandoval
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Facultad de Medicina, Universidad San Sebastián, Valdivia, Chile
| | - Luis J V Galietta
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Department of Translational Medical Sciences (DISMET), University of Napoli "Federico II", Napoli, Italy
| | - Carlos A Flores
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Facultad de Medicina, Universidad San Sebastián, Valdivia, Chile
| |
Collapse
|
9
|
Duy PQ, Dylik B, Deniz E. Precision medicine in the pediatric and neonatal intensive care units through genomics. Curr Opin Pediatr 2025; 37:211-215. [PMID: 40298123 PMCID: PMC12055474 DOI: 10.1097/mop.0000000000001471] [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] [Indexed: 04/30/2025]
Abstract
PURPOSE OF REVIEW Genome-wide sequencing technologies have revolutionized the understanding of human disorders and advanced precision medicine, especially for pediatric disorders. Here, we discuss the utility of genomic technologies in advancing the care of children admitted to the pediatric and neonatal intensive care units. RECENT FINDINGS Rapid molecular diagnosis permitted by genomic medicine has yielded clinically actionable findings that influence decision-making and facilitate timely therapeutic interventions. Identifying a genetic association provides a causal anchor to understanding disease biology at the single nucleotide resolution, revealing hidden biological heterogeneity that may be obscured by traditional imaging, laboratory, and pathological workup. The importance of a genetic diagnosis is further highlighted by the promise of gene therapy to correct the underlying genetic perturbation, as evidenced by the recent emergence of FDA-approved gene therapies for childhood genetic conditions. SUMMARY We predict that whole-genome sequencing, in conjunction with other omic technologies, will become critical diagnostic adjuncts in the clinical workup of critically ill children.
Collapse
Affiliation(s)
- Phan Q. Duy
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Benjamin Dylik
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Engin Deniz
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
10
|
Wang Y, Zhang Y, An X, Jiang Y, Wang F. New LFA-1 inhibitor Orientin reduces angiotensin II-induced vascular remodeling. Eur J Pharmacol 2025; 995:177426. [PMID: 39993698 DOI: 10.1016/j.ejphar.2025.177426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 02/21/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
BACKGROUND The interaction between monocytes and vascular endothelium often leads to inflammatory reactions and vascular remodeling. The lymphocyte function-associated antigen 1 (LFA1) plays a crucial role in promoting leukocyte adhesion and migration during inflammatory diseases. However, the role of LFA1 in angiotensin II (Ang II)-induced hypertension and vascular remodeling is not clear. Orientin (Ori) has antioxidant, anti-inflammatory, anticancer, and resistance to myocardial remodeling. However, the role of Orientin remains unclear in angiotensin II (Ang II)-induced hypertension and vascular remodeling. METHODS In this study, Ang II was used to induce hypertension in mice. We employed various techniques including blood pressure monitoring, pathological staining, Immunofluorescent and Immunohistochemical staining, real time-PCR, vasodilation analysis and other methods to study whether LFA1 antibody and Orientin can regulate vascular remoding. RESULTS Our results showed that LFA1 significantly improved Ang II-induced hypertension, inflammation, fibrosis, and oxidative stress. Furthermore, in vitro experiments have substantiated that the use of neutralizing antibodies targeting LFA1 can effectively hinder the migration of macrophages to endothelial cells, which is triggered by Ang II. Additionally, the antibodies also reduce the extent of DNA damage and oxidative stress. Orientin can interact with LFA-1. Then, it was proved by pathological staining that Orientin can inhibit Ang II-induced vascular remodeling. CONCLUSION Here, we identified Orientin as a small molecule inhibitor of LFA-1 with anti-vascular remodeling function. These findings not only suggest that Orientin is a promising compound for the clinical treatment of vascular injury and hypertension, but also provide strategies for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Yitong Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China, No.49 Huayuanbei Road, Beijing, 100191, China; Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ying Zhang
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiangbo An
- Department of Interventional Therapy, First Affiliated Hospital of Dalian Medical University, Dalian, China; Department of Cardiology, An Zhen Hospital, Capital Medical University, No.2 Anzhen Road, Chao Yang district, Beijing, 100029, China.
| | - Yinong Jiang
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Feng Wang
- Department of Interventional Therapy, First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
11
|
Sun F, Desevin K, Fu Y, Parameswaran S, Mayall J, Rinaldi V, Krietenstein N, Manukyan A, Yin Q, Galan C, Yang CH, Shindyapina AV, Gladyshev VN, Garber M, Schjenken JE, Rando OJ. A single cell atlas of the mouse seminal vesicle. G3 (BETHESDA, MD.) 2025; 15:jkaf045. [PMID: 40036847 DOI: 10.1093/g3journal/jkaf045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 02/16/2025] [Indexed: 03/06/2025]
Abstract
During mammalian reproduction, sperm are delivered to the female reproductive tract bathed in a complex medium known as seminal fluid, which plays key roles in signaling to the female reproductive tract and in nourishing sperm for their onwards journey. Along with minor contributions from the prostate and the epididymis, the majority of seminal fluid is produced by a somewhat understudied organ known as the seminal vesicle. Here, we report the first single-cell RNA-seq atlas of the mouse seminal vesicle, generated using tissues obtained from 23 mice of varying ages, exposed to a range of dietary challenges. We define the transcriptome of the secretory cells in this tissue, identifying a relatively homogeneous population of the epithelial cells which are responsible for producing the majority of seminal fluid. We also define the immune cell populations-including large populations of macrophages, dendritic cells, T cells, and NKT cells-which have the potential to play roles in producing the various immune mediators present in seminal plasma. Together, our data provide a resource for understanding the composition of an understudied reproductive tissue, with potential implications for paternal control of offspring development and metabolism.
Collapse
Affiliation(s)
- Fengyun Sun
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Kathleen Desevin
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Yu Fu
- Department of Systems Biology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Shanmathi Parameswaran
- Hunter Medical Research Institute Infertility and Reproduction Research Program, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2305, Australia
| | - Jemma Mayall
- Immune Health Program, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW 2305, Australia
| | - Vera Rinaldi
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Nils Krietenstein
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Artür Manukyan
- Department of Bioinformatics and Integrative Biology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Qiangzong Yin
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Carolina Galan
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Chih-Hsiang Yang
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Anastasia V Shindyapina
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Manuel Garber
- Department of Bioinformatics and Integrative Biology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - John E Schjenken
- Hunter Medical Research Institute Infertility and Reproduction Research Program, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, Callaghan, NSW 2305, Australia
| | - Oliver J Rando
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| |
Collapse
|
12
|
Yue S, Gong L, Tan Y, Zhang X, Liao F. IsoalloLCA-intervened regulatory T cell exosomes alleviate inflammatory bowel disease by inhibiting NF-κB-associated inflammation in intestinal epithelial cells. Int Immunopharmacol 2025; 154:114501. [PMID: 40174336 DOI: 10.1016/j.intimp.2025.114501] [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: 11/17/2024] [Revised: 02/11/2025] [Accepted: 03/14/2025] [Indexed: 04/04/2025]
Abstract
Regulatory T cells (Tregs) are the principal immune cells that exert anti-inflammatory effects within the organism. Their exosomes exhibit therapeutic efficacy across a broad spectrum of diseases owing to their high stability, low immunogenicity, and substantial penetration capacity. Recent research have indicated that isoallolithocholic acid (isoalloLCA), a metabolite associated with bile acid metabolism, may enhance Treg activity by upregulating forkhead box protein3 (Foxp3) expression. Hence, metabolite-based strategies for reinforcing Tregs may offer novel intervention options for treating related diseases. In this study, tumor necrosis factor (TNF)-α and dextran sulfate sodium (DSS) were employed to establish cellular and animal models of inflammatory bowel disease (IBD), further evaluating the therapeutic efficacy of isoalloLCA-intervened regulatory T cell exosomes (isoalloLCA-Exo) within this model. Our findings demonstrated that isoalloLCA-Exo effectively inhibit colitis progression in a murine model, as indicated by reduced inflammation, decreased apoptosis of intestinal epithelial cells, and improved intestinal barrier function. Furthermore, in vitro analyses elucidated the molecular mechanisms underlying the anti-inflammatory effects of isoalloLCA-Exo, revealing that the intervention effectively reversed TNF-α-induced inflammation and apoptosis in intestinal epithelial cells by modulating the NF-κB pathway. In conclusion, isoalloLCA-Exo can decelerate inflammatory bowel disease progression and suppress inflammatory response in intestinal epithelial cells by inhibiting NF-κB pathway. Notably, isoalloLCA-Exo exhibit superior efficacy to the traditional drug mesalazine and conventional treg exosome(NC-Exo). These findings have significant implications for optimizing Treg-derived exosome-based therapies for inflammation-related diseases.
Collapse
Affiliation(s)
- Simei Yue
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lingjiao Gong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yulin Tan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaodan Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fei Liao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Wuhan University Shenzhen Research Institute, Shenzhen, Guangdong 518000, China; Central Laboratory of Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
13
|
Liu C, Peng H, Yu J, Luo P, Xiong C, Chen H, Fan H, Ma Y, Ou W, Zhang S, Yang C, Zhao L, Zhang Y, Guo X, Ke Q, Wang T, Deng C, Li W, Xiang AP, Xia K. Impaired ketogenesis in Leydig Cells drives testicular aging. Nat Commun 2025; 16:4224. [PMID: 40328805 DOI: 10.1038/s41467-025-59591-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 04/17/2025] [Indexed: 05/08/2025] Open
Abstract
Testicular aging commonly leads to testosterone deficiency and impaired spermatogenesis, yet the underlying mechanisms remain elusive. Here, we show that Leydig cells are particularly vulnerable to aging processes in testis. Single-cell RNA sequencing identifies the expression of Hmgcs2, the gene encoding rate-limiting enzyme of ketogenesis, decreases significantly in Leydig cells from aged mice. Additionally, the concentrations of ketone bodies β-hydroxybutyric acid and acetoacetic acid in young testes are substantially higher than that in serum, but significantly diminish in aged testes. Silencing of Hmgcs2 in young Leydig cells drives cell senescence and accelerated testicular aging. Mechanistically, β-hydroxybutyric acid upregulates the expression of Foxo3a by facilitating histone acetylation, thereby mitigating Leydig cells senescence and promoting testosterone production. Consistently, enhanced ketogenesis by genetic manipulation or oral β-hydroxybutyric acid supplementation alleviates Leydig cells senescence and ameliorates testicular aging in aged mice. These findings highlight defective ketogenesis as a pivotal factor in testicular aging, suggesting potential therapeutic avenues for addressing age-related testicular dysfunction.
Collapse
Affiliation(s)
- Congyuan Liu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hao Peng
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiajie Yu
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Peng Luo
- Reproductive Medicine Center, The First Affiliated Hospital, Sun Yat-sen University, The Key Laboratory for Reproductive Medicine of Guangdong Province, Guangzhou, Guangdong, China
| | - Chuanfeng Xiong
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hong Chen
- Center for Stem Cells Translational Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong, China
- Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Hang Fan
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuanchen Ma
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wangsheng Ou
- State Key Laboratory of Ophthalmology, Zhong Shan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Suyuan Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Cuifeng Yang
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lerong Zhao
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuchen Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaolu Guo
- Center for Stem Cells Translational Medicine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong, China
- Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Qiong Ke
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tao Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chunhua Deng
- Department of Urology and Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Weiqiang Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China.
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Kai Xia
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China.
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
14
|
Gao Y, Peng Y, Zhou Y, Zhu J, Fu S, Chen Y, Cai C, Han Y, Shen H, Zeng S, Mao L, Xiao Z. Mitochondrial gene SLC25A24 regulated anti-tumor immunity and inhibited the proliferation and metastasis of colorectal cancer by PKG1-dependent cGMP/PKG1 pathway. Int Immunopharmacol 2025; 157:114664. [PMID: 40334626 DOI: 10.1016/j.intimp.2025.114664] [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: 01/29/2025] [Revised: 03/30/2025] [Accepted: 04/09/2025] [Indexed: 05/09/2025]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related mortality globally, with metastasis playing a key role in its unfavorable prognosis. Emerging research has emphasized the pivotal role of mitochondria in tumor immune regulation. Nevertheless, the clinical relevance and functional role of SLC25A24, a mitochondrial solute carrier, in CRC remain largely unexplored. Through bioinformatics analyses and validation in clinical cohorts, this study identifies SLC25A24 as an independent prognostic marker in CRC, significantly linked to immune infiltration in CRC tissues. Our findings demonstrated that SLC25A24 expression is markedly reduced in CRC cell lines and tissues. Kaplan-Meier survival analysis revealed that lower SLC25A24 expression is associated with worse overall survival and progression-free survival in CRC patients. Interestingly, SLC25A24 expression was higher in microsatellite instability (MSI) CRC, which shows greater responsiveness to immune checkpoint inhibitors (ICIs). Functional experiments indicated that SLC25A24 overexpression suppresses CRC cell proliferation, migration, and invasion. Mechanistic studies revealed that SLC25A24 positively regulates the cGMP/PKG1 signaling pathway in CRC, influencing mitochondrial potential, apoptosis, and proliferation-related markers. This research highlights the SLC25A24-PKG1 axis as a potential therapeutic target to bolster anti-tumor immunity and curb CRC progression.
Collapse
Affiliation(s)
- Yan Gao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Yinghui Peng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yulai Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Microbiology, Immunology & Molecular Genetics, University of Texas Long School of Medicine, UT Health Science Center, San Antonio, TX 78229, USA
| | - Jiang Zhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shenao Fu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yihong Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Lei Mao
- Department of Oncology, Yueyang People's Hospital, Yueyang Hospital Afliated to Hunan Normal University, Yueyang 414000, Hunan, China.
| | - Zemin Xiao
- Department of Oncology, Changde Hospital, Xiangya School of Medicine, Central South University 818 Renmin Rd, Wuling District, Changde, Hunan 415000, China.
| |
Collapse
|
15
|
Tümmler B, Pallenberg ST, Dittrich AM, Graeber SY, Naehrlich L, Sommerburg O, Mall MA. Progress of personalized medicine of cystic fibrosis in the times of efficient CFTR modulators. Mol Cell Pediatr 2025; 12:6. [PMID: 40320452 PMCID: PMC12050259 DOI: 10.1186/s40348-025-00194-0] [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: 01/20/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Cystic fibrosis (CF) is a systemic disorder of exocrine glands that is caused by mutations in the CFTR gene. MAIN BODY The basic defect in people with CF (pwCF) leads to impaired epithelial transport of chloride and bicarbonate that can be assessed by CFTR biomarkers, i.e. the β-adrenergic sweat rate and sweat chloride concentration (SCC), chloride conductance of the nasal respiratory epithelium (NPD), urine secretion of bicarbonate, intestinal current measurements (ICM) of chloride secretory responses in rectal biopsies and in bioassays of chloride transport in organoids or cell cultures. CFTR modulators are a novel class of drugs that improve defective posttranslational processing, trafficking and function of mutant CFTR. By April 2025, triple combination therapy with the CFTR potentiator ivacaftor (IVA) and the CFTR correctors elexacaftor (ELX) and tezacaftor (TEZ) has been approved in Europe for the treatment of all pwCF who do not carry two minimal function CFTR mutations. Previous phase 3 and post-approval phase 4 studies in pwCF who harbour one or two alleles of the major mutation F508del consistently reported significant improvements of lung function and anthropometry upon initiation of ELX/TEZ/IVA compared to baseline. Normalization of SCC, NPD and ICM correlated with clinical outcomes on the population level, but the restoration of CFTR function was diverse and not predictive for clinical outcome in the individual patient. Theratyping of non-F508del CF genotypes in patient-derived organoids and cell cultures revealed for most cases clinically meaningful increases of CFTR activity upon exposure to ELX/TEZ/IVA. Likewise, every second CF patient with non-F508del genotypes improved in SCC and clinical outcome upon exposure to ELX/TEZ/IVA indicating that triple CFTR modulator therapy is potentially beneficial for all pwCF who do not carry two minimal function CFTR mutations. This group who is not eligible for CFTR modulators may opt for gene addition therapy in the future, as the first-in-human trial with a recombinant lentiviral vector is underway. FUTURE DIRECTIONS The upcoming generation of pwCF will probably experience a rather normal life in childhood and adolescence. To classify the upcoming personal signatures of CF disease in the times of efficient modulators, we need more sensitive CFTR biomarkers that address the long-term course of airway and gut microbiome, host defense, epithelial homeostasis and multiorgan metabolism.
Collapse
Affiliation(s)
- Burkhard Tümmler
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, 30625, Germany.
- German Center for Lung Research, Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover Medical School, Hannover, Germany.
| | - Sophia Theres Pallenberg
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, 30625, Germany
| | - Anna-Maria Dittrich
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, 30625, Germany
- German Center for Lung Research, Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover Medical School, Hannover, Germany
| | - Simon Y Graeber
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, and Cystic Fibrosis Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | - Lutz Naehrlich
- Department of Pediatrics, Justus Liebig University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany
| | - Olaf Sommerburg
- Division of Pediatric Pneumology and Allergy, and Cystic Fibrosis Center, Department of Pediatrics, University of Heidelberg, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL),, University of Heidelberg, Heidelberg, Germany
| | - Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, and Cystic Fibrosis Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| |
Collapse
|
16
|
Banjac I, Maimets M, Tsang IHC, Dioli M, Hansen SL, Krizic K, Bressan RB, Lövkvist C, Jensen KB. Fate mapping in mouse demonstrates early secretory differentiation directly from Lgr5+ intestinal stem cells. Dev Cell 2025; 60:1281-1289.e6. [PMID: 39793582 DOI: 10.1016/j.devcel.2024.12.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 08/10/2024] [Accepted: 12/11/2024] [Indexed: 01/13/2025]
Abstract
The intestinal epithelium has a remarkably high turnover in homeostasis. It remains unresolved how this is orchestrated at the cellular level and how the behavior of stem and progenitor cells ensures tissue maintenance. To address this, we combined quantitative fate mapping in three complementary mouse models with mathematical modeling and single-cell RNA sequencing. Our integrated approach generated a spatially and temporally defined model of crypt maintenance based on two cycling populations: stem cells at the crypt-bottom and transit-amplifying (TA) cells above them. Subsequently, we validated the predictions from the mathematical model, demonstrating that fate decisions between the secretory and absorptive lineages are made within the stem cell compartment, whereas TA cell divisions contribute specifically to the absorptive lineage. These quantitative insights provide further direct evidence for crypt-bottom stem cells as the dominant driver of the intestinal epithelium replenishment.
Collapse
Affiliation(s)
- Isidora Banjac
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Martti Maimets
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Ingrid H C Tsang
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Marius Dioli
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Stine Lind Hansen
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Kata Krizic
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Raul Bardini Bressan
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Cecilia Lövkvist
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark.
| | - Kim B Jensen
- Novo Nordisk Foundation Center for Stem Cell Medicine, reNEW, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark.
| |
Collapse
|
17
|
Hu DJK, Cai XT, Simons J, Yun J, Elstrott J, Jasper H. Non-canonical Wnt signaling promotes epithelial fluidization in the repairing airway. Nat Commun 2025; 16:4124. [PMID: 40319020 PMCID: PMC12049509 DOI: 10.1038/s41467-025-59320-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 04/17/2025] [Indexed: 05/07/2025] Open
Abstract
Concerted migration of basal stem cells (BCs) in the airway, also known as epithelial fluidization, has been implicated in epithelial repair after injury. How BC migration is regulated, and how it influences the success of epithelial repair, remains unclear. Here we have identified non-canonical Wnt signaling through Ptk7, Fzd7, and YAP as a critical regulator of BC migration in the mouse trachea. Using live imaging and genetic studies in the mouse, we find that Ptk7 is required for the concerted movement of BCs after injury, and that this requirement extends to BC proliferation and subsequent restoration of epithelial homeostasis after injury. We demonstrate that Ptk7 exerts this function in conjunction with Wnt5a and Fzd7, and through YAP activation in BCs. Our data provide mechanistic insight into the regulation of epithelial repair in the airway.
Collapse
Affiliation(s)
- Daniel Jun-Kit Hu
- Regenerative Medicine, Genentech Inc., South San Francisco, CA, USA.
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA.
| | - Xiaoyu Tracy Cai
- Regenerative Medicine, Genentech Inc., South San Francisco, CA, USA
- Department of Medicine, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Jesse Simons
- Regenerative Medicine, Genentech Inc., South San Francisco, CA, USA
| | - Jina Yun
- Regenerative Medicine, Genentech Inc., South San Francisco, CA, USA
| | - Justin Elstrott
- Translational Imaging, Genentech Inc., South San Francisco, CA, USA
| | - Heinrich Jasper
- Regenerative Medicine, Genentech Inc., South San Francisco, CA, USA.
| |
Collapse
|
18
|
Cao M, Deng Y, Hao Q, Yan H, Wang QL, Dong C, Wu J, He Y, Huang LB, Xia X, Gao Y, Chen HN, Zhang WH, Zhang YJ, Zhuo X, Dai L, Hu H, Peng Y, Zhang F, Liu Z, Huang W, Zhang H, Yang L, Shu Y, Zhang W, Zhang Y, Xu H. Single-cell transcriptomic analysis reveals gut microbiota-immunotherapy synergy through modulating tumor microenvironment. Signal Transduct Target Ther 2025; 10:140. [PMID: 40312419 PMCID: PMC12045981 DOI: 10.1038/s41392-025-02226-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 05/03/2025] Open
Abstract
The gut microbiota crucially regulates the efficacy of immune checkpoint inhibitor (ICI) based immunotherapy, but the underlying mechanisms remain unclear at the single-cell resolution. Using single-cell RNA sequencing and subsequent validations, we investigate gut microbiota-ICI synergy by profiling the tumor microenvironment (TME) and elucidating critical cellular interactions in mouse models. Our findings reveal that intact gut microbiota combined with ICIs may synergistically increase the proportions of CD8+, CD4+, and γδ T cells, reduce glycolysis metabolism, and reverse exhausted CD8+ T cells into memory/effector CD8+ T cells, enhancing antitumor response. This synergistic effect also induces macrophage reprogramming from M2 protumor Spp1+ tumor-associated macrophages (TAMs) to Cd74+ TAMs, which act as antigen-presenting cells (APCs). These macrophage subtypes show a negative correlation within tumors, particularly during fecal microbiota transplantation. Depleting Spp1+ TAMs in Spp1 conditional knockout mice boosts ICI efficacy and T cell infiltration, regardless of gut microbiota status, suggesting a potential upstream role of the gut microbiota and highlighting the crucial negative impact of Spp1+ TAMs during macrophage reprogramming on immunotherapy outcomes. Mechanistically, we propose a γδ T cell-APC-CD8+ T cell axis, where gut microbiota and ICIs enhance Cd40lg expression on γδ T cells, activating Cd40 overexpressing APCs (e.g., Cd74+ TAMs) through CD40-CD40L-related NF-κB signaling and boosting CD8+ T cell responses via CD86-CD28 interactions. These findings highlight the potential importance of γδ T cells and SPP1-related macrophage reprogramming in activating CD8+ T cells, as well as the synergistic effect of gut microbiota and ICIs in immunotherapy through modulating the TME.
Collapse
Affiliation(s)
- Minyuan Cao
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yun Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Hao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Huayun Yan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Quan-Lin Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China
| | - Chunyan Dong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yajiao He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Li-Bin Huang
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xuyang Xia
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Gastrointestinal Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yongchao Gao
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China
| | - Hai-Ning Chen
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Wei-Han Zhang
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yan-Jing Zhang
- Core Facilities, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaozhen Zhuo
- Department of Cardiology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Lunzhi Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hongbo Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Feng Zhang
- Center for Precision Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Zhaoqian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China
| | - Weihua Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China
| | - Huiyuan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Shu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central Laboratory of Hunan Cancer Hospital, Central South University, Changsha, China.
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
| | - Yan Zhang
- Lung Cancer Center/Lung Cancer Institute, Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, China.
| | - Heng Xu
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
- Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, China.
- Tianfu Jincheng Laboratory, Chengdu, China.
| |
Collapse
|
19
|
Domènech-Moreno E, Lim WW, Montrose MG, Sévigny M, Brandt A, Lemmetyinen TT, Viitala EW, Mäkelä TP, Cook SA, Ollila S. Interleukin-11 expressed in the polyp-enriched fibroblast subset is a potential therapeutic target in Peutz-Jeghers syndrome. J Pathol 2025; 266:66-80. [PMID: 40070038 DOI: 10.1002/path.6408] [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: 07/24/2024] [Revised: 12/12/2024] [Accepted: 01/17/2025] [Indexed: 04/12/2025]
Abstract
Peutz-Jeghers syndrome (PJS) is associated with early-onset gastrointestinal polyposis caused by hereditary inactivating pathogenic variants in the tumor suppressor gene STK11 (LKB1). Due to lack of prophylactic therapies, management of PJS polyps requires frequent surveillance. Interestingly, studies in mouse models have revealed that stromal cells drive the polyp formation, but detailed understanding of the cell types and interactions involved has been lacking. Using single-cell RNA sequencing of PJS mouse model polyps, we here identify a polyp-enriched crypt top fibroblast (pCTF) cluster characterized by a transcriptional signature also enriched in PJS patient polyps. The pCTF signature was also noted in primary fibroblasts in vitro following acute STK11 loss. Targeted deletion of Stk11 in crypt top fibroblasts using Foxl1-Cre led to upregulation of the pCTF signature genes and later to polyposis. pCTFs displayed similarity to inflammation-associated fibroblasts, and polyposis was exacerbated by inflammation. Cell-cell communication analysis identified interleukin 11 (IL-11) as a potential pCTF inducer, and consistent with this, IL-11 was required for fibroblast reprogramming toward pCTFs following STK11 loss. Importantly, a neutralizing IL-11 antibody efficiently reduced polyp formation in a PJS model indicating a key, targetable role for IL-11 in polyp development. Together the results characterize pCTFs as a PJS polyp-enriched fibroblast subset and identify IL-11 as a key mediator of fibroblast reprogramming and a potential therapeutic target in PJS. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Eva Domènech-Moreno
- HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Wei-Wen Lim
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Melissa G Montrose
- HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Myriam Sévigny
- Translational Cancer Medicine Program, University of Helsinki, Helsinki, Finland
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Anders Brandt
- HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Toni T Lemmetyinen
- Translational Cancer Medicine Program, University of Helsinki, Helsinki, Finland
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Emma W Viitala
- Translational Cancer Medicine Program, University of Helsinki, Helsinki, Finland
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Tomi P Mäkelä
- HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Stuart A Cook
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
- MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, London, UK
| | - Saara Ollila
- Translational Cancer Medicine Program, University of Helsinki, Helsinki, Finland
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| |
Collapse
|
20
|
Maurice MM, Angers S. Mechanistic insights into Wnt-β-catenin pathway activation and signal transduction. Nat Rev Mol Cell Biol 2025; 26:371-388. [PMID: 39856369 DOI: 10.1038/s41580-024-00823-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 01/27/2025]
Abstract
In multicellular organisms, Wnt proteins govern stem and progenitor cell renewal and differentiation to regulate embryonic development, adult tissue homeostasis and tissue regeneration. Defects in canonical Wnt signalling, which is transduced intracellularly by β-catenin, have been associated with developmental disorders, degenerative diseases and cancers. Although a simple model describing Wnt-β-catenin signalling is widely used to introduce this pathway and has largely remained unchanged over the past 30 years, in this Review we discuss recent studies that have provided important new insights into the mechanisms of Wnt production, receptor activation and intracellular signalling that advance our understanding of the molecular mechanisms that underlie this important cell-cell communication system. In addition, we review the recent development of molecules capable of activating the Wnt-β-catenin pathway with selectivity in vitro and in vivo that is enabling new lines of study to pave the way for the development of Wnt therapies for the treatment of human diseases.
Collapse
Affiliation(s)
- Madelon M Maurice
- Center for Molecular Medicine, University Medical Center, Utrecht, Netherlands.
- Oncode Institute, Utrecht, Netherlands.
| | - Stephane Angers
- Donnelly Centre for Cellular and Biomolecular Research and Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
21
|
Liu Z, Jiang X, Ke Z, Wang W, Tang J, Dai Y. PAR2 deficiency impairs antitumor immunity and attenuates anti-PD1 efficacy in colorectal cancer. Pharmacol Res 2025; 215:107721. [PMID: 40174816 DOI: 10.1016/j.phrs.2025.107721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
A T cell-inflamed tumor microenvironment is predictive of better prognosis and clinical response to immunotherapy. Proteinase-activated receptor 2 (PAR2), a member of G-protein coupled receptors is involved in inflammatory process and the progression of various cancers. However, the role of PAR2 in modulating the tumor microenvironment remains unclear. Here, we found that PAR2 high-expression was associated with a favorable prognosis in patients with colorectal cancer. Intriguingly, PAR2 expression in human colorectal cancer was mainly confined to tumor cells and was significantly associated with CD8+ T cell infiltration. Tumor-intrinsic PAR2 deficiency blunted antitumor immune responses to promote tumor growth and attenuated the therapeutic efficacy of anti-PD1 in a mouse model of colon cancer. Tumors with downregulated PAR2 showed decreased CD8+ T cell infiltration and impaired effector function. Mechanistically, PAR2 activation in tumor cells induced CXCL9 and CXCL10 production via PI3K/AKT/mTOR signaling, thereby enhancing CD8+ T cell recruitment in the tumor microenvironment. In addition, PAR2 was essential for dendritic cell activation and differentiation towards conventional type 1 subset. PAR2 deficiency in dendritic cells markedly impaired their ability to prime CD8+ T cells and control tumor growth in vivo. Thus, our findings identify new roles for PAR2 in promoting antitumor immunity and provide a promising target to improve immunotherapy efficacy in colorectal cancer.
Collapse
Affiliation(s)
- Zilin Liu
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Xuehui Jiang
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Ziliang Ke
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Weihong Wang
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Jianqiang Tang
- Department of General Surgery, Peking University First Hospital, Beijing, China.
| | - Yun Dai
- Department of Gastroenterology, Peking University First Hospital, Beijing, China.
| |
Collapse
|
22
|
Gulbransen BD, Heuckeroth RO. Could Diverse Subtypes of Canonical and Schwann-like Enteric Glia Explain Diverse Glial Functions? Gastroenterology 2025; 168:874-876. [PMID: 39788210 DOI: 10.1053/j.gastro.2024.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 12/26/2024] [Indexed: 01/12/2025]
Affiliation(s)
- Brian D Gulbransen
- Department of Physiology, Michigan State University, East Lansing, Michigan.
| | - Robert O Heuckeroth
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The Children's Hospital of Philadelphia Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
23
|
Mazzilli SA, Rahal Z, Rouhani MJ, Janes SM, Kadara H, Dubinett SM, Spira AE. Translating premalignant biology to accelerate non-small-cell lung cancer interception. Nat Rev Cancer 2025; 25:379-392. [PMID: 39994467 DOI: 10.1038/s41568-025-00791-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/02/2025] [Indexed: 02/26/2025]
Abstract
Over the past decade, substantial progress has been made in the development of targeted and immune-based therapies for patients with advanced non-small-cell lung cancer. To further improve outcomes for patients with lung cancer, identifying and intercepting disease at the earliest and most curable stages are crucial next steps. With the recent implementation of low-dose computed tomography scan screening in populations at high risk, there is an emerging unmet need for new diagnostic, prognostic and therapeutic tools to help treat patients suspected of harbouring premalignant lesions and minimally invasive non-small-cell lung cancer. Continued advances in the identification of the earliest drivers of lung carcinogenesis are poised to address these unmet needs. Employing multimodal approaches to chart the temporal and spatial maps of the molecular events driving lung premalignant lesion progression will refine our understanding of early carcinogenesis. Elucidating the molecular drivers of premalignancy is critical to the development of biomarkers to detect those incubating a premalignant lesion, to stratify risk for progression to invasive cancer and to identify novel therapeutic targets to intercept that process. In this Review, we summarize emerging insights into the earliest cellular and molecular events associated with lung squamous and adenocarcinoma carcinogenesis and highlight the growing opportunity for translating these insights into clinical tools for early detection and disease interception to transform the outcomes for those at risk for lung cancer.
Collapse
Affiliation(s)
- Sarah A Mazzilli
- Sectional Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| | - Zahraa Rahal
- Division of Pathology-Lab Medicine, Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Maral J Rouhani
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Sam M Janes
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Humam Kadara
- Division of Pathology-Lab Medicine, Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Steven M Dubinett
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, and Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Avrum E Spira
- Sectional Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- Johnson & Johnson Innovative Medicine, Boston, MA, USA.
| |
Collapse
|
24
|
Conrad C, Magnen M, Tsui J, Wismer H, Naser M, Venkataramani U, Samad B, Cleary SJ, Qiu L, Tian JJ, De Giovanni M, Mende N, Leavitt AD, Passegué E, Laurenti E, Combes AJ, Looney MR. Decoding functional hematopoietic progenitor cells in the adult human lung. Blood 2025; 145:1975-1986. [PMID: 40014797 PMCID: PMC7617544 DOI: 10.1182/blood.2024027884] [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: 12/04/2024] [Revised: 01/10/2025] [Accepted: 01/19/2025] [Indexed: 03/01/2025] Open
Abstract
ABSTRACT Although the bone marrow is the main site of blood cell production in adults, rare pools of hematopoietic stem and progenitor cells have been found in extramedullary organs. In mice, we have previously shown that the lung contains hematopoietic progenitor cells and is a site of platelet production. Here, in the adult human lung, we show that functional hematopoietic precursors reside in the extravascular spaces with a frequency similar to the bone marrow and are capable of proliferation and engraftment in mice. The gene signature of pulmonary and medullary CD34+ hematopoietic progenitors indicates greater baseline activation of immune-, megakaryocyte/platelet-, and erythroid-related pathways in lung progenitors. Spatial transcriptomics mapped blood progenitors in the lung to an alveolar interstitium niche with only a few cells identified in an intravascular location. In human blood samples collected for stem cell transplantation, CD34+ cells with a lung signature enriched the mobilized pool of hematopoietic stem cells. These results identify the lung as a pool for uniquely programmed blood stem and progenitor cells with the potential to support hematopoiesis in humans.
Collapse
Affiliation(s)
- Catharina Conrad
- Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Mélia Magnen
- Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Jessica Tsui
- UCSF CoLabs, University of California, San Francisco, CAUSA
| | | | - Mohammad Naser
- UCSF CoLabs, University of California, San Francisco, CAUSA
| | | | - Bushra Samad
- UCSF CoLabs, University of California, San Francisco, CAUSA
| | - Simon J. Cleary
- Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Longhui Qiu
- Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Jennifer J. Tian
- Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Marco De Giovanni
- Department of Microbiology & Immunology, University of California, San Francisco; San Francisco, CA, USA
| | - Nicole Mende
- Wellcome – MRC Cambridge Stem Cell Institute, Department of Hematology, University of Cambridge; Cambridge, UK
| | - Andrew D. Leavitt
- Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Emmanuelle Passegué
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center; New York, NY, USA
| | - Elisa Laurenti
- Wellcome – MRC Cambridge Stem Cell Institute, Department of Hematology, University of Cambridge; Cambridge, UK
| | - Alexis J. Combes
- Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
- UCSF CoLabs, University of California, San Francisco, CAUSA
- Department of Pathology, University of California, San Francisco; San Francisco, CA, USA
- Bakar ImmunoX Initiative, University of California, San Francisco; San Francisco, CA, USA
- Biomedical Sciences Program, University of California, San Francisco, CA, USA
| | - Mark R. Looney
- Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
- Bakar ImmunoX Initiative, University of California, San Francisco; San Francisco, CA, USA
- Biomedical Sciences Program, University of California, San Francisco, CA, USA
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
25
|
Elsner RA, Shlomchik MJ. Coordinated Regulation of Extrafollicular B Cell Responses by IL-12 and IFNγ. Immunol Rev 2025; 331:e70027. [PMID: 40211749 PMCID: PMC11986407 DOI: 10.1111/imr.70027] [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: 02/28/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025]
Abstract
Upon activation, B cells undergo either the germinal center (GC) or extrafollicular (EF) response. While GC are known to generate high-affinity memory B cells and long-lived plasma cells, the role of the EF response is less well understood. Initially, it was thought to be limited to that of a source of fast but lower-quality antibodies until the GC can form. However, recent evidence strongly supports the EF response as an important component of the humoral response to infection. EF responses are now also recognized as a source of pathogenic B cells in autoimmune diseases. The EF response itself is dynamic and regulated by pathways that are only recently being uncovered. We have identified that the cytokine IL-12 acts as a molecular switch, enhancing the EF response and suppressing GC through multiple mechanisms. These include direct effects on both B cells themselves and the coordinated differentiation of helper CD4 T cells. Here, we explore this pathway in relation to other recent advancements in our understanding of the EF response's role and highlight areas for future research. A better understanding of how the EF response forms and is regulated is essential for advancing treatments for many disease states.
Collapse
Affiliation(s)
- Rebecca A. Elsner
- Department of ImmunologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Mark J. Shlomchik
- Department of ImmunologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
26
|
Xie Z, Rose L, Feng J, Zhao Y, Lu Y, Kane H, Hibberd TJ, Hu X, Wang Z, Zang K, Yang X, Richardson Q, Othman R, Venezia O, Zhakyp A, Gao F, Abe N, Vigeland K, Wang H, Branch C, Duizer C, Deng L, Meng X, Zamidar L, Hauptschein M, Bergin R, Dong X, Chiu IM, Kim BS, Spencer NJ, Hu H, Jackson R. Enteric neuronal Piezo1 maintains mechanical and immunological homeostasis by sensing force. Cell 2025; 188:2417-2432.e19. [PMID: 40132579 PMCID: PMC12048284 DOI: 10.1016/j.cell.2025.02.031] [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: 05/06/2024] [Revised: 11/19/2024] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
The gastrointestinal (GI) tract experiences a myriad of mechanical forces while orchestrating digestion and barrier immunity. A central conductor of these processes, the enteric nervous system (ENS), detects luminal pressure to regulate peristalsis independently of extrinsic input from the central and peripheral nervous systems. However, how the ∼500 million enteric neurons that reside in the GI tract sense and respond to force remains unknown. Herein, we establish that the mechanosensor Piezo1 is functionally expressed in cholinergic enteric neurons. Optogenetic stimulation of Piezo1+ cholinergic enteric neurons drives colonic motility, while Piezo1 deficiency reduces cholinergic neuronal activity and slows peristalsis. Additionally, Piezo1 deficiency in cholinergic enteric neurons abolishes exercise-induced acceleration of GI motility. Finally, we uncover that enteric neuronal Piezo1 function is required for motility alterations in colitis and acts to prevent aberrant inflammation and tissue damage. This work uncovers how the ENS senses and responds to mechanical force.
Collapse
Affiliation(s)
- Zili Xie
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Lillian Rose
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Jing Feng
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA; Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yonghui Zhao
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Yisi Lu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Harry Kane
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Timothy J Hibberd
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Xueming Hu
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Zhen Wang
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Kaikai Zang
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Xingliang Yang
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | | | - Rahmeh Othman
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Olivia Venezia
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Ademi Zhakyp
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Fang Gao
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Nobuya Abe
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Keren Vigeland
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Hongshen Wang
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Camren Branch
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Coco Duizer
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Liwen Deng
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Xia Meng
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Lydia Zamidar
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Max Hauptschein
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Ronan Bergin
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Issac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Brian S Kim
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Allen Discovery Center for Neuroimmune Interactions, New York, NY 10029, USA
| | - Nick J Spencer
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Hongzhen Hu
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Allen Discovery Center for Neuroimmune Interactions, New York, NY 10029, USA; The Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Ruaidhrí Jackson
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
27
|
Boxer E, Feigin N, Tschernichovsky R, Darnell NG, Greenwald AR, Hoefflin R, Kovarsky D, Simkin D, Turgeman S, Zhang L, Tirosh I. Emerging clinical applications of single-cell RNA sequencing in oncology. Nat Rev Clin Oncol 2025; 22:315-326. [PMID: 40021788 DOI: 10.1038/s41571-025-01003-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2025] [Indexed: 03/03/2025]
Abstract
Single-cell RNA sequencing (scRNA-seq) has revolutionized our understanding of complex tissues both in health and in disease. Over the past decade, scRNA-seq has been applied to tumour samples obtained from patients with cancer in hundreds of studies, thereby advancing the view that each tumour is a complex ecosystem and uncovering the diverse states of both cancer cells and the tumour microenvironment. Such studies have primarily investigated and provided insights into the basic biology of cancer, although considerable research interest exists in leveraging these findings towards clinical applications. In this Review, we summarize the available data from scRNA-seq studies investigating samples from patients with cancer with a particular focus on findings that are of potential clinical relevance. We highlight four main research objectives of scRNA-seq studies and describe some of the most relevant findings towards such goals. We also describe the limitations of scRNA-seq, as well as future approaches in this field that are anticipated to further advance clinical applicability.
Collapse
Affiliation(s)
- Emily Boxer
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Nisan Feigin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Roi Tschernichovsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
| | - Noam Galili Darnell
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Alissa R Greenwald
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Rouven Hoefflin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Daniel Kovarsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Dor Simkin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Shira Turgeman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Lingling Zhang
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Itay Tirosh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
28
|
Jin J, Wang Z, Liu Y, Chen J, Jiang M, Lu L, Xu J, Gao F, Wang J, Zhang J, Xu GT, Jin C, Tian H, Zhao J, Ou Q. miR-143-3p boosts extracellular vesicles to improve the dermal fibrosis of localized scleroderma. J Autoimmun 2025; 153:103422. [PMID: 40273600 DOI: 10.1016/j.jaut.2025.103422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 03/15/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
Localized scleroderma (LoSc) is an autoimmune disease that features extensive fibrosis of the skin. Due to its severity and limited understanding, no effective treatments have been developed to date. Bone marrow mesenchymal stem cells (BMSCs) derived extracellular vesicles (EVs) have been demonstrated promising therapeutic effects on the LoSc mouse model in our previous study. However, identifying the targets and underlying mechanisms of EVs remains a significant challenge for therapeutic applications. miR-143-3p, a critical and abundant factor in BMSC-EVs identified through miRNA sequencing, mediates antifibrotic effects in a LoSc mouse model and is significantly lacking in the dermis of LoSc patients. This microRNA inhibits myofibroblast formation and collagen synthesis, contributing to the therapeutic effects of BMSC-EVs in the LoSc mouse model. Moreover, miR-143-3p-reinforced BMSC-EVs demonstrated enhanced therapeutic efficacy compared to normal BMSC-EVs, reducing dermal thickening, collagen deposition, fibroblast differentiation into myofibroblasts, and promoting skin tissue remodeling. IGF1R, highly expressed in the skin of LoSc, was identified as a potential target of miR-143-3p and was inhibited by miR-143-3p-reinforced EVs, thereby modulating the IGF1/IGF1R-AKT/MAPK pathway. In conclusion, miR-143-3p-enriched EVs could be a more efficient candidate for treating dermal fibrosis in LoSc.
Collapse
Affiliation(s)
- Jiahui Jin
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Dermatology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhe Wang
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Physiology, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
| | - Yifan Liu
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jie Chen
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Miao Jiang
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lixia Lu
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jingying Xu
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Furong Gao
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Juan Wang
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jieping Zhang
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guo-Tong Xu
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Caixia Jin
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Haibin Tian
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Jingjun Zhao
- Department of Dermatology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qingjian Ou
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
29
|
Heiser BJ, Veyssi A, Ghosh D. Recent strategies for enhanced delivery of mRNA to the lungs. Nanomedicine (Lond) 2025; 20:1043-1069. [PMID: 40190037 PMCID: PMC12051540 DOI: 10.1080/17435889.2025.2485669] [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: 12/20/2024] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
mRNA-based therapies have emerged as a transformative tool in modern medicine, gaining significant attention following their successful use in COVID-19 vaccines. Delivery to the lungs offers several compelling advantages for mRNA delivery. The lungs are one of the most vascularized organs in the body, which provides an extensive surface area that can facilitate efficient drug transport. Local delivery to the lungs bypasses gastrointestinal degradation, potentially enhancing therapeutic efficacy. In addition, the extensive capillary network of the lungs provides an ideal target for systemic delivery. However, developing effective mRNA therapies for the lungs presents significant challenges. The complex anatomy of the lungs and the body's immune response to foreign particles create barriers to delivery. This review discusses key approaches for overcoming these challenges and improving mRNA delivery to the lungs. It examines both local and systemic delivery strategies aimed at improving lung delivery while mitigating off-target effects. Although substantial progress has been made in lung-targeted mRNA therapies, challenges remain in optimizing cellular uptake and achieving therapeutic efficacy within pulmonary tissues. The continued refinement of delivery strategies that enhance lung-specific targeting while minimizing degradation is critical for the clinical success of mRNA-based pulmonary therapies.
Collapse
Affiliation(s)
- Brittany J. Heiser
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Arian Veyssi
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Debadyuti Ghosh
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
30
|
Li J, Ma Y, Cao Y, Zheng G, Ren Q, Chen C, Zhu Q, Zhou Y, Lu Y, Zhang Y, Deng C, Chen WH, Su J. Integrating microbial GWAS and single-cell transcriptomics reveals associations between host cell populations and the gut microbiome. Nat Microbiol 2025; 10:1210-1226. [PMID: 40195537 DOI: 10.1038/s41564-025-01978-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025]
Abstract
Microbial genome-wide association studies (GWAS) have uncovered numerous host genetic variants associated with gut microbiota. However, links between host genetics, the gut microbiome and specific cellular contexts remain unclear. Here we use a computational framework, scBPS (single-cell Bacteria Polygenic Score), to integrate existing microbial GWAS and single-cell RNA-sequencing profiles of 24 human organs, including the liver, pancreas, lung and intestine, to identify host tissues and cell types relevant to gut microbes. Analysing 207 microbial taxa and 254 host cell types, scBPS-inferred cellular enrichments confirmed known biology such as dominant communications between gut microbes and the digestive tissue module and liver epithelial cell compartment. scBPS also identified a robust association between Collinsella and the central-veinal hepatocyte subpopulation. We experimentally validated the causal effects of Collinsella on cholesterol metabolism in mice through single-nuclei RNA sequencing on liver tissue to identify relevant cell subpopulations. Mechanistically, oral gavage of Collinsella modulated cholesterol pathway gene expression in central-veinal hepatocytes. We further validated our approach using independent microbial GWAS data, alongside single-cell and bulk transcriptomic analyses, demonstrating its robustness and reproducibility. Together, scBPS enables a systematic mapping of the host-microbe crosstalk by linking cell populations to their interacting gut microbes.
Collapse
Affiliation(s)
- Jingjing Li
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yunlong Ma
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yue Cao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Gongwei Zheng
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qing Ren
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Cheng Chen
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qunyan Zhu
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yijun Zhou
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yu Lu
- The Second School of Clinical Medicine, Institution of Medical Artificial Intelligence, Binzhou Medical University, Yantai, China
| | - Yaru Zhang
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Chunyu Deng
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Wei-Hua Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- The Second School of Clinical Medicine, Institution of Medical Artificial Intelligence, Binzhou Medical University, Yantai, China.
- School of Biological Science, Jining Medical University, Rizhao, China.
| | - Jianzhong Su
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
31
|
Xie Z, Zheng G, Niu L, Du K, Li R, Dan H, Duan L, Wu H, Ren G, Dou X, Dai S, Feng F, Zhang J, Zheng J. SPP1 + macrophages in colorectal cancer: Markers of malignancy and promising therapeutic targets. Genes Dis 2025; 12:101340. [PMID: 40092488 PMCID: PMC11907465 DOI: 10.1016/j.gendis.2024.101340] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/19/2024] [Accepted: 04/03/2024] [Indexed: 03/19/2025] Open
Abstract
SPP1+ macrophages have been identified as key players in the colorectal cancer (CRC) tumor microenvironment, but their function remains unclear. This study integrated single-cell and spatial transcriptomics with bulk sequencing to investigate the roles and mechanisms of SPP1 + macrophages in CRC. Our findings revealed a pronounced elevation of SPP1 + macrophages in CRC, especially within tumor territories. These macrophages served as markers for CRC initiation, progression, metastasis, and potential prognosis. Furthermore, they showed heightened transcriptional activity in genes linked to angiogenesis, epithelial-mesenchymal transition, glycolysis, hypoxia, and immunosuppression. SPP1 protein amplified CRC cell migration and invasion, potentially mediating cellular crosstalk via the SPP1-CD44, SPP1-PTGER4, and SPP1-a4b1 complex axes. Patients with a high proportion of SPP1 + macrophages could benefit more from immune checkpoint blockade therapy. Interestingly, CSF1R expression was significantly enriched in C1QC + macrophages versus SPP1 + macrophages, possibly explaining limited anti-CSF1R monotherapy effects. In conclusion, we propose an SPP1 + macrophage model in CRC, highlighting such macrophages as a promising therapeutic target due to their malignancy markers.
Collapse
Affiliation(s)
- Zhenyu Xie
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Medical University, Xi'an, Shaanxi 710032, China
| | - Gaozan Zheng
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Medical University, Xi'an, Shaanxi 710032, China
| | - Liaoran Niu
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Medical University, Xi'an, Shaanxi 710032, China
| | - Kunli Du
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Medical University, Xi'an, Shaanxi 710032, China
| | - Ruikai Li
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Medical University, Xi'an, Shaanxi 710032, China
| | - Hanjun Dan
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Medical University, Xi'an, Shaanxi 710032, China
| | - Lili Duan
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Medical University, Xi'an, Shaanxi 710032, China
| | - Hongze Wu
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Medical University, Xi'an, Shaanxi 710032, China
| | - Guangming Ren
- Xi'an Medical University, Xi'an, Shaanxi 710021, China
| | - Xinyu Dou
- Xi'an Medical University, Xi'an, Shaanxi 710021, China
| | - Songchen Dai
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110016, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning 110016, China
| | - Fan Feng
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Medical University, Xi'an, Shaanxi 710032, China
| | - Jian Zhang
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Jianyong Zheng
- Department of Digestive Surgery, Xijing Hospital of Digestive Diseases, Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
32
|
Roerden M, Spranger S. Cancer immune evasion, immunoediting and intratumour heterogeneity. Nat Rev Immunol 2025; 25:353-369. [PMID: 39748116 DOI: 10.1038/s41577-024-01111-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2024] [Indexed: 01/04/2025]
Abstract
Cancers can avoid immune-mediated elimination by acquiring traits that disrupt antitumour immunity. These mechanisms of immune evasion are selected and reinforced during tumour evolution under immune pressure. Some immunogenic subclones are effectively eliminated by antitumour T cell responses (a process known as immunoediting), which results in a clonally selected tumour. Other cancer cells arise to resist immunoediting, which leads to a tumour that includes several distinct cancer cell populations (referred to as intratumour heterogeneity (ITH)). Tumours with high ITH are associated with poor patient outcomes and a lack of responsiveness to immune checkpoint blockade therapy. In this Review, we discuss the different ways that cancer cells evade the immune system and how these mechanisms impact immunoediting and tumour evolution. We also describe how subclonal antigen presentation in tumours with high ITH can result in immune evasion.
Collapse
Affiliation(s)
- Malte Roerden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute for Technology, Cambridge, MA, USA
| | - Stefani Spranger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute for Technology, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute for Technology, Cambridge, MA, USA.
- Ragon Institute of Mass General Hospital, Massachusetts Institute for Technology and Harvard, Cambridge, MA, USA.
| |
Collapse
|
33
|
Green EH, Kotrannavar SR, Rutherford ME, Lunnemann HM, Kaur H, Heiser CN, Ding H, Simmons AJ, Liu X, Lacy DB, Washington MK, Shrubsole MJ, Liu Q, Lau KS, Sears CL, Coffey RJ, Drewes JL, Markham NO. Multiomic spatial atlas shows deleted in malignant brain tumors 1 (DMBT1) glycoprotein is lost in colonic dysplasia. J Pathol 2025; 266:51-65. [PMID: 40026233 PMCID: PMC11985286 DOI: 10.1002/path.6406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 12/03/2024] [Accepted: 01/15/2025] [Indexed: 03/05/2025]
Abstract
Colorectal cancer (CRC) is responsible for over 900,000 annual deaths worldwide. Emerging evidence supports pro-carcinogenic bacteria in the colonic microbiome are at least promotional in CRC development and may be causal. We previously showed toxigenic C. difficile from human CRC-associated bacterial biofilms accelerates tumorigenesis in ApcMin/+ mice, both in specific pathogen-free mice and in gnotobiotic mice colonized with a defined consortium of bacteria. To further understand host-microbe interactions during colonic tumorigenesis, we combined single-cell RNA-sequencing (scRNA-seq), spatial transcriptomics, and immunofluorescence to define the molecular spatial organization of colonic dysplasia in our consortium model with or without C. difficile. Our data show a striking bipartite regulation of Deleted in Malignant Brain Tumors 1 (DMBT1) in the inflamed versus dysplastic colon. From scRNA-seq, differential gene expression analysis of normal absorptive colonocytes at 2 weeks postinoculation showed DMBT1 upregulated by C. difficile compared to colonocytes from mice without C. difficile exposure. In contrast, our spatial transcriptomic analysis showed DMBT1 dramatically downregulated in dysplastic foci compared with normal-adjacent tissue. We further integrated our datasets to generate custom colonic dysplasia scores and ligand-receptor mapping. Validation with immunofluorescence showed DMBT1 protein downregulated in dysplastic foci from three mouse models of colonic tumorigenesis and in adenomatous dysplasia from human samples. Finally, we used mouse and human organoids to implicate WNT signaling in the downregulation of DMBT1 mRNA and protein. Together, our data reveal cell type-specific regulation of DMBT1, a potential mechanistic link between bacteria and colonic tumorigenesis. Published 2025. This article is a U.S. Government work and is in the public domain in the USA. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Emily H Green
- Department of Pathology, Microbiology, and ImmunologyVanderbilt University Medical CenterNashvilleTNUSA
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTNUSA
| | | | - Megan E Rutherford
- Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Hannah M Lunnemann
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTNUSA
- Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Harsimran Kaur
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTNUSA
- Chemical and Physical Biology ProgramVanderbilt UniversityNashvilleTNUSA
| | - Cody N Heiser
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTNUSA
- Department of Cell and Developmental BiologyVanderbilt UniversityNashvilleTNUSA
| | - Hua Ding
- Department of Microbiology and Molecular ImmunologyBloomberg School of Public HealthBaltimoreMDUSA
| | - Alan J Simmons
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTNUSA
- Department of Cell and Developmental BiologyVanderbilt UniversityNashvilleTNUSA
| | - Xiao Liu
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTNUSA
| | - D Borden Lacy
- Department of Pathology, Microbiology, and ImmunologyVanderbilt University Medical CenterNashvilleTNUSA
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTNUSA
- Department of Veterans AffairsTennessee Valley Healthcare SystemNashvilleTNUSA
| | - M Kay Washington
- Department of Pathology, Microbiology, and ImmunologyVanderbilt University Medical CenterNashvilleTNUSA
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTNUSA
| | - Martha J Shrubsole
- Vanderbilt Epidemiology CenterVanderbilt University Medical CenterNashvilleTNUSA
- Vanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTNUSA
| | - Qi Liu
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTNUSA
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTNUSA
| | - Ken S Lau
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTNUSA
- Department of Cell and Developmental BiologyVanderbilt UniversityNashvilleTNUSA
- Vanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTNUSA
| | - Cynthia L Sears
- Department of Microbiology and Molecular ImmunologyBloomberg School of Public HealthBaltimoreMDUSA
- Department of Medicine, Division of Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMDUSA
- Department of OncologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Robert J Coffey
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTNUSA
- Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
- Department of Cell and Developmental BiologyVanderbilt UniversityNashvilleTNUSA
- Vanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTNUSA
| | - Julia L Drewes
- Department of Medicine, Division of Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMDUSA
- Department of OncologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Nicholas O Markham
- Department of Pathology, Microbiology, and ImmunologyVanderbilt University Medical CenterNashvilleTNUSA
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTNUSA
- Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
- Department of Veterans AffairsTennessee Valley Healthcare SystemNashvilleTNUSA
- Vanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTNUSA
| |
Collapse
|
34
|
Song EM, Saqib J, Joo YH, Ramsha Z, Moon CM, Jung SA, Kim J. Inflammatory transcriptomic signatures and cell type compositions in inflamed and non-inflamed colonic mucosa of ulcerative colitis. Genes Dis 2025; 12:101447. [PMID: 39926325 PMCID: PMC11804533 DOI: 10.1016/j.gendis.2024.101447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 07/28/2024] [Indexed: 02/11/2025] Open
Affiliation(s)
- Eun Mi Song
- Departments of Internal Medicine, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Jahanzeb Saqib
- School of Systems Biomedical Science, Soongsil University, 369 Sangdo-Ro, Dongjak-Gu, Seoul 06978, Republic of Korea
| | - Yang Hee Joo
- Departments of Internal Medicine, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Zehra Ramsha
- School of Systems Biomedical Science, Soongsil University, 369 Sangdo-Ro, Dongjak-Gu, Seoul 06978, Republic of Korea
| | - Chang Mo Moon
- Departments of Internal Medicine, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Sung-Ae Jung
- Departments of Internal Medicine, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Junil Kim
- School of Systems Biomedical Science, Soongsil University, 369 Sangdo-Ro, Dongjak-Gu, Seoul 06978, Republic of Korea
| |
Collapse
|
35
|
Lee HJ, Park SW, Lee JH, Chang SY, Oh SM, Mun S, Kang J, Park JE, Choi JK, Kim TI, Kim JY, Kim P. Differential cellular origins of the extracellular matrix of tumor and normal tissues according to colorectal cancer subtypes. Br J Cancer 2025; 132:770-782. [PMID: 40032993 PMCID: PMC12041468 DOI: 10.1038/s41416-025-02964-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: 04/16/2024] [Revised: 01/07/2025] [Accepted: 02/13/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Understanding the proteomic-level heterogeneity of the tumor microenvironment (TME) in colorectal cancer (CRC) is crucial due to its well-known heterogeneity. While heterogenous CRC has been extensively characterized at the molecular subtype level, research into the functional heterogeneity of fibroblasts, particularly their relationship with extracellular matrix (ECM) alterations, remains limited. Addressing this gap is essential for a comprehensive understanding of CRC progression and the development of targeted therapies. METHODS 24 tissue samples from 21 CRC patients, along with adjacent normal tissues (NAT), were collected and decellularized using a detergent-based method to enrich the ECM component. Proteomic analysis of ECM-enriched samples was performed using tandem mass tag (TMT) spectrometry, followed by statistical analysis including differential expression protein (DEP) analysis. Single-cell RNA sequencing (scRNA-Seq) data from public datasets were integrated and analyzed to delineate cell states within the TME. Bulk tissue RNA-Seq and bioinformatics analysis, including consensus molecular subtype (CMS) classification and single-cell level deconvolution of TCGA bulk RNA-seq data, were conducted to further explore gene expression patterns and TME composition. RESULTS Differential cellular origin of the NAT and tumorous ECM proteins were identified, revealing 110 ECM proteins enriched in NAT and 28 ECM proteins in tumor tissues. Desmoplastic and WNT5A+ inflammatory fibroblasts were indicated as the sources of tumor-enriched ECM proteins, while ADAMDEC1+ expressing fibroblasts and PI16+ expressing fibroblast were identified as the sources of NAT-enriched ECM proteins. Deconvolution of bulk RNA-seq of CRC tissues discriminated CMS-specific fibroblast state, reflecting the biological traits of each CMS subtype. Specially, seven ECM genes specific to mesenchymal subtype (CMS4), including PI16+ fibroblast-related 4 genes (SFRP2, PRELP, OGN, SRPX) and desmoplastic fibroblast-related 3 genes (THBS2, CTHRC1, BGN), showed a significant association with poorer survival in patient with CRC. CONCLUSION We conducted an extracellular matrix (ECM)-focused profiling of the TME by integrating quantitative proteomics with single-cell RNA sequencing (scRNA-seq) data from CRC patients. We identified the ECM proteins of NAT and tumor tissue, and established a cell-matrisome database. We defined mesenchymal subtype-specific molecules associated with specific fibroblast subtypes showing a significant association with poorer survival in patients with CRC. Our ECM-focused profiling of tumor stroma provides new insights as indicators for biological processes and clinical endpoints.
Collapse
Affiliation(s)
- Hyun Jin Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Sang Woo Park
- Korea Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, 28119, Republic of Korea
| | - Jun Hyeong Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Shin Young Chang
- Department of Internal Medicine, Institute of Gastroenterology, Brain Korea 21 Project for Medical Science, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Sang Mi Oh
- Department of Internal Medicine, Institute of Gastroenterology, Brain Korea 21 Project for Medical Science, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Siwon Mun
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Junho Kang
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Jong-Eun Park
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
- SCL-KAIST Institute of Translational Research, KAIST, Daejeon, Republic of Korea
| | - Jung Kyoon Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
- SCL-KAIST Institute of Translational Research, KAIST, Daejeon, Republic of Korea
| | - Tae Il Kim
- Department of Internal Medicine, Institute of Gastroenterology, Brain Korea 21 Project for Medical Science, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Jin Young Kim
- Korea Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, 28119, Republic of Korea.
| | - Pilnam Kim
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea.
- SCL-KAIST Institute of Translational Research, KAIST, Daejeon, Republic of Korea.
| |
Collapse
|
36
|
Zhang H, Zhang N, Yang X, Wang C, Yang Q, Luo J, Ye T. BRAF mutation cancer, colorectal cancer, tumor associated lymph node structure and immune microenvironment study: MAPK protein kinase molecular action and SIRPG-CD47 protein signaling pathway. Int J Biol Macromol 2025; 307:142191. [PMID: 40101830 DOI: 10.1016/j.ijbiomac.2025.142191] [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: 01/14/2025] [Revised: 03/11/2025] [Accepted: 03/15/2025] [Indexed: 03/20/2025]
Abstract
BRAF mutation affects the biological characteristics and microenvironment of the tumor during the development of colorectal cancer. Tumor-associated lymph nodes are the key sites of immune response. This study aimed to systematically evaluate the impact of BRAF gene mutations on the remodeling of the CRC immune microenvironment, with a particular focus on their effects on the maturation and function of TLS·In this study, clinical samples of CRC patients were collected, and immune cell subsets were analyzed by single-cell RNA sequencing, and pseudo-temporal locus analysis and spatial transcriptome analysis were performed to explore intercellular communication and functional enrichment analysis. The distribution and maturity of TLS were evaluated by immunohistochemistry and multiple fluorescence staining techniques, and statistical analysis was performed.The results showed that BRAF mutation significantly affected the number and maturity of lymphatic structures infiltrated by tumors, and was negatively correlated with patient prognosis. BRAF mutations lead to alterations in T cell subsets, particularly the dual role of CD4+ CXCL13 cells in TLS maturation. B-cell subpopulation analysis revealed functional deficits in CRC patients with BRAF mutations, which further drove the remodeling of the tumor immune microenvironment.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Oncology, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Nenglin Zhang
- Department of Gastroenterology, First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232007, China
| | - Xiaodi Yang
- Department of Oncology, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Chen Wang
- Department of Oncology, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Qinghui Yang
- Department of Oncology, Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Jing Luo
- Department of Oncology, Minhang Hospital, Fudan University, Shanghai 201199, China; Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201199, China.
| | - Tao Ye
- Department of Oncology, Minhang Hospital, Fudan University, Shanghai 201199, China; Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hospital & AHS, Fudan University, Shanghai 201199, China.
| |
Collapse
|
37
|
Cuesta-Margolles G, Schlecht-Louf G, Bachelerie F. ACKR3 in Skin Homeostasis, an Overlooked Player in the CXCR4/CXCL12 Axis. J Invest Dermatol 2025; 145:1039-1049. [PMID: 39466217 DOI: 10.1016/j.jid.2024.08.022] [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: 04/26/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 10/29/2024]
Abstract
CXCL12 and its receptor CXCR4 emerge as critical regulators within the intricate network of processes ensuring skin homeostasis. In this review, we discuss their spatial distribution and function in steady-state skin; delve into their role in acute wound healing, with emphasis on fibrotic and regenerative responses; and explore their relevance in skin responses to commensals and pathogens. Given the lack of knowledge surrounding ACKR3, the atypical receptor of CXCL12, we speculate whether and how it might be involved in the processes mentioned earlier. Is ACKR3 the (a)typical friend who enjoys missing the party, or do we need to take a closer look?
Collapse
Affiliation(s)
| | - Géraldine Schlecht-Louf
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| | - Françoise Bachelerie
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| |
Collapse
|
38
|
Gómez-López S, Alhendi ASN, Przybilla MJ, Bordeu I, Whiteman ZE, Butler T, Rouhani MJ, Kalinke L, Uddin I, Otter KEJ, Chandrasekharan DP, Lebrusant-Fernandez M, Shurr AYL, Durrenberger PF, Moore DA, Falzon M, Reading JL, Martincorena I, Simons BD, Campbell PJ, Janes SM. Aberrant basal cell clonal dynamics shape early lung carcinogenesis. Science 2025:eads9145. [PMID: 40310937 DOI: 10.1126/science.ads9145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 04/10/2025] [Indexed: 05/03/2025]
Abstract
Preinvasive squamous lung lesions are precursors of lung squamous cell carcinoma (LUSC). The cellular events underlying lesion formation are unknown. Using a carcinogen-induced model of LUSC with no added genetic hits or cell type bias, we find that carcinogen exposure leads to non-neutral competition among basal cells, aberrant clonal expansions, and basal cell mobilization along the airways. Ultimately, preinvasive lesions develop from a few highly mutated clones that dominate most of the bronchial tree. Multi-site sequencing in human patients confirms the presence of clonally related preinvasive lesions across distinct airway regions. Our work identifies a transition in basal cell clonal dynamics, and an associated shift in basal cell fate, as drivers of field cancerization in the lung.
Collapse
Affiliation(s)
- Sandra Gómez-López
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Ahmed S N Alhendi
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Moritz J Przybilla
- Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Ignacio Bordeu
- Departamento de Física, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile, Santiago, Chile
| | - Zoe E Whiteman
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Timothy Butler
- Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Maral J Rouhani
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Lukas Kalinke
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Imran Uddin
- Cancer Research UK City of London Centre Single Cell Genomics Facility, UCL Cancer Institute, University College London, London, UK
- Genomics Translational Technology Platform, UCL Cancer Institute, University College London, London, UK
| | - Kate E J Otter
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | | | - Marta Lebrusant-Fernandez
- Pre-Cancer Immunology Laboratory, UCL Cancer Institute, University College London, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Abigail Y L Shurr
- Pre-Cancer Immunology Laboratory, UCL Cancer Institute, University College London, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Pascal F Durrenberger
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - David A Moore
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Department of Cellular Pathology, University College London Hospitals NHS Trust, London, UK
| | - Mary Falzon
- Department of Cellular Pathology, University College London Hospitals NHS Trust, London, UK
| | - James L Reading
- Pre-Cancer Immunology Laboratory, UCL Cancer Institute, University College London, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Iñigo Martincorena
- Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Benjamin D Simons
- Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Cambridge, UK
- Gurdon Institute, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Peter J Campbell
- Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Sam M Janes
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
- University College London Hospitals NHS Trust, London, UK
| |
Collapse
|
39
|
Zhang G, Lian Y, Li Q, Zhou S, Zhang L, Chen L, Tang J, Liu H, Li N, Pan Q, Gu Y, Lin N, Wang H, Wang X, Guo J, Zhang W, Jin Z, Xu B, Su X, Lin M, Han Q, Qin J. Vagal pathway activation links chronic stress to decline in intestinal stem cell function. Cell Stem Cell 2025; 32:778-794.e10. [PMID: 40120585 DOI: 10.1016/j.stem.2025.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 01/11/2025] [Accepted: 02/26/2025] [Indexed: 03/25/2025]
Abstract
Chronic stress adversely affects intestinal health, but the specific neural pathways linking the brain to intestinal tissue are not fully understood. Here, we show that chronic stress-induced activation of the central amygdala-dorsal motor nucleus of the vagus (CeA-DMV) pathway accelerates premature aging and impairs the stemness of intestinal stem cells (ISCs). This pathway influences ISC function independently of the microbiota, the hypothalamic-pituitary-adrenal (HPA) axis, the immune response, and the sympathetic nervous system (SNS). Under chronic stress, DMV-mediated vagal activation prompts cholinergic enteric neurons to release acetylcholine (ACh), which engages ISCs via the M3 muscarinic acetylcholine receptor (CHRM3). This interaction activates the p38 mitogen-activated protein kinase (MAPK) pathway, triggering growth arrest and mitochondrial fragmentation, thereby accelerating an aging-like decline in ISCs. Together, our findings provide insights into an alternative neural mechanism that links stress to intestinal dysfunction. Strategies targeting the DMV-associated vagal pathway represent potential therapeutic approaches for stress-induced intestinal diseases.
Collapse
Affiliation(s)
- Guoying Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Yannan Lian
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Qingguo Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Shanghai 200032, China
| | - Shudi Zhou
- Department of Endocrinology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Lili Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Liting Chen
- Department of Emergency and Critical Disease, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Department of Anatomy and Physiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Junzhe Tang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Shanghai 200032, China
| | - Hailong Liu
- Department of General Surgery, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ni Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Qiang Pan
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Yongqiang Gu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Naiheng Lin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Hanling Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xuege Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Jiacheng Guo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Wei Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Zige Jin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Beitao Xu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xiao Su
- Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Moubin Lin
- Department of General Surgery, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Qi Han
- Department of Emergency and Critical Disease, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Department of Anatomy and Physiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Jun Qin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China.
| |
Collapse
|
40
|
Donado CA, Theisen E, Zhang F, Nathan A, Fairfield ML, Rupani KV, Jones D, Johannes KP, Raychaudhuri S, Dwyer DF, Jonsson AH, Brenner MB. Granzyme K activates the entire complement cascade. Nature 2025; 641:211-221. [PMID: 39914456 DOI: 10.1038/s41586-025-08713-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 01/29/2025] [Indexed: 03/21/2025]
Abstract
Granzymes are a family of serine proteases that are mainly expressed by CD8+ T cells, natural killer cells and innate-like lymphocytes1. Although their primary function is thought to be the induction of cell death in virally infected cells and tumours, accumulating evidence indicates that some granzymes can elicit inflammation by acting on extracellular substrates1. We previously found that most tissue CD8+ T cells in rheumatoid arthritis synovium, and in inflamed organs for some other diseases, express granzyme K (GZMK)2, a tryptase-like protease with poorly defined function. Here, we show that GZMK can activate the complement cascade by cleaving the C2 and C4 proteins. The nascent C4b and C2b fragments form a C3 convertase that cleaves C3, enabling the assembly of a C5 convertase that cleaves C5. The resulting convertases generate all the effector molecules of the complement cascade: the anaphylatoxins C3a and C5a, the opsonins C4b and C3b, and the membrane attack complex. In rheumatoid arthritis synovium, GZMK is enriched in regions with abundant complement activation, and fibroblasts are the main producers of complement proteins that serve as substrates for GZMK-mediated complement activation. Furthermore, Gzmk-deficient mice are significantly protected from inflammatory disease, exhibiting reduced arthritis and dermatitis, with concomitant decreases in complement activation. Our findings describe the discovery of a previously unidentified mechanism of complement activation that is driven entirely by lymphocyte-derived GZMK. Given the widespread abundance of GZMK-expressing T cells in tissues in chronic inflammatory diseases, GZMK-mediated complement activation is likely to be an important contributor to tissue inflammation in multiple disease contexts.
Collapse
Affiliation(s)
- Carlos A Donado
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Erin Theisen
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA, USA
| | - Fan Zhang
- Division of Rheumatology and Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Aparna Nathan
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Madison L Fairfield
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Karishma Vijay Rupani
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Dominique Jones
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kellsey P Johannes
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Soumya Raychaudhuri
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniel F Dwyer
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - A Helena Jonsson
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Michael B Brenner
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
41
|
Janto NV, Gleizes AR, Sun SJ, Ari G, Rao V, Gracz AD. Tritrichomonas muris sensitizes the intestinal epithelium to doxorubicin-induced apoptosis. Am J Physiol Gastrointest Liver Physiol 2025; 328:G594-G609. [PMID: 40243204 DOI: 10.1152/ajpgi.00242.2024] [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: 08/12/2024] [Revised: 09/26/2024] [Accepted: 04/03/2025] [Indexed: 04/18/2025]
Abstract
Doxorubicin (DXR) is a widely used chemotherapy drug that can induce severe intestinal mucositis. Although the influence of gut bacteria on DXR-induced damage has been documented, the role of eukaryotic commensals remains unexplored. We discovered Tritrichomonas muris (Tmu) in one of our mouse colonies exhibiting abnormal tuft cell hyperplasia, prompting an investigation into its impact on DXR-induced intestinal injury. Mice from Tmu-colonized and Tmu-excluded facilities were injected with DXR. Tissue morphology and gene expression were evaluated at acute injury (6 h) and regenerative (72 h and 120 h) phases. Changes to crypt and villus morphology were more subtle than previously reported and region-specific, with significantly shorter jejunal villi in Tmu+ mice at 72 h post-DXR compared with Tmu- controls. Most notably, we observed elevated rates of DXR-induced apoptosis, measured by cleaved caspase 3 (CC3) staining, in Tmu+ intestinal crypts at 6 h post-DXR. Tmu+ mice also exhibited reduced expression of active intestinal stem cell (aISC) marker Lgr5 and facultative ISC (fISC) marker Ly6a at 6 h post-DXR compared with Tmu- controls. Tmu, but not DXR, was associated with increased inflammation and expression of type 2 cytokines IL-5 and IL-13. Tmu+ mice also exhibited a decreased fecal abundance of Lactobacillus, which promotes gut barrier integrity, and reduced claudin expression, indicating potential barrier dysfunction that could explain the increase in DXR-induced apoptosis. These findings highlight the significant influence of commensal microbiota, particularly eukaryotic organisms like Tmu, on intestinal biology and response to chemotherapy, underscoring the complexity of gut microbiota interactions in drug-induced mucositis.NEW & NOTEWORTHY Our study found that the eukaryotic commensal Tritrichomonas muris (Tmu) significantly increases DXR-induced intestinal apoptosis in mice. Tmu also reduces Lgr5 expression post-DXR injury and elevates inflammation and type 2 cytokine expression in the absence of injury. 16S sequencing identifies decreased abundance of protective Lactobacillus in Tmu colonized mice, as well as decreased expression of barrier-forming claudins, which may explain increased apoptosis. These findings emphasize the complex role of microbiota in drug-induced intestinal damage.
Collapse
Affiliation(s)
- Nicolas V Janto
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, Georgia, United States
- Graduate Program in Genetics and Molecular Biology, Emory University, Atlanta, Georgia, United States
| | - Antoine R Gleizes
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, Georgia, United States
| | - Siyang J Sun
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, Georgia, United States
| | - Gurel Ari
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, Georgia, United States
| | - Vivek Rao
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, Georgia, United States
| | - Adam D Gracz
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, Georgia, United States
- Graduate Program in Genetics and Molecular Biology, Emory University, Atlanta, Georgia, United States
- Department of Human Genetics, Emory University, Atlanta, Georgia, United States
| |
Collapse
|
42
|
Windster JD, Kakiailatu NJM, Kuil LE, Antanaviciute A, Sacchetti A, MacKenzie KC, Peulen-Zink J, Kan TW, Bindels E, de Pater E, Doukas M, van den Bosch TPP, Yousefi S, Barakat TS, Meeussen CJHM, Sloots PCEJ, Wijnen RMH, Parikh K, Boesmans W, Melotte V, Hofstra RMW, Simmons A, Alves MM. Human Enteric Glia Diversity in Health and Disease: New Avenues for the Treatment of Hirschsprung Disease. Gastroenterology 2025; 168:965-979.e12. [PMID: 39725172 DOI: 10.1053/j.gastro.2024.12.011] [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: 10/12/2023] [Revised: 11/16/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND & AIMS The enteric nervous system (ENS), which is composed of neurons and glia, regulates intestinal motility. Hirschsprung disease (HSCR) results from defects in ENS formation; however, although neuronal aspects have been studied extensively, enteric glia remain disregarded. This study aimed to explore enteric glia diversity in health and disease. METHODS Full-thickness intestinal resection material from pediatric controls and patients with HSCR was collected, dissociated, and enriched for the ENS population through fluorescence-activated cell sorting. Single-cell RNA sequencing was performed to uncover the transcriptomic diversity of the ENS in controls and HSCR patients, as well as in wild-type and ret mutant zebrafish. Immunofluorescence and fluorescence in situ hybridization confirmed the presence of distinct subtypes. RESULTS Two major enteric glial classes emerged in the pediatric intestine: Schwann-like enteric glia, which are reminiscent of Schwann cells, and enteric glia expressing classical glial markers. Comparative analysis with previously published datasets confirmed our classification and revealed that although classical enteric glia are predominant prenatally, Schwann-like enteric glia become more abundant postnatally. In HSCR, ganglionic segments mirrored controls and aganglionic segments featured only Schwann-like enteric glia. Leveraging the regenerative potential of Schwann cells, we explored therapeutic options using a ret mutant zebrafish. Prucalopride, a serotonin-receptor (5-HT) agonist, induced neurogenesis partially rescuing the HSCR phenotype in ret+/- mutants. CONCLUSIONS Two major enteric glial classes were identified in the pediatric intestine, highlighting the significant postnatal contribution of Schwann-like enteric glia to glial heterogeneity. Crucially, these glial subtypes persist in aganglionic segments of patients with HSCR, offering a new target for their treatment using 5-HT agonists.
Collapse
Affiliation(s)
- Jonathan D Windster
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands; Department of Pediatric Surgery, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Naomi J M Kakiailatu
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Laura E Kuil
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Agne Antanaviciute
- MRC Translational Immune Discovery Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford, United Kingdom; MRC WIMM Centre for Computational Biology, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Andrea Sacchetti
- Department of Pathology, Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Katherine C MacKenzie
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Joke Peulen-Zink
- Department of Hematology, Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Tsung W Kan
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Eric Bindels
- Department of Hematology, Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Emma de Pater
- Department of Hematology, Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Michail Doukas
- Department of Pathology, Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Thierry P P van den Bosch
- Department of Pathology, Erasmus Medical Center Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Soheil Yousefi
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Tahsin-Stefan Barakat
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Conny J H M Meeussen
- Department of Pediatric Surgery, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Pim C E J Sloots
- Department of Pediatric Surgery, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Rene M H Wijnen
- Department of Pediatric Surgery, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Kaushal Parikh
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Werend Boesmans
- Biomedical Research Institute, Hasselt University, Hasselt, Belgium; Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Veerle Melotte
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands; Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Robert M W Hofstra
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Alison Simmons
- MRC WIMM Centre for Computational Biology, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Maria M Alves
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands; Department of Pediatric Surgery, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, the Netherlands.
| |
Collapse
|
43
|
Tao B, Li X, Hao M, Tian T, Li Y, Li X, Yang C, Li Q, Feng Q, Zhou H, Zhao Y, Wang D, Liu W. Organoid-Guided Precision Medicine: From Bench to Bedside. MedComm (Beijing) 2025; 6:e70195. [PMID: 40321594 PMCID: PMC12046123 DOI: 10.1002/mco2.70195] [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: 07/31/2024] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 05/08/2025] Open
Abstract
Organoid technology, as an emerging field within biotechnology, has demonstrated transformative potential in advancing precision medicine. This review systematically outlines the translational trajectory of organoids from bench to bedside, emphasizing their construction methodologies, key regulatory factors, and multifaceted applications in personalized healthcare. By recapitulating physiological architectures and disease phenotypes through three-dimensional culture systems, organoids leverage natural and synthetic scaffolds, stem cell sources, and spatiotemporal cytokine regulation to model tissue-specific microenvironments. Diverse organoid types-including skin, intestinal, lung, and tumor organoids-have facilitated breakthroughs in modeling tissue development, drug efficacy and toxicity screening, disease pathogenesis studies, and patient-tailored diagnostics. For instance, patient-derived tumor organoids preserve tumor heterogeneity and genomic profiles, serving as predictive platforms for individualized chemotherapy responses. In precision medicine, organoid-guided multiomics analyses identify actionable biomarkers and resistance mechanisms, while clustered regularly interspaced short palindromic repeats-based functional screens optimize therapeutic targeting. Despite preclinical successes, challenges persist in standardization, vascularization, and ethical considerations. Future integration of artificial intelligence, microfluidics, and spatial transcriptomics will enhance organoid scalability, reproducibility, and clinical relevance. By bridging molecular insights with patient-specific therapies, organoids are poised to revolutionize precision medicine, offering dynamic platforms for drug development, regenerative strategies, and individualized treatment paradigms.
Collapse
Affiliation(s)
- Boqaing Tao
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| | - Xiaolan Li
- Laboratory of Allergy and Precision MedicineChengdu Institute of Respiratory Healththe Third People's Hospital of ChengduAffiliated Hospital of Southwest Jiaotong UniversityChengduChina
| | - Ming Hao
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| | - Tian Tian
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Yuyang Li
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| | - Xiang Li
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| | - Chun Yang
- College of Basic MedicineBeihua UniversityJilinChina
| | - Qirong Li
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Qiang Feng
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Hengzong Zhou
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Yicheng Zhao
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Dongxu Wang
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
- Zhichuang Gene Editing Animal Model Research CenterWenzhou Institute of TechnologyWenzhouChina
| | - Weiwei Liu
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| |
Collapse
|
44
|
Le Maître M, Guerrier T, Sanges S, Chepy A, Collet A, Launay D. Beyond circulating B cells: Characteristics and role of tissue-infiltrating B cells in systemic sclerosis. Autoimmun Rev 2025; 24:103782. [PMID: 40010623 DOI: 10.1016/j.autrev.2025.103782] [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: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025]
Abstract
B cells play a key role in the pathophysiology of systemic sclerosis (SSc). While they are less characterized than their circulating counterparts, tissue-infiltrating B cells may have a more direct pathological role in tissues. In this review, we decipher the multiple evidence of B cells infiltration in the skin and lungs of SSc patients and animal models of SSc but also of other chronic fibrotic diseases with similar pathological mechanisms such as chronic graft versus host disease, idiopathic pulmonary fibrosis or morphea. We also recapitulate the current knowledge about mechanisms of B cells infiltration and their functions in tissues. Finally, we discuss B cell targeted therapies, and their specific impact on infiltrated B cells. Understanding the local consequences of infiltrating B cells is an important step for a better management of patients and the improvement of therapies in SSc.
Collapse
Affiliation(s)
- Mathilde Le Maître
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France.
| | - Thomas Guerrier
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
| | - Sébastien Sanges
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France; CHU Lille, Département de Médecine Interne et Immunologie Clinique, F-59000 Lille, France; Centre National de Référence Maladies Auto-immunes Systémiques Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), F-59000 Lille, France; Health Care Provider of the European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases (ReCONNET), France
| | - Aurélien Chepy
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France; CHU Lille, Département de Médecine Interne et Immunologie Clinique, F-59000 Lille, France; Centre National de Référence Maladies Auto-immunes Systémiques Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), F-59000 Lille, France; Health Care Provider of the European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases (ReCONNET), France
| | - Aurore Collet
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France; CHU Lille, Institut d'Immunologie, Pôle de Biologie Pathologie Génétique, Lille, France
| | - David Launay
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France; CHU Lille, Département de Médecine Interne et Immunologie Clinique, F-59000 Lille, France; Centre National de Référence Maladies Auto-immunes Systémiques Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), F-59000 Lille, France; Health Care Provider of the European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases (ReCONNET), France
| |
Collapse
|
45
|
Finger AM, Hendley AM, Figueroa D, Gonzalez H, Weaver VM. Tissue mechanics in tumor heterogeneity and aggression. Trends Cancer 2025:S2405-8033(25)00096-2. [PMID: 40307158 DOI: 10.1016/j.trecan.2025.04.004] [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: 08/27/2024] [Revised: 03/10/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025]
Abstract
Tumorigenesis ensues within a heterogeneous tissue microenvironment that promotes malignant transformation, metastasis and treatment resistance. A major feature of the tumor microenvironment is the heterogeneous population of cancer-associated fibroblasts and myeloid cells that stiffen the extracellular matrix. The heterogeneously stiffened extracellular matrix in turn activates cellular mechanotransduction and creates a hypoxic and metabolically hostile microenvironment. The stiffened extracellular matrix and elevated mechanosignaling also drive tumor aggression by fostering tumor cell growth, survival, and invasion, compromising antitumor immunity, expanding cancer stem cell frequency, and increasing mutational burden, which promote intratumor heterogeneity. Delineating the molecular mechanisms whereby tissue mechanics regulate these phenotypes should help to clarify the basis for tumor heterogeneity and cancer aggression and identify novel therapeutic targets that could improve patient outcome. Here, we discuss the role of the extracellular matrix in driving cancer aggression through its impact on tumor heterogeneity.
Collapse
Affiliation(s)
- Anna-Marie Finger
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA 94143; Current address: Liver Disease Research, Global Drug Discovery, Novo Nordisk A/S, Malov, Denmark
| | - Audrey Marie Hendley
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA 94143
| | - Diego Figueroa
- Department of Radiation Oncology, Department of Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Hugo Gonzalez
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA 94143; Current address: Laboratory of Tumor Microenvironment and Metastasis, Centro Ciencia & Vida, Santiago, Chile
| | - Valerie Marie Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA 94143; Department of Radiation Oncology, Department of Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
46
|
Zhao Q, Shao M, Ma L, Zhou R. Insights into Modeling Inflammatory Bowel Disease from Stem Cell Derived Intestinal Organoids. Stem Cell Rev Rep 2025:10.1007/s12015-025-10887-8. [PMID: 40299197 DOI: 10.1007/s12015-025-10887-8] [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] [Accepted: 04/13/2025] [Indexed: 04/30/2025]
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease (CD) and ulcerative colitis (UC), is a multifactorial, immune-mediated condition marked by chronic gastrointestinal inflammation. This condition significantly impairs patients' quality of life and represents a major public health challenge globally. Pathogenesis of IBD arises from complex interplay among genetic predisposition, environmental factors, immune dysregulation, and microbial dysbiosis. Although significant strides have been made in unraveling these mechanisms, existing therapeutic options remain inadequate in addressing the full spectrum of clinical needs, underscoring the urgent demand for innovative strategies. Regenerative medicine has emerged as a promising frontier, offering novel tools for therapeutic development. We briefly consolidated current knowledge on IBD pathogenesis and treatments, emphasized the pivotal potential of human intestinal organoids (including adult stem cell-derived organoids and pluripotent stem cell- derived organoids) as a robust platform for mechanistic studies and treatment exploration. Leveraging this technology, we aim to advance personalized and next-generation therapies for IBD.
Collapse
Affiliation(s)
- Qi Zhao
- The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou, Zhejiang Province, China
| | - Miaoli Shao
- The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou, Zhejiang Province, China
| | - Lisha Ma
- The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou, Zhejiang Province, China
| | - Renfang Zhou
- The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou, Zhejiang Province, China.
| |
Collapse
|
47
|
Wang R, Zhu J, Zhou J, Li J, Wang M, Wu Y, Zhao D, Chen X, Chen X, Wang Y, Zou J. Bioinspired Claw-Engaged Adhesive Microparticles Armed with γGC Alleviate Ulcerative Colitis via Targeted Suppression of Macrophage Ferroptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2503903. [PMID: 40298904 DOI: 10.1002/advs.202503903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/13/2025] [Indexed: 04/30/2025]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease, characterized by focal iron overload. Herein, we reported that γ-glutamylcysteine (γGC) deletion in UC lesions intensified the disease by depleting intracellular GSH to induce macrophage ferroptosis, leading to macrophage M1 reprogramming and eventually exacerbating inflammation. To counteract this, the advanced microparticles (MPs)-based delivery system is selected to encapsulate γGC. The resulting γGC-MPs displayed the same porous and spiky morphology as their substrate's natural pollens, resulting in improved intestinal adhesion and enhanced lesion contact of γGC-MPs. Our results demonstrated that exogenous γGC supplementation could inhibit macrophage M1 polarization by restraining ferroptosis, as well as suppressing the PI3K/AKT pathway and TNF signaling pathway. Compared with free γGC, γGC-MPs significantly alleviated typical UC symptoms in dextran sulfate sodium (DSS)-induced colitis, evidenced by reduced intestinal inflammation, restored intestinal barrier function, and improved microbiota composition. Consequently, this study addressed critical gaps in understanding the causes of ferroptosis and its impact on macrophage reprogramming in UC, offering a novel synergistic therapeutic strategy for UC.
Collapse
Affiliation(s)
- Rong Wang
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, 210046, China
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan, 410219, China
| | - Jianwei Zhu
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, 210046, China
- Departments of Diagnostic Radiology Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Jinyi Zhou
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, 210046, China
| | - Jinyang Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Min Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Yuqi Wu
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, 210046, China
| | - Danshan Zhao
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, Jiangsu, 210046, China
| | - Xiancheng Chen
- Department of Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210029, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
| | - Yuetong Wang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, Jiangsu, 210046, China
| | - Jianhua Zou
- Departments of Diagnostic Radiology Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
| |
Collapse
|
48
|
Gonzalez-Acera M, Patankar JV, Erkert L, Cineus R, Gamez-Belmonte R, Leupold T, Bubeck M, Bao LL, Dinkel M, Wang R, Schickedanz L, Limberger H, Stolzer I, Gerlach K, Diemand L, Mascia F, Gupta P, Naschberger E, Koop K, Plattner C, Sturm G, Weigmann B, Günther C, Wirtz S, Stürzl M, Hildner K, Kühl AA, Siegmund B, Gießl A, Atreya R, Hegazy AN, Trajanoski Z, Neurath MF, Becker C. Integrated multimodel analysis of intestinal inflammation exposes key molecular features of preclinical and clinical IBD. Gut 2025:gutjnl-2024-333729. [PMID: 40301114 DOI: 10.1136/gutjnl-2024-333729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 04/16/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND IBD is a chronic inflammatory condition driven by complex genetic and immune interactions, yet preclinical models often fail to fully recapitulate all aspects of the human disease. A systematic comparison of commonly used IBD models is essential to identify conserved molecular mechanisms and improve translational relevance. OBJECTIVE We performed a multimodel transcriptomic analysis of 13 widely used IBD mouse models to uncover coregulatory gene networks conserved between preclinical colitis/ileitis and human IBD and to define model-specific and conserved cellular, subcellular and molecular signatures. DESIGN We employed comparative transcriptomic analyses with curated and a priori statistical correlative methods between mouse models versus IBD patient datasets at both bulk and single-cell levels. RESULTS We identify IBD-related pathways, ontologies and cellular compositions that are translatable between mouse models and patient cohorts. We further describe a conserved core inflammatory signature of IBD-associated genes governing T-cell homing, innate immunity and epithelial barrier that translates into the new mouse gut Molecular Inflammation Score (mMIS). Moreover, specific mouse IBD models have distinct signatures for B-cell, T-cell and enteric neurons. We discover that transcriptomic relatedness of models is a function of the mode of induction, not the canonical immunotype (Th1/Th2/Th17). Moreover, the model compendium database is made available as a web explorer (http://trr241.hosting.rrze.uni-erlangen.de/SEPIA/). CONCLUSION This integrated multimodel approach provides a framework for systematically assessing the molecular landscape of intestinal inflammation. Our findings reveal conserved inflammatory circuits, refine model selection, offering a valuable resource for the IBD research community.
Collapse
Affiliation(s)
- Miguel Gonzalez-Acera
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Jay V Patankar
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Lena Erkert
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Roodline Cineus
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Deutsches Rheuma-Forschungszentrum, ein Institut der Leibniz-Gemeinschaft, Berlin, Germany
| | - Reyes Gamez-Belmonte
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Tamara Leupold
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Marvin Bubeck
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Li-Li Bao
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Martin Dinkel
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Ru Wang
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Laura Schickedanz
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Heidi Limberger
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Iris Stolzer
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Katharina Gerlach
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Leonard Diemand
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Fabrizio Mascia
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Pooja Gupta
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Bayern, Germany
| | - Elisabeth Naschberger
- Department of Surgery, Universitätsklinikum, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Kristina Koop
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Christina Plattner
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Gregor Sturm
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Benno Weigmann
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Claudia Günther
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Michael Stürzl
- Department of Surgery, Universitätsklinikum, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Kai Hildner
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Anja A Kühl
- iPATH.Berlin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Britta Siegmund
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas Gießl
- Department of Ophthalmology, Universitätsklinikum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Raja Atreya
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Ahmed N Hegazy
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Deutsches Rheuma-Forschungszentrum, ein Institut der Leibniz-Gemeinschaft, Berlin, Germany
| | - Zlatko Trajanoski
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus F Neurath
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsche Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universitat Erlangen-Nurnberg, Erlangen, Germany
| |
Collapse
|
49
|
Mörseburg A, Zhao Y, Kentistou KA, Perry JRB, Ong KK, Day FR. Genetic determinants of proteomic aging. NPJ AGING 2025; 11:30. [PMID: 40287427 PMCID: PMC12033249 DOI: 10.1038/s41514-025-00205-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 02/21/2025] [Indexed: 04/29/2025]
Abstract
Changes in the proteome and its dysregulation have long been known to be a hallmark of aging. We derived a proteomic aging trait using data on 1459 plasma proteins from 44,435 UK Biobank individuals measured using an antibody-based assay. This metric is strongly associated with four age-related disease outcomes, even after adjusting for chronological age. Survival analysis showed that one-year older proteomic age, relative to chronological age, increases all-cause mortality hazard by 13 percent. We performed a genome-wide association analysis of proteomic age acceleration (proteomic aging trait minus chronological age) to identify its biological determinants. Proteomic age acceleration showed modest genetic correlations with four epigenetic clocks (Rg = 0.17 to 0.19) and telomere length (Rg = -0.2). Once we removed associations that were explained by a single pQTL, we were left with three signals mapping to BRCA1, POLR2A and TET2 with apparent widespread effects on plasma proteomic aging. Genetic variation at these three loci has been shown to affect other omics-related aging measures. Mendelian randomisation analyses showed causal effects of higher BMI and type 2 diabetes on faster proteomic age acceleration. This supports the idea that obesity and other features of metabolic syndrome have an adverse effect on the processes of human aging.
Collapse
Affiliation(s)
- Alexander Mörseburg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| | - Yajie Zhao
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Katherine A Kentistou
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - John R B Perry
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Ken K Ong
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Felix R Day
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| |
Collapse
|
50
|
Zhu X, Zhao L, Teng F, Meng S, Xie M. ScAGCN: Graph Convolutional Network with Adaptive Aggregation Mechanism for scRNA-seq Data Dimensionality Reduction. Interdiscip Sci 2025:10.1007/s12539-025-00702-w. [PMID: 40281370 DOI: 10.1007/s12539-025-00702-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 04/29/2025]
Abstract
With the development of single-cell RNA-sequencing (scRNA-seq) technology, scRNA-seq data analysis suffers huge challenges due to large scale, high dimensionality, high noise, and high sparsity. To achieve accurately embedded representation in the large-scale scRNA-seq data, we try to design a novel graph convolutional network with an adaptive aggregation mechanism. Based on the assumption that the aggregation order of different cells would be different, a graph convolutional network with an adaptive aggregation-based dimensionality reduction algorithm for scRNA-seq data is developed, named scAGCN. In scAGCN, a preprocessing consisting of quality control and feature selection is implemented. Then, an approximate nearest neighbor graph is rapidly constructed. Finally, a graph convolutional network with an adaptive aggregation mechanism is constructed, in which the neighborhood selection strategy based on node distribution and similarity boxplots is designed, and the aggregation function is optimized by defining a similarity measurement between neighborhood nodes and the central node. The results show that scAGCN outperforms existing dimensionality reduction methods on 15 real scRNA-seq datasets, especially in 10 large-scale scRNA-seq datasets.
Collapse
Affiliation(s)
- Xiaoshu Zhu
- School of Computer and Information Security, Guangxi Key Laboratory of Trusted Software, Guilin University of Electronic Technology, Guilin, 541004, China.
| | - Liquan Zhao
- School of Computer and Information Security, Guangxi Key Laboratory of Trusted Software, Guilin University of Electronic Technology, Guilin, 541004, China
| | - Fei Teng
- School of Computer and Information Security, Guangxi Key Laboratory of Trusted Software, Guilin University of Electronic Technology, Guilin, 541004, China
| | - Shuang Meng
- School of Computer Science and Engineering, Guangxi Normal University, Guilin, 541006, China
| | - Miao Xie
- School of Computer Science and Engineering, Yulin Normal University, Yulin, 537000, China.
| |
Collapse
|