1
|
Pawlina-Tyszko K, Semik-Gurgul E, Podstawski P, Herc W, Witkowski M, Ropka-Molik K. Altered expression of collagen gene family members and its epigenetic background in equine Sarcoids. Res Vet Sci 2025; 190:105656. [PMID: 40288239 DOI: 10.1016/j.rvsc.2025.105656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 03/18/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025]
Abstract
Alterations in the genes involved in the creation of the extracellular matrix (ECM) were observed in our earlier transcriptome studies of sarcoids and their cell culture model. For a complete characterization of the underlying molecular pathways, it is imperative to comprehend the involvement of ECM modifications in the oncogenic transformation of sarcoid fibroblasts. Thus, the aim of this investigation was to describe the expression patterns of a set of genes that are essential for the rearrangements of the extracellular matrix, namely collagen genes, and elucidate possible mechanisms underlying the observed disruptions. To this end, we applied the RT-qPCR method on BPV-negative skin samples and sarcoid samples (n = 6 and 7; respectively) to perform relative quantification of the expression level of eight genes belonging to the collagen family and carried out an integrative analysis of the obtained data with previously characterized epigenetic signatures. The results showed aberrations in the level of chosen collagen genes in the sarcoids compared to the control, manifesting in their elevated levels in the tumor samples (p-value≤0.05). The upregulation of Col1A2, Col11A1, Col6A3, Col5A2, Col4A1, Col6A6, Col5A1, Col6A2 genes was detected in sarcoid samples. The identified changes were statistically significant (p-value≤0.05) and ranged from 1.43 (Col6A2) to 1.88 (Col6A3). Further investigation into the potential involvement of epigenetic mechanisms in the regulation of collagen gene levels in sarcoids revealed compelling evidence of DNA methylation and microRNAs playing significant roles. The findings suggest a complex interplay between gene expression, epigenetic regulation, and the dysregulation of the ECM in sarcoid pathogenesis.
Collapse
Affiliation(s)
- Klaudia Pawlina-Tyszko
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1 st, 32-083 Balice, Poland
| | - Ewelina Semik-Gurgul
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1 st, 32-083 Balice, Poland
| | - Przemysław Podstawski
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1 st, 32-083 Balice, Poland
| | - Weronika Herc
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1 st, 32-083 Balice, Poland
| | - Maciej Witkowski
- Institute of Veterinary Medicine, University Centre of Veterinary Medicine JU-AU, Mickiewicza 24/28 st., 30-059 Kraków, Poland; Horse Clinic Służewiec, Puławska 266 st, 02-684 Warsaw, Poland
| | - Katarzyna Ropka-Molik
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1 st, 32-083 Balice, Poland.
| |
Collapse
|
2
|
Kolliopoulos V, Mikos AG. Decellularized extracellular matrix as a drug delivery carrier. J Control Release 2025; 382:113661. [PMID: 40139392 DOI: 10.1016/j.jconrel.2025.113661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 03/18/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025]
Abstract
Tissue engineering and regenerative medicine approaches seek to enhance biomaterial mimicry with the goal of driving cell recruitment, proliferation, and differentiation. Decellularized extracellular matrix (dECM) biomaterials have emerged as a promising platform for biomaterials development as they capture the complexity of native tissues and offer a rich environment of signals to guide cellular responses. However, the decellularization process can affect both the structure and composition of the ECM. Recent efforts have focused on leveraging dECM as drug delivery carriers for controlled release of bioactive molecules. This review highlights current strategies for incorporating therapeutic agents into dECM which include encapsulation within hydrogel formulations, direct bulk absorption of biomolecules, and affinity-based binding and conjugation. Each method offers unique advantages for modulating release profiles, which can range from rapid initial burst to prolonged, sustained release, depending on factors such as crosslinking density, degradation rate, and specific interactions of biomolecules with dECM components such as glycosaminoglycans.
Collapse
Affiliation(s)
- Vasiliki Kolliopoulos
- Department of Bioengineering, Rice University, Houston, TX 77030, United States of America
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX 77030, United States of America.
| |
Collapse
|
3
|
von Mentzer U, Havemeister F, Råberg L, Kothuru Chinnadurai H, Erensoy G, Esbjörner EK, Stubelius A. Glycosylation-driven interactions of nanoparticles with the extracellular matrix: Implications for inflammation and drug delivery. BIOMATERIALS ADVANCES 2025; 171:214230. [PMID: 39983501 DOI: 10.1016/j.bioadv.2025.214230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/22/2025] [Accepted: 02/13/2025] [Indexed: 02/23/2025]
Abstract
Cationic nanoparticles (NPs) are emerging as promising carriers for intra-articular drug delivery, particularly for osteoarthritis (OA) where treatment options are limited. However, the clinical translation is challenged by an incomplete understanding of NP interactions within pathological environments. While the influence of the protein coronas on NP behavior has been extensively studied, the specific role of glycoproteins in the extracellular matrix (ECM) remains underexplored, representing a significant knowledge gap. This study investigates how glycosylation-driven interactions between polymeric NPs and enzyme-degraded cartilage biomolecules such as glycosaminoglycans (GAGs) affect NP-ECM aggregate formation and subsequent inflammatory responses. Using an ex vivo model of cartilage degradation induced by catabolic enzymes- hyaluronidase, ADAMTS5 and collagenase- a novel model system was developed to specifically study the behavior of small (<10 nm) and large (∼270 nm) cationic NPs in glycoprotein-enriched environments. Atomic force microscopy and dynamic light scattering revealed distinct mesh-like structures formed by the NP aggregates following different enzymatic treatments, confirming the adsorption of glycosylated fragments onto the particles. While total protein content showed minimal differences between NP samples, smaller NPs demonstrated a prominent association with GAGs such as hyaluronic acid and aggrecan, as demonstrated by circular dichroism. These ECM-NP interactions significantly influenced the immunological response, as evidenced by differential cytokine production from macrophages exposed to the aggregates. Our findings underscore the crucial, yet underappreciated, role of glycoproteins in determining NP behavior in pathological environments. Accounting for glycoprotein interactions into the design of nanomaterial and drug delivery systems could significantly improve therapeutic outcomes by enhanced targeting precision, optimized delivery, and effectively modulating immune responses in OA and other complex diseases.
Collapse
Affiliation(s)
- Ula von Mentzer
- Division of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Fritjof Havemeister
- Division of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Loise Råberg
- Division of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | | | - Gizem Erensoy
- Division of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Elin K Esbjörner
- Division of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Alexandra Stubelius
- Division of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden.
| |
Collapse
|
4
|
Bounaama A, Djerdjouri B. Matrix metalloproteinase 9 implication during colorectal carcinogenesis. Effect of doxycycline. Fundam Clin Pharmacol 2025; 39:e70012. [PMID: 40273927 DOI: 10.1111/fcp.70012] [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: 02/12/2025] [Revised: 03/14/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Matrix metalloproteinases (MMPs), including MMP9, play a significant role in colorectal cancer (CRC) progression, mainly by extracellular matrix remodeling. However, little is known about MMP9 role in aberrant crypt foci (ACF) cluster formation, the earliest colon preneoplastic lesions. AIMS AND METHODS We conducted a bioinformatics analysis of MMPs expression in CRC using Gene Expression Profiling Interactive Analysis2 (GEPIA2). Subsequently, we investigated MMP9 expression during the early stage of colon carcinogenesis in mice and assessed the effect of doxycycline (DOX), a global inhibitor of MMPs, on ACF cluster formation. Thus, NMRI mice received two weekly injections of 1,2-Dimethylhydrazine (DMH, 20 mg/kg, subcutaneously), followed or not by DOX (100 mg/kg, orally, from the 4th to the 6th week). RESULTS GEPIA2 analysis indicated that among the 28 identified MMPs with collagenase and doxycycline-sensitive activities, MMPs 1, 3, 7, 9, and 13 were overexpressed in CRC tissues. Moreover, only MMP1 and MMP9 correlated well with collagen expression in colorectal tumors. In vivo, methylene blue-stained DMH-treated colons revealed multiple ACF clusters at week 6, associated with mucosa remodeling and sustained nitrosative stress as attested by enhanced collagen fibers, malondialdehyde level, and nitrotyrosine deposits. Pyrosequencing showed increased methylation at the tenth CpG site of the MMP9 promoter, which was associated with increased MMP9 expression. Interestingly, DOX attenuated the number and size of ACF clusters and mucosa remodeling without rebalancing nitrosative stress. CONCLUSION Overexpression of MMP9 occurs early during colorectal carcinogenesis, and doxycycline may control the pathological remodeling of colon mucosa into ACF clusters by attenuating MMP9 activity.
Collapse
Affiliation(s)
- Abdelkader Bounaama
- Tamayouz_Laboratory of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), Algiers, Algeria
| | - Bahia Djerdjouri
- Tamayouz_Laboratory of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Sciences and Technology Houari Boumediene (USTHB), Algiers, Algeria
| |
Collapse
|
5
|
Capik O, Karatas OF. Pathways and outputs orchestrated in tumor microenvironment cells by hypoxia-induced tumor-derived exosomes in pan-cancer. Cell Oncol (Dordr) 2025; 48:539-557. [PMID: 39928285 PMCID: PMC12119682 DOI: 10.1007/s13402-025-01042-z] [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] [Accepted: 01/30/2025] [Indexed: 02/11/2025] Open
Abstract
Hypoxia is a critical microenvironmental condition that plays a major role in driving tumorigenesis and cancer progression. Increasing evidence has revealed novel functions of hypoxia in intercellular communication. The hypoxia induced tumor derived exosomes (hiTDExs) released in high quantities by tumor cells under hypoxia are packed with unique cargoes that are essential for cancer cells' interactions within their microenvironment. These hiTDExs facilitate not only immune evasion but also promote cancer cell growth, survival, angiogenesis, EMT, resistance to therapy, and the metastatic spread of the disease. Nevertheless, direct interventions targeting hypoxia signaling in cancer therapy face challenges related to tumor progression and resistance, limiting their clinical effectiveness. Therefore, deepening our understanding of the molecular processes through which hiTDExs remodels tumors and their microenvironment, as well as how tumor cells adjust to hypoxic conditions, remains essential. This knowledge will pave the way for novel approaches in treating hypoxic tumors. In this review, we discuss recent work revealing the hiTDExs mediated interactions between tumor and its microenvironment. We have described key hiTDExs cargos (lncRNA, circRNAs, cytokines, etc.) and their targets in the receipt cells, responsible for various biological effects. Moreover, we emphasized the importance of hiTDExs as versatile elements of cell communication in the tumor microenvironment. Finally, we highlighted the effects of hiTDExs on the molecular changes in target cells by executing molecular cargo transfer between cells and altering signaling pathways. Currently, hiTDExs show promise in the treatment of diseases. Understanding the molecular processes through which hiTDExs influence tumor behavior and their microenvironment, along with how tumor cells adapt to and survive in low-oxygen conditions, remains a central focus in cancer research, paving the way for innovative strategies in treating hypoxic tumors and enhancing immunotherapy.
Collapse
Affiliation(s)
- Ozel Capik
- Department of Molecular Biology and Genetics, Erzurum Technical University, Omer Nasuhi Bilmen Mah. Havaalani Yolu Cad. No: 53 Yakutiye, Erzurum, Turkey.
- Cancer Therapeutics Laboratory, High Technology Application and Research Center, Erzurum Technical University, Erzurum, Turkey.
| | - Omer Faruk Karatas
- Department of Molecular Biology and Genetics, Erzurum Technical University, Omer Nasuhi Bilmen Mah. Havaalani Yolu Cad. No: 53 Yakutiye, Erzurum, Turkey
- Cancer Therapeutics Laboratory, High Technology Application and Research Center, Erzurum Technical University, Erzurum, Turkey
| |
Collapse
|
6
|
Wu L, Coletta DK. Obesity and type 2 diabetes mellitus: insights from skeletal muscle extracellular matrix remodeling. Am J Physiol Cell Physiol 2025; 328:C1752-C1763. [PMID: 40244268 DOI: 10.1152/ajpcell.00154.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/23/2024] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are metabolic diseases at epidemic proportions. The economic burden for these diseases is at an all-time high, and as such, there is an urgent need for advancements in identifying targets for treating these complex disorders. The extracellular matrix (ECM), comprising collagen, fibronectin, laminin, elastin, and proteoglycan, surrounds skeletal muscles and plays a critical role in maintaining tissue homeostasis by providing structural support and facilitating cell-to-cell communication. Disruption of the ECM signaling results in changes to its micro/macroenvironment, thereby modifying tissue homeostasis. Skeletal muscle ECM remodeling has been shown to be associated with insulin resistance, an underlying feature of obesity and T2DM. This narrative review explores the critical components of skeletal muscle ECM and its accumulation and remodeling in metabolic diseases. In addition, we discuss potential treatments to mitigate the effects of ECM remodeling in skeletal muscle. We conclude that targeting ECM remodeling in skeletal muscle represents a promising yet underexplored therapeutic avenue in the management of metabolic disorders.
Collapse
Affiliation(s)
- Linda Wu
- Department of Physiology, University of Arizona, Tucson, Arizona, United States
| | - Dawn K Coletta
- Department of Physiology, University of Arizona, Tucson, Arizona, United States
- Division of Endocrinology, Department of Medicine, University of Arizona, Tucson, Arizona, United States
- Center for Disparities in Diabetes, Obesity, and Metabolism, University of Arizona, Tucson, Arizona, United States
| |
Collapse
|
7
|
Xiong S, Zhang S, Yue N, Cao J, Wu C. CAR-T cell therapy in the treatment of relapsed or refractory primary central nervous system lymphoma: recent advances and challenges. Leuk Lymphoma 2025; 66:1045-1057. [PMID: 39898872 DOI: 10.1080/10428194.2025.2458214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 01/01/2025] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
Primary central nervous system lymphoma (PCNSL) is a rare and aggressive lymphoma that is isolated in the central nervous system (CNS) or vitreoretinal space. High-dose methotrexate (HD-MTX)-based immunochemotherapy is the frontline for its treatment, with a high early response rate. However, relapsed or refractory (R/R) patients present numerous difficulties and challenges in clinical treatment. Chimeric antigen receptor (CAR)-T cells offer a promising option for the treatment of hematologic malignancies, especially in the R/R B-cell lymphoma and multiple myeloma. Despite the exclusion of most PCNSL cases from pivotal CAR-T cell trials due to their specific tumor microenvironment (TME), available preclinical and clinical studies with small cohorts suggest an overall acceptable safety profile and remarkable anti-tumor effects. In this review, we will provide the development process of CAR-T cells and summarize the research progress, limitations, and future perspectives of CAR-T cell therapy in patients with R/R PCNSL.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Central Nervous System Neoplasms/therapy
- Central Nervous System Neoplasms/pathology
- Central Nervous System Neoplasms/immunology
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Tumor Microenvironment/immunology
- Neoplasm Recurrence, Local/therapy
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/immunology
- Drug Resistance, Neoplasm
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Treatment Outcome
- Lymphoma/therapy
- Lymphoma/pathology
- Lymphoma/immunology
- Animals
- Clinical Trials as Topic
- Receptors, Antigen, T-Cell
Collapse
Affiliation(s)
- Shuzhen Xiong
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | | | | | | | | |
Collapse
|
8
|
Zhao Z, Qiu S, Zhang X, Liu S, Wang L, Guan H, He J, Hu Y, Li X, Luo S, Chen Z, Mo T, Zhang Y, Zhao X, Pan Y, Ding H, Cao J, Pan J. Characterization of a novel cell line established from mice gastrointestinal stromal model by chemical induction. Transl Oncol 2025; 56:102388. [PMID: 40233502 PMCID: PMC12022689 DOI: 10.1016/j.tranon.2025.102388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 02/03/2025] [Accepted: 04/05/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are a type of tumor that originates from gastrointestinal mesenchymal tissue. Although several somatic or germline mutation GIST mice were established, however, there is still a lack of an authentic mice GIST cell lines for further experimental study. METHODS We developed a chemically induced C57BL/6 J GIST model using 3- methylcholanthrene. Tumor characteristics were confirmed through histology and IHC. Primary cells were isolated to establish the mGSTc01 cell line, and molecular profiling was conducted. Additionally, we established GIST model in immunocompetent mice to evaluate their sensitivity to imatinib. RESULTS Our study successfully developed a chemically induced murine GIST model, characterized by positive staining of c-kit and DOG-1. The mGSTc01 monoclonal cell line exhibited slender morphology and expressed the c-kit marker, Whole exome sequencing uncovered mutations of Lamb1, MMP9, and c-kit in GIST cells and provided a detailed picture of the entire genome's copy number variations. RNA sequencing indicated genes associated with cell adhesion and focal adhesion were enriched in mGSTc01 cells. The mGSTc01 cells demonstrated obvious malignant behaviors, notably elevated migration, adhesion, and proliferation. In immunocompetent mice, subcutaneous xenografts not only reserved the aggressive phenotype but also displayed a response to imatinib, underscoring the model's applicability for advancing therapeutic research. CONCLUSION We firstly established a mGSTc01 cell line derived from C57BL/6 J mice GIST tumor offers, which closely mimicking human disease characteristics. It is a potent platform for investigating tumor microenvironment of GIST in mice model, and provides a novel way for new therapeutic discoveries in GIST.
Collapse
Affiliation(s)
- Zhan Zhao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Shenghui Qiu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China; Department of General Surgery, Guangzhou First People's Hospital, Guangzhou, 510180, PR China
| | - Xiangwei Zhang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Shijin Liu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Lu Wang
- Institute of Precision Cancer Medicine and Pathology, Jinan University Medical College, Guangzhou, Guangdong, 510632, PR China
| | - Hanyang Guan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Jiashuai He
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Yangzhi Hu
- The Affiliated Hospital of Xiangnan University, Chenzhou, Hunan, PR China
| | - Xiaobo Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
| | - Simin Luo
- Department of Bone and Joint Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Zuyang Chen
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Tianmu Mo
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Yiran Zhang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Xiaoxu Zhao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Yunlong Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China
| | - Hui Ding
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China.
| | - Jie Cao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China; Department of General Surgery, Guangzhou First People's Hospital, Guangzhou, 510180, PR China.
| | - Jinghua Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, 510632, Guangzhou, Guangdong, PR China.
| |
Collapse
|
9
|
Lin Y, Chen J, Tan J, Yu Z, Pi R, Xiong J, Ding Y, Chen M, Bai X. Pericytes in the Pulmonary Microenvironment: Guardians or Adversaries? Lung 2025; 203:65. [PMID: 40448710 DOI: 10.1007/s00408-025-00820-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 05/11/2025] [Indexed: 06/02/2025]
Abstract
Pericytes, specialized mural cells residing within the basement membrane of pulmonary microvessels, participate in various biological processes, including vascular homeostasis, immunomodulation, and tissue repair. However, these beneficial physiological roles can be detrimental under pathological conditions. Numerous pulmonary fibrosis models have demonstrated pericyte differentiation into scar-forming myofibroblasts, leading to collagen deposition and matrix remodeling, thereby contributing to tissue fibrosis. Similarly, pericytes play crucial roles in inflammatory diseases. This review aims to explore the dual roles of pericytes in the lung and the underlying mechanisms of their role conversion, providing insights for developing therapeutic strategies targeting these cells.
Collapse
Affiliation(s)
- Yan Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiaqi Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiale Tan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zihang Yu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ruozheng Pi
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jingrong Xiong
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yi Ding
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| | - Minfeng Chen
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China.
| | - Xue Bai
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
10
|
Liang F, Yu Q, Li T, Meng H, Huang X, Sheng S, Jiang Y, Ren F. Functionalized liposomes induce cascade degradation of extracellular matrix by hyaluronidase and photodynamic therapy for synergistic suppression of breast cancer. Int J Biol Macromol 2025:144794. [PMID: 40449783 DOI: 10.1016/j.ijbiomac.2025.144794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 05/27/2025] [Accepted: 05/28/2025] [Indexed: 06/03/2025]
Abstract
Treatment of breast cancer (BC) remains to face clinical challenges due to poor tumor penetration of therapeutic agents, which is a direct consequence of the high content of hyaluronan (HA) within the extracellular matrix (ECM) of tumor tissues. Herein, we reported the synthesis of phototherapy liposomes which integrated hyaluronidase (HAase) conjugated with a matrix metalloproteinase 2 (MMP-2) responsive peptide in their phospholipid membrane, and preloaded with indocyanine green (ICG), CaO2, and L-buthionine sulfoximine (BSO). Under near-infrared (NIR) laser irradiation, the responsive release of HAase and photodynamic therapy (PDT) effect generated by ICG enabled the controlled degradation of HA in the ECM. Moreover, the photothermal effect of ICG inactivated the residual HAase. This sequential cascade of events facilitated the highly efficient penetration of liposomes into tumor and degrades HA into oligosaccharides (oHA) through adjusting NIR laser irradiation intensity and duration. The phototherapy liposomes also alleviated hypoxia and depleted glutathione (GSH) within the tumor microenvironment. Combined treatment with our liposomes and laser irradiation led to a cell survival rate of <20 % for both 4 T1 and MDA-MB-231 cells. In 4 T1 tumor-bearing mice, the liposome group under NIR light irradiation significantly enhanced tumor ablation, achieving an inhibition rate of 64.9 %, demonstrating that the combination of PDT/PTT with oHA improves the therapeutic efficacy. As expected, the generation of oHA reduced the adverse effects triggered by HAase. These phototherapy liposomes, by promoting the penetration of PDT/PTT drugs, offer a promising strategy to enhance the treatment effectiveness for BC.
Collapse
Affiliation(s)
- Futu Liang
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qin Yu
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Tianyang Li
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Haimei Meng
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xinfeng Huang
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Sizhe Sheng
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yufei Jiang
- First clinical medicine college, Southern Medical University, Guangzhou 510515, China
| | - Fei Ren
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
11
|
Zhang X, Zheng J, Zhang L, Zhang J, Feng L, Zhang L, Huang X. Transcriptomic and proteomic integrated analysis reveals molecular mechanisms of 3D bioprinted vaginal scaffolds in vaginal regeneration. Sci Rep 2025; 15:18601. [PMID: 40436951 PMCID: PMC12119898 DOI: 10.1038/s41598-025-00507-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 04/29/2025] [Indexed: 06/01/2025] Open
Abstract
3D Bioprinting technology has been applied to vaginal reconstruction with satisfactory results. Understanding the transcriptome and proteome of regenerated vaginas is essential for knowing how biomaterials and seed cells contribute to vaginal regeneration. There are no reports on the systemic analysis of vaginal regeneration transcriptomes or proteomes. This study aims to explore the transcriptomic and proteomic features of vaginal tissue reconstructed with 3D bioprinted scaffolds. The scaffolds were made with biomaterials and bone marrow-derived mesenchymal stem cells (BMSCs) and then transplanted into a rabbit model.rna sequencing was used to analyze the transcriptomes of reconstructed and normal vaginal tissues, identifying 11,956 differentially expressed genes (DEGs). Proteomic analysis using liquid chromatography-tandem mass spectrometry (LC-MS/MS) and data-independent acquisition (DIA) identified 7,363 differentially expressed proteins (DEPs). Gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) enrichment analyses were performed on DEGs and deps. Results showed that DEGs and deps.were involved in extracellular matrix remodeling, angiogenesis, inflammatory response, epithelialization, and muscle formation. This study shows that 3D bioprinted scaffolds are feasible for vaginal reconstruction and offers new insights into the molecular mechanisms involved.
Collapse
Affiliation(s)
- Xuemei Zhang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei Province, China
- Department of Pelvic floor clinic, Cangzhou Central Hospital, Cangzhou, 061600, Hebei Province, China
| | - Jiahua Zheng
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei Province, China
| | - Liye Zhang
- Chengde Medical University, Chengde, 067000, Hebei Province, China
| | - Jingkun Zhang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei Province, China
| | - Li Feng
- Department of Gynecology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, 050000, Hebei Province, China
| | - Lin Zhang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei Province, China.
| | - Xianghua Huang
- Department of Obstetrics and Gynecology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, Hebei Province, China.
| |
Collapse
|
12
|
Lee JJ, Ng KY, Bakhtiar A. Extracellular matrix: unlocking new avenues in cancer treatment. Biomark Res 2025; 13:78. [PMID: 40426238 PMCID: PMC12117852 DOI: 10.1186/s40364-025-00757-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/05/2025] [Indexed: 05/29/2025] Open
Abstract
The extracellular matrix (ECM) plays a critical role in cancer progression by influencing tumor growth, invasion, and metastasis. This review explores the emerging therapeutic strategies that target the ECM as a novel approach in cancer treatment. By disrupting the structural and biochemical interactions within the tumor microenvironment, ECM-targeted therapies aim to inhibit cancer progression and overcome therapeutic resistance. We examine the current state of ECM research, focusing on key components such as collagen, laminin, fibronectin, periostin, and hyaluronic acid, and their roles in tumor biology. Additionally, we discuss the challenges associated with ECM-targeted therapies, including drug delivery, specificity, and potential side effects, while highlighting recent advancements and future directions. This review underscores the potential of ECM-focused strategies to enhance the efficacy of existing treatments and contribute to more effective cancer therapies.
Collapse
Affiliation(s)
- Jia Jing Lee
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| | - Athirah Bakhtiar
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia.
| |
Collapse
|
13
|
Liu Z, Liao X, Zhao H, Ruan B, Jia F, He X, Long R. miR-29a-3p compositely regulates the COL6A6/PTEN-PI3K/Akt/CUX1 feedback loop to participate in the proliferation and invasion of pituitary adenomas. J Mol Histol 2025; 56:172. [PMID: 40419838 DOI: 10.1007/s10735-025-10436-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/21/2025] [Indexed: 05/28/2025]
Abstract
Pituitary adenoma (PA) is one of the most common intracranial tumors, and owing to its special biological morphology and behavior, there is currently no effective treatment. miRNAs play crucial roles as diagnostic indicators and targets for the treatment of numerous cancer types. The objective of this research was to explore how miR-29a-3p influences the development of PA. We collected 25 pairs of PA tissue and normal pituitary tissue, followed by the subcutaneous injection of 5 × 107 HP75 cells into the left axilla of nude mice, creating a heterotopic PA xenograft tumor model for experimental study. TtT/GF and HP75 cell proliferation and tumor growth in nude mice were assessed using CCK-8, Transwell, and immunohistochemistry tests. Western blotting, RT‒qPCR and RIP were used to detect the expression and interaction of related proteins and genes. The expression of miR-29a-3p was upregulated in PA. Knockdown of miR-29a-3p can inhibit the proliferation, invasion and migration of TtT/GF and HP75 cells and reduce the epithelial mesenchymal transformation (EMT) of these cells. Furthermore, reducing miR-29a-3p levels suppressed the expression of Ki-67 in the PA tissues of nude mice and slowed tumor growth. From a mechanistic standpoint, miR-29a-3p can target COL6A6 and PTEN. Knockdown of miR-29a-3p inhibits the PI3K/Akt/CUX1 signaling pathway through simultaneously increasing COL6A6 and PTEN expression, thus inhibiting the proliferation, invasion, migration and EMT of PA cells and alleviating the progression of PA. Conversely, CUX1 can promote the expression of miR-29a-3p through a positive feedback loop and accelerate the development of PA. Our study suggests that downregulating the expression of miR-29a-3p may be a new target for the treatment of PA.
Collapse
Affiliation(s)
- Zhuohui Liu
- Department of Otolaryngology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Xiufu Liao
- Department of Otolaryngology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
- Department of Otolaryngology, Chongqing General Hospital, Chongqing, 401147, Chongqing, China
| | - Hexiang Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Biao Ruan
- Department of Otolaryngology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Fengfeng Jia
- Department of Otolaryngology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Xuzhi He
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, 400042, Chongqing, China.
| | - Ruiqing Long
- Department of Otolaryngology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China.
| |
Collapse
|
14
|
Qi J, Luo Z, Li CY, Wang J, Ding W. Interpretable niche-based cell‒cell communication inference using multi-view graph neural networks. NATURE COMPUTATIONAL SCIENCE 2025:10.1038/s43588-025-00809-6. [PMID: 40425827 DOI: 10.1038/s43588-025-00809-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 04/22/2025] [Indexed: 05/29/2025]
Abstract
Cell‒cell communication (CCC) is a fundamental biological process for the harmonious functioning of biological systems. Increasing evidence indicates that cells of the same type or cluster may exhibit different interaction patterns under varying niches, yet most prevailing methods perform CCC inference at the cell type or cluster level while disregarding niche heterogeneity. Here we introduce the Spatial Transcriptomics-based cell‒cell Communication And Subtype Exploration (STCase) tool, which can describe CCC events at the single-cell/spot level based on spatial transcriptomics (ST). STCase includes an interpretable multi-view graph neural network via CCC-aware attention to identify niches for each cell type and uncover niche-specific CCC events. We show that STCase outperforms state-of-the-art approaches and accurately captures reported immune-related CCC events in human bronchial glands. We also identify three distinct niches of oral squamous cell carcinoma that may be obscured by agglomerative methods, and discover niche-specific CCC events that could influence tumor prognosis.
Collapse
Affiliation(s)
- Juntian Qi
- State Key Laboratory of Gene Function and Modulation Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhengchao Luo
- Department of Big Data and Biomedical AI, College of Future Technology, Peking University, Beijing, China
| | - Chuan-Yun Li
- State Key Laboratory of Gene Function and Modulation Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China.
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Jinzhuo Wang
- Department of Big Data and Biomedical AI, College of Future Technology, Peking University, Beijing, China.
| | - Wanqiu Ding
- State Key Laboratory of Gene Function and Modulation Research, Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China.
- Bioinformatics Core Facility, College of Future Technology, Peking University, Beijing, China.
| |
Collapse
|
15
|
Zhang Z, Ren J, Tang K, Hu X, Liu J, Li C. Matrix stiffness enhances viability, migration, invasion and invadopodia formation of oral cancer cells via PI3K/AKT pathway in vitro. Eur J Med Res 2025; 30:413. [PMID: 40410903 PMCID: PMC12103043 DOI: 10.1186/s40001-025-02666-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Accepted: 05/05/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND Oral cancer (OC) is one of the major types of cancer and the most common cause of cancer-related mortality in Asia. In recent years, matrix stiffness in the tumor microenvironment has been found to play an important role in regulating tumor cell behavior. However, the regulatory mechanisms associated with matrix stiffness in OC cells remain unclear. METHODS In this study, polyacrylamide gels with different stiffness were prepared to simulate low versus high matrix stiffness environments in tumor tissues by adjusting the acrylamide and cross-linker concentrations. Subsequently, the effects of different stiffness on OC cell survival, migration, invasion and invadopodia formation were explored based on cell counting kit-8 (CCK-8), Transwell and confocal microscopy. Meanwhile, the levels of markers relevant to phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT), apoptosis (BAX and BCL2) as well as metastasis (Cadherin-1, CDH1; Cadherin-2, and CDH2) were calculated via western blotting and real-time quantitative PCR. RESULTS According to the results, high matrix stiffness was seen to contribute to the increased number of migrated and invaded cells as well as the enhanced viability of OC cells, along with the aggravated invadopodia formation and the up-regulation in CDH2 and BCL2 levels yet the down-regulation in CDH1 and BAX levels. Elevated PI3K/AKT phosphorylation levels were also seen in high matrix stiffness-mediated OC cells, and the intervention using LY294002 could visibly overturned the effects of high matrix stiffness on the cell migration, invasion and invadopodia formation of OC cells. CONCLUSIONS This study reveals that matrix stiffness may enhance the invasiveness and anti-apoptotic ability of OC cells by activating the PI3K/AKT pathway, which provides a new idea for exploring the microenvironmental regulation of tumor mechanics and targeted intervention strategies.
Collapse
Affiliation(s)
- Zihao Zhang
- Department of Oral and Maxillofacial Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Jiayi Ren
- Department of Clinical Medicine, Harbin Medical University ("5+3" Integration), Harbin, 150000, China
| | - Ke Tang
- College of Pharmacy, Harbin Medical University, Harbin, 150000, China
| | - Xinyi Hu
- Department of General Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Jinhui Liu
- Department of Prosthodontics, Dalian Stomatological Hospital, Dalian, 151600, China
| | - Chunming Li
- Department of Oral and Maxillofacial Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China.
| |
Collapse
|
16
|
Wang C, Fan M, Heo SC, Adams SM, Li T, Liu Y, Li Q, Loebel C, Burdick JA, Lu XL, Birk DE, Alisafaei F, Mauck RL, Han L. Structure, Mechanics, and Mechanobiology of Fibrocartilage Pericellular Matrix Mediated by Type V Collagen. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e14750. [PMID: 40407177 DOI: 10.1002/advs.202414750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 05/01/2025] [Indexed: 05/28/2025]
Abstract
The pericellular matrix (PCM) is the immediate microniche surrounding cells in various tissues, regulating matrix turnover, cell-matrix interactions, and disease. This study elucidates the structure-mechanical properties and mechanobiology of the PCM in fibrocartilage, using the murine meniscus as the model. The fibrocartilage PCM is comprised of thin, randomly oriented collagen fibrils that entrap proteoglycans, contrasting with the densely packed, highly aligned collagen fibers in the bulk extracellular matrix (ECM). Compared to the ECM, the PCM exhibits lower modulus and greater isotropy, but has similar relative viscoelastic properties. In Col5a1+/- menisci, the reduction of collagen V results in thicker, more heterogeneous collagen fibrils, reduced modulus, loss of isotropy and faster viscoelastic relaxation in the PCM. Such altered PCM leads to impaired matrix-to-cell strain transmission, and in turn, disrupts mechanotransduction of meniscal cells, as illustrated by reduced calcium signaling activities and alters expression of matrix genes. In vitro, Col5a1+/- cells produce a weakened PCM with inferior properties and reduced protection of cells against tensile stretch. These findings highlight the PCM as a distinctive microstructure in fibrocartilage mechanobiology, underscoring a pivotal role of collagen V in PCM function. Targeting the PCM or its constituents offers potential for improving meniscus regeneration, osteoarthritis intervention and broader fibrocartilage-related therapies.
Collapse
Affiliation(s)
- Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Mingyue Fan
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Su Chin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sheila M Adams
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Thomas Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Yuchen Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Qing Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Claudia Loebel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jason A Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, 80309, USA
| | - X Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE, 19716, USA
| | - David E Birk
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Farid Alisafaei
- Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA, 19104, USA
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| |
Collapse
|
17
|
Zhou M, Wu W, Wang Y, Zhang B, Zhao X, Zhou H, Cao Y, Wu P, Wang M, Wang J. Targeting COL5A1 enhances anoikis thus attenuating malignancy of glioblastoma via inhibiting the Wnt/β-catenin signaling pathway. J Neurooncol 2025:10.1007/s11060-025-05036-7. [PMID: 40402199 DOI: 10.1007/s11060-025-05036-7] [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: 02/25/2025] [Accepted: 04/03/2025] [Indexed: 05/23/2025]
Abstract
PURPOSE As one of the most prevalent primary brain tumors, glioblastoma (GBM) is characterized by its severe malignancy and extremely poor prognosis. Recent studies have demonstrated that targeting anoikis and malignancy showed impressed efficiency for treatment in a wide range of solid tumors, however, relevant research on GBM still remains unclarified. METHODS In this study, genes related with malignancy and anoikis of GBM were identified by utilizing the Cancer Genome Atlas (TCGA), the Chinese Glioma Genome Atlas (CGGA) and the Molecular Signatures Database (MSigDB). Subsequently, the role of the key gene was validated via proliferation, invasion and migration experiments both in conditions with and without attachment. Moreover, RNA sequencing analysis was employed to reveal further mechanisms. RESULTS Here, Type V collagen alpha 1 (COL5A1) was identified as a critical gene associated with anoikis and poor outcomes. Additionally, COL5A1 knockdown induced significant reduction in malignancy of GBM both in vitro and in vivo. Moreover, cell anoikis was remarkable enhanced by reduced expression of COL5A1 after low-attachment cell culture. Mechanically, RNA sequencing analysis revealed that the activity of the Wnt/β-catenin signaling pathway was diminished following COL5A1 knockdown, which indicated that COL5A1 reduced anoikis via regulating Wnt/β-catenin signaling pathway thus promoted malignancies of GBM cells. CONCLUSION These findings demonstrated the novel evidence that COL5A1 serves as an essential regulatory factor influencing both anoikis and malignancy of GBM cells by regulating Wnt/β-catenin signaling pathway, indicating that COL5A1 could be a novel prognosis-related biomarker and potential therapeutic target for GBM.
Collapse
Affiliation(s)
- Mingjing Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta West Road, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Wei Wu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta West Road, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yichang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta West Road, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Beichen Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta West Road, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xuyan Zhao
- Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Haoyu Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta West Road, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yiyang Cao
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta West Road, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Pancheng Wu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta West Road, Xi'an, Shaanxi, 710061, China
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Maode Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta West Road, Xi'an, Shaanxi, 710061, China.
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Jia Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277, Yanta West Road, Xi'an, Shaanxi, 710061, China.
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
18
|
Xu DM, Chen LX, Xue T, Zhuang XY, Wei LC, Han H, Mo M. Decoding the impact of MMP1+ malignant subsets on tumor-immune interactions: insights from single-cell and spatial transcriptomics. Cell Death Discov 2025; 11:244. [PMID: 40394037 DOI: 10.1038/s41420-025-02503-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/22/2025] Open
Abstract
Matrix metalloproteinase 1 plays a pivotal role in tumor biology and immune modulation through its enzymatic remodeling of the extracellular matrix, facilitating tumor progression. In this study, we utilized large-scale single-cell RNA sequencing and spatial transcriptomics to investigate MMP1 expression, its cellular localization, and its impact on tumor progression and immune modulation. Our findings reveal that MMP1 expression is elevated in various tumor types and is strongly correlated with metastatic potential. High MMP1 expression was associated with increased activity in epithelial-mesenchymal transition signaling and TNFα/NF-κB pathways, which are known to promote tumor progression. Furthermore, MMP1+ malignant cells exhibited significant interactions with immune cells, particularly macrophages and CD8+ T cells. MMP1 expression correlated with enhanced macrophage infiltration and impaired CD8+ T-cell function, contributing to an immunosuppressive tumor microenvironment. Notably, the CXCL16-CXCR6 and ANXA1-FPR3 signaling axes were identified as key mediators of these interactions. Inhibition of MMP1 in vitro demonstrated reduced cell invasion, stemness, and proliferation, while increasing reactive oxygen species levels and promoting apoptosis. Our findings position MMP1 as a key player in the "tumor-immune" vicious cycle and a promising therapeutic target to enhance anti-tumor responses and improve patient outcomes.
Collapse
Affiliation(s)
- Da-Ming Xu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Ling-Xiao Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Ting Xue
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Xiao-Yu Zhuang
- Department of Anesthesiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, P. R. China
| | - Li-Chao Wei
- Department of Organ Transplantation, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Hui Han
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China.
| | - Miao Mo
- Department of Urology, Xiangya Hospital, Central South University, Changsha, P.R. China.
| |
Collapse
|
19
|
Zeng C, Lv X, Wang F, Huang Y, Ren Y, Zhang H. Matrix Remodeling Associated Genes (MXRAs): structural diversity, functional significance, and therapeutic potential in tumor microenvironments. Discov Oncol 2025; 16:833. [PMID: 40394417 PMCID: PMC12092922 DOI: 10.1007/s12672-025-02728-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 05/16/2025] [Indexed: 05/22/2025] Open
Abstract
The Matrix Remodeling Associated Genes (MXRAs) family, comprising eight distinct members (MXRA1-8), plays a crucial role in the development and maintenance of higher vertebrate cells. These proteins are primarily involved in the regulation of intercellular adhesion and the remodeling of the extracellular matrix (ECM). Recent investigations have highlighted the significant roles of MXRAs in the modulation of tumor growth and progression, establishing them as vital components in the oncogenic landscape. Notably, each MXRA member exhibits unique structural characteristics and functional properties, contributing to a diverse array of regulatory effects within the tumor context. This review seeks to provide a comprehensive analysis of the structural attributes, functional contributions, and activities of MXRAs within the tumor microenvironment. By elucidating the underlying mechanisms of action, this paper aims to offer novel insights and strategic approaches for the identification of early diagnostic biomarkers, as well as potential therapeutic targets that may facilitate molecular interventions aimed at inhibiting tumor development.
Collapse
Affiliation(s)
- Chao Zeng
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Xiao Lv
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
- Department of Obstetrics and Gynecology, Key Laboratory of Gynecologic Oncology Gansu Province, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Feng Wang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China
- The First School of Clinical Medicne, Lanzhou University, Lanzhou, 730030, China
| | - Yaomin Huang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China
- The First School of Clinical Medicne, Lanzhou University, Lanzhou, 730030, China
| | - Yanxian Ren
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China
- The First School of Clinical Medicne, Lanzhou University, Lanzhou, 730030, China
- Gansu Province Key Laboratory of Biological Therapy and Regenerative Medicine Transformation, Lanzhou, 730030, China
| | - Hengwei Zhang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
- The First School of Clinical Medicne, Lanzhou University, Lanzhou, 730030, China.
- Gansu Province Key Laboratory of Biological Therapy and Regenerative Medicine Transformation, Lanzhou, 730030, China.
| |
Collapse
|
20
|
Bai L, Li J, Du S, Lei W, Zhou F, Chen Y, Si Y, Wang Y, Li L, Li Y. Lingguizhugan decoction inhibits the cleavage of LYVE-1 by MMP-9 and promotes lymphangiogenesis to improve myocardial infarction. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156863. [PMID: 40412056 DOI: 10.1016/j.phymed.2025.156863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 04/30/2025] [Accepted: 05/13/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Myocardial remodeling plays a crucial role in determining patient outcomes after myocardial infarction (MI). Emerging evidence from both preclinical and clinical studies highlights the beneficial effects of cardiac lymphangiogenesis in improving cardiac function and prognosis post-MI. Lingguizhugan decoction (LD), a traditional Chinese medicine, is extensively used in the treatment of ischemic heart disease. Nevertheless, its potential mechanisms are still not well understood. PURPOSE To determine whether LD can enhance post-MI myocardial remodeling by promoting lymphangiogenesis and to elucidate its molecular mechanisms METHODS: Surgical ligation of the left anterior descending artery (LAD) was utilized to establish MI rat model. Cardiac structure and function were assessed using histopathological staining and echocardiography. A transgenic zebrafish model was used to confirm that lymphangiogenesis plays a key role in LD's cardioprotective effects. Network pharmacology analysis was conducted to predict the potential mechanisms underlying LD's therapeutic action in MI. The expression levels of matrix metalloproteinase-9 (MMP-9) and lymphatic vessel endothelial receptor-1 (LYVE-1) were assessed in MI rats. Transcriptomic data mining from MI patients and in vitro protein interaction validation were conducted to explore the relationship between MMP-9 and LYVE-1. Key proteins involved in the interleukin-17 (IL-17) signaling pathway were analyzed using western blotting and qRT-PCR. Furthermore, mass spectrometry imaging was conducted to identify potential bioactive compounds in LD that regulate the IL-17 signaling pathway. RESULTS LD significantly improved cardiac function and mitigated adverse myocardial remodeling in left anterior descending artery-ligated rats. Notably, LD promoted cardiac lymphangiogenesis and improved cardiac function in transgenic zebrafish treated with verapamil, with further validation using lymphangiogenesis inhibitors. Based on network pharmacology findings and previous studies, MMP-9 and LYVE-1 were identified as key targets of LD. LD decreased MMP-9 expression and increased LYVE-1 levels in MI rat hearts. A strong correlation was observed between MMP-9 and LYVE-1, suggesting a potential regulatory relationship. Additionally, LD downregulated key proteins involved in the IL-17 signaling pathway, indicating its role in modulating inflammatory responses. Finally, four biologically active compounds dehydrotrametenonic acid, ethyl p-methoxycinnamate, atractylon, and licoricone were identified in cardiac tissue as potential regulators of the IL-17R/MMP-9/LYVE-1 axis, contributing to LD's lymphangiogenesis-promoting effects. CONCLUSIONS LD enhances post-MI ventricular remodeling by promoting lymphangiogenesis and modulating the IL-17R/MMP-9/LYVE-1 signaling pathway. Dehydrotrametenonic acid, ethyl p-methoxycinnamate, atractylon, and licoricone may serve as key bioactive compounds responsible for LD's therapeutic effects.
Collapse
Affiliation(s)
- Liding Bai
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiaxin Li
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Siqi Du
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Wei Lei
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Fengjie Zhou
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yao Chen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China
| | - Yuxue Si
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Yanyan Wang
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China.
| | - Lin Li
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yuhong Li
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
21
|
Guvatova ZG, Kudasheva ER, Efremov YM, Timashev PS, Fedorova MS, Pudova EA, Snezhkina AV, Kudryavtseva AV, Kobelyatskaya AA, Moskalev AA. Changes in Gene Expression Patterns in Young and Senescent Fibroblasts in Glycated Three-Dimensional Collagen Matrices. Int J Mol Sci 2025; 26:4769. [PMID: 40429909 PMCID: PMC12112436 DOI: 10.3390/ijms26104769] [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: 04/20/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Glycation, or non-enzymatic glycosylation, has recently attracted increasing interest in the context of its impact on aging. Advanced glycation end products (AGEs) contribute to various age-related pathological conditions such as inflammation, fibrosis, and vascular calcification. However, the molecular mechanisms underlying glycation-induced disruption of cell-matrix interactions during cellular senescence are not fully understood. The aim of this study was to investigate transcriptomic changes in young and senescent dermal fibroblasts (HdFbs) cultured in 3D post-glycated collagen type I matrices after 10 and 17 days. Our findings indicate that D-ribose-mediated glycation increases the accumulation of fluorescent AGEs and the stiffness of matrices in a dose-dependent manner. The transcriptome alterations in cells encompassed the modulation of age-related genes and signaling pathways, including activation of genes related to senescence-associated secretory phenotype (SASP). Notably, the alterations in the transcriptome profiles due to glycation were more pronounced (in terms of both the number of genes and their fold changes) after 10 days of culture compared to day 17 in both passages. These findings suggest that cellular responses to glycation and resulting stiffness depend on both the concentration of reducing sugar and the time spent under those conditions.
Collapse
Affiliation(s)
- Zulfiya G. Guvatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Evelina R. Kudasheva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Russian Clinical Research Center for Gerontology, Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, 127994 Moscow, Russia
| | - Yuri M. Efremov
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Peter S. Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Maria S. Fedorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Elena A. Pudova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasiya V. Snezhkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anna V. Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasiya A. Kobelyatskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Longevity Institute of Russian Research Surgery Center, 119435 Moscow, Russia
| | - Alexey A. Moskalev
- Longevity Institute of Russian Research Surgery Center, 119435 Moscow, Russia
| |
Collapse
|
22
|
Zhang XH, He YD, Wang H, Cao Y, Ying SQ, Liu JN, Lei X, Liu L, Cai XY, Mu SH, Zhang KC, Yuan Y, Liu YH, Xu HK, Chen J, Liang JF, Jin Y, Jin F, Sui BD, Zheng CX. Development-Inspired Biomimetic Cell-Niche Coaggregates Safeguard Tooth Stem Cell-Based Functional Tissue Regeneration. Adv Healthc Mater 2025:e2501550. [PMID: 40376872 DOI: 10.1002/adhm.202501550] [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: 03/24/2025] [Indexed: 05/18/2025]
Abstract
Harnessing natural developmental programs to repair and replace damaged organs represents promising approaches in regenerative medicine. However, effective strategies are still lacking for tissue regeneration in complicated conditions, such as the periodontal bone defect. Here, human dental follicle stem cells (hDFSCs) and their aggregates (hDFSCA) are cultured and characterized, which are formed based on the inherent property of these stem cells self-assembly into compact spheroid-like structures, mimicking mesenchymal condensation in development. A periodontal tissue-specific microenvironment simulation material is then established, human decellularized alveolar bone matrix particles (hDABMPs), which possess favorable physicochemical and biological properties for regenerative use. hDFSCs co-cultured with hDABMPs exhibit improved cell function, and hDFSCA-hDABMP co-aggregates are subsequently constructed, which activate the developmental gene expression in hDFSCA and initiate hypoxic adaptation mechanisms for tissue regeneration. Indeed, hDFSCA-hDABMP co-aggregates significantly promote regeneration after implantation in alveolar bone defects with good biosafety. Interestingly, during the early stages of implantation, hDABMPs enhance hDFSC survival and expansion, thereby providing a sufficient source of cells for tissue regeneration. Collectively, this study reveals a development-inspired, engineered cell-niche co-aggregation strategy for enhancing CA therapeutic potential by simulating tissue-specific microenvironments, offering novel insights for functional tissue regeneration.
Collapse
Affiliation(s)
- Xiao-Hui Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yi-De He
- Department of Operative Dentistry and Endodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Hao Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yuan Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Si-Qi Ying
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jia-Ning Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xiao Lei
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Lu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xin-Yue Cai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Shi-Han Mu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Kai-Chao Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yuan Yuan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yi-Han Liu
- Department of Stomatology, the First Medical Center, Chinese PLA General Hospital, Beijing, Beijing, 100039, China
| | - Hao-Kun Xu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Ji Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Oral Implantology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jian-Fei Liang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Yan Jin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Fang Jin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Bing-Dong Sui
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Chen-Xi Zheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| |
Collapse
|
23
|
Liu Y, Gilchrist AE, Johansson PK, Guan Y, Deras JD, Liu YC, Ceva S, Huang MS, Navarro RS, Enejder A, Peltz G, Heilshorn SC. Engineered Hydrogels for Organoid Models of Human Nonalcoholic Fatty Liver Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e17332. [PMID: 40364726 DOI: 10.1002/advs.202417332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/22/2025] [Indexed: 05/15/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by increased lipid accumulation and excessive deposition of extracellular matrix (ECM) that results in tissue stiffening. The potential interplay between matrix stiffness and hepatocyte lipid accumulation during NAFLD has not been established. Here, an in vitro NAFLD model is developed using chemically defined, engineered hydrogels and human induced pluripotent stem cell-derived hepatic organoids (HOs). Specifically, dynamic covalent chemistry crosslinking, along with transient small molecule competitors, are used to create dynamic stiffening hydrogels that enable the reproducible culture of HOs. Within matrices that mimic the stiffness of healthy to diseased tissue (≈1-6 kPa), lipid droplet accumulation in HOs is triggered by exposure to an NAFLD-associated free fatty acid. These NAFLD model suggests that higher stiffness microenvironments result in increased hepatic lipid droplet accumulation, increased expression of fibrosis markers, and increased metabolic dysregulation. By targeting the ROCK mechanosignaling pathway, the synergy between matrix stiffness and lipid droplet accumulation is disrupted. The in vitro model of NAFLD has the potential to understand the role of mechanosignaling in disease progression and identify new pathways for therapeutic intervention.
Collapse
Affiliation(s)
- Yueming Liu
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Aidan E Gilchrist
- Department of Biomedical Engineering, University of California, Davis, CA, 95616, USA
| | - Patrik K Johansson
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Yuan Guan
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jaydon D Deras
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Yu-Chung Liu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sofia Ceva
- Department of Biology, Stanford University, Stanford, CA, 94305, USA
| | - Michelle S Huang
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Renato S Navarro
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Annika Enejder
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Gary Peltz
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
24
|
Joshi J, Bhutada S, Martin DR, Guzowski J, Blankenberg D, Apte SS. DICED (Database of Identified Cleavage Sites Endemic to Diseases States): A Searchable Web Interface for Terminomics/Degradomics. Proteomics 2025:e202500007. [PMID: 40351053 DOI: 10.1002/pmic.202500007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 04/09/2025] [Accepted: 04/25/2025] [Indexed: 05/14/2025]
Abstract
Proteolysis is an irreversible posttranslational modification with immense biological impact. Owing to its high disease significance, there is growing interest in investigating proteolysis on the proteome scale, termed degradomics. We developed 'Database of Identified Cleavage sites Endemic to Disease states' (DICED; https://diced.lerner.ccf.org/), as a searchable knowledgebase to promote collaboration and knowledge sharing in degradomics. DICED was designed and constructed using Python, JavaScript, HTML, and PostgreSQL. Django (https://www.djangoproject.com) was chosen as the primary framework for its security features and support for agile development. DICED can be utilized on major web browsers and operating systems for easy access to high-throughput mass spectrometry-identified cleaved protein termini. The data was obtained using N-terminomics, comprising N-terminal protein labeling, labeled peptide enrichment, mass spectrometry and positional peptide annotation. The DICED database contains experimentally derived N-terminomics peptide datasets from tissues, diseases, or digests of tissue protein libraries using individual proteases and is searchable using UniProt ID, protein name, gene symbol or up to 100 peptide sequences. The tabular output format can be exported as a CSV file. Although DICED presently accesses data from a single laboratory, it is freely available as a Galaxy tool and the underlying database is scalable, permitting addition of new datasets and features.
Collapse
Affiliation(s)
- Jayadev Joshi
- Center for Computational Life Sciences, Cleveland Clinic Research, Cleveland, Ohio, USA
| | - Sumit Bhutada
- Department of Biomedical Engineering, Cleveland Clinic Research, Cleveland, Ohio, USA
| | - Daniel R Martin
- Department of Biomedical Engineering, Cleveland Clinic Research, Cleveland, Ohio, USA
| | - Joyce Guzowski
- Central Administration, Cleveland Clinic Research, Cleveland, Ohio, USA
| | - Daniel Blankenberg
- Center for Computational Life Sciences, Cleveland Clinic Research, Cleveland, Ohio, USA
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland Clinic Research, Cleveland, Ohio, USA
| |
Collapse
|
25
|
Chissico Júnior F, Santos da Silva T, Vieira Meirelles F, Monzani PS, Fornari Laurindo L, Maria Barbalho S, Miglino MA. A Review on Bioengineering the Bovine Mammary Gland: The Role of the Extracellular Matrix and Reconstruction Prospects. Bioengineering (Basel) 2025; 12:501. [PMID: 40428120 PMCID: PMC12108683 DOI: 10.3390/bioengineering12050501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
The mammary gland is a modified sweat gland responsible for milk production. It is affected by diseases that reduce animals' quality of life, consequently leading to economic losses in livestock. With advancements in tissue bioengineering and regenerative medicine, studying the extracellular matrix (ECM) of the bovine mammary gland can improve our understanding of its physiology and the processes that affect it. This knowledge could also enable the development of sustainable therapeutic alternatives for both the dairy production chain and human oncology research. A common approach in regenerative medicine is decellularization, a process that removes all cells from tissue while preserving its architecture and ECM components for subsequent recellularization. The success of recellularization depends on obtaining immunologically compatible scaffolds and using appropriate cell culture sources and methods to ensure tissue functionality. However, tissue culture technology still faces challenges due to specific requirements and high costs. Here, we review the literature on biomaterials and tissue engineering, providing an overview of the ECM of the bovine mammary gland and advances in its bioengineering, with a focus on regenerative medicine for bovine species. The methodology employed consists of a structured search of scientific databases, including PubMed, Google Scholar, and SciELO, using specific keywords related to tissue engineering and the bovine mammary gland. The selection criteria prioritized peer-reviewed articles published between 2002 and 2025 that demonstrated scientific relevance and contributed to the understanding of bovine mammary gland bioengineering. Although research on this topic has advanced, vascularization, tissue maturation, and scalability remain key barriers to widespread application and economic viability.
Collapse
Affiliation(s)
- Fernando Chissico Júnior
- Department of Surgery, School of Veterinary Medicine and Animal Science, Universidade de São Paulo (USP), São Paulo 05508-270, SP, Brazil; (F.C.J.); (T.S.d.S.)
- Department of Veterinary Medicine and Animal Science, School of Veterinary Medicine and Animal Science, Universidade Save (UniSave), Chongoene 1200, Mozambique
| | - Thamires Santos da Silva
- Department of Surgery, School of Veterinary Medicine and Animal Science, Universidade de São Paulo (USP), São Paulo 05508-270, SP, Brazil; (F.C.J.); (T.S.d.S.)
| | - Flávio Vieira Meirelles
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, Universidade de São Paulo (USP), Pirassununga Campus, Pirassununga 13635-900, SP, Brazil;
| | - Paulo Sérgio Monzani
- Department of Food Engineering, Faculty of Animal Science and Food Engineering, Universidade de São Paulo (USP), Pirassununga Campus, Pirassununga 13635-900, SP, Brazil;
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil or (L.F.L.); (S.M.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil or (L.F.L.); (S.M.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Maria Angélica Miglino
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Postgraduate Program in Animal Health, Production and Environment, School of Veterinary Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Department of Animal Anatomy, School of Veterinary Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| |
Collapse
|
26
|
Miller J, Perrier Q, Rengaraj A, Bowlby J, Byers L, Peveri E, Jeong W, Ritchey T, Gambelli AM, Rossi A, Calafiore R, Tomei A, Orlando G, Asthana A. State of the Art of Bioengineering Approaches in Beta-Cell Replacement. CURRENT TRANSPLANTATION REPORTS 2025; 12:17. [PMID: 40342868 PMCID: PMC12055624 DOI: 10.1007/s40472-025-00470-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2025] [Indexed: 05/11/2025]
Abstract
Purpose of the Review Despite recent advancements in technology for the treatment of type 1 diabetes (T1D), exogenous insulin delivery through automated devices remains the gold standard for treatment. This review will explore progress made in pancreatic islet bioengineering within the field of beta-cell replacement for T1D treatment. Recent Findings First, we will focus on the use of decellularized extracellular matrices (dECM) as a platform for pancreatic organoid development. These matrices preserve microarchitecture and essential biochemical signals for cell differentiation, offering a promising alternative to synthetic matrices. Second, advancements in 3D bioprinting for creating complex organ structures like pancreatic islets will be discussed. This technology allows for increased precision and customization of cellular models, crucial for replicating native pancreatic islet functionality. Finally, this review will explore the use of stem cell-derived organoids to generate insulin-producing islet-like cells. While these organoids face challenges such as functional immaturity and poor vascularization, they represent a significant advancement for disease modeling, drug screening, and autologous islet transplantation. Summary These innovative approaches promise to revolutionize T1D treatment by overcoming the limitations of traditional therapies based on human pancreatic islets.
Collapse
Affiliation(s)
- Jake Miller
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | - Quentin Perrier
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
- Univ. Grenoble Alpes, Department of Pharmacy, Grenoble Alpes University Hospital, Grenoble, France
| | - Arunkumar Rengaraj
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| | - Joshua Bowlby
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| | - Lori Byers
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| | - Emma Peveri
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | - Wonwoo Jeong
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | - Thomas Ritchey
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | | | - Arianna Rossi
- Department of Engineering, University of Perugia, Perugia, Italy
| | | | - Alice Tomei
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL USA
| | - Giuseppe Orlando
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| | - Amish Asthana
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| |
Collapse
|
27
|
Dobrzyn K, Kopij G, Kiezun M, Zaobidna E, Gudelska M, Zarzecka B, Kisielewska K, Rak A, Smolinska N, Kaminski T. The effect of visfatin on the transcriptomic profile of porcine anterior pituitary cells during periimplantation period. Sci Rep 2025; 15:15858. [PMID: 40328795 PMCID: PMC12056127 DOI: 10.1038/s41598-025-00766-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 04/30/2025] [Indexed: 05/08/2025] Open
Abstract
Females' reproductive capacity is closely related to the actual metabolic status of the organism. The pituitary, an element of the regulatory hypothalamic-pituitary-ovarian axis, is one of the most important endocrine glands regulating reproductive system activity. Undisturbed functioning of pituitary ensures the regular course of pregnancy through, among others, the modulation of steroid hormones production, which is critical in the early stages of gestation. Visfatin, a hormone belonging to the adipokines family, may belong to a group of factors regulating the reproductive functions in response to the female's metabolic status. Herein we verified the hypothesis assuming a modulatory effect of visfatin on the porcine anterior pituitary transcriptome on days 15 to 16 of gestation (beginning of implantation). RNA-seq analysis of the porcine anterior pituitary cells revealed changes in the expression of 203 genes (121 up-regulated and 82 down-regulated, when compared to the non-treated controls), assigned to 325 gene ontology terms. The presence of visfatin affected the frequency of alternative splicing events (194 cases), as well as long noncoding RNA expression (64 cases). Visfatin expression and the occurrence of alternative splicing events of genes that are responsible, directly or indirectly, for regulation of the secretory functions of the pituitary, including those critical for reproductive functions suggests that the adipokine may be a key agent in ensuring the appropriate hormonal milieu during the peri-implantation period.
Collapse
Affiliation(s)
- Kamil Dobrzyn
- Department of Zoology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego st. 5, Olsztyn, 10-719, Poland.
| | - Grzegorz Kopij
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Marta Kiezun
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Ewa Zaobidna
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Marlena Gudelska
- Department of Human Histology and Embryology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Barbara Zarzecka
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Katarzyna Kisielewska
- Department of Human Histology and Embryology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Agnieszka Rak
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Nina Smolinska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Tadeusz Kaminski
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
28
|
Timmer LT, den Hertog E, Versteeg D, Post H, Verdonschot JAJ, Monshouwer-Kloots J, Kyriakopoulou E, Perini I, Koopmans T, van der Kraak P, Zentilin L, Heymans SRB, Vink A, Giacca M, Heck AJR, van Rooij E. Cardiomyocyte SORBS2 expression increases in heart failure and regulates integrin interactions and extracellular matrix composition. Cardiovasc Res 2025; 121:585-600. [PMID: 39957251 PMCID: PMC12054630 DOI: 10.1093/cvr/cvaf021] [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: 04/25/2024] [Revised: 10/26/2024] [Accepted: 12/05/2024] [Indexed: 02/18/2025] Open
Abstract
AIMS In this study, we aimed to uncover genes associated with stressed cardiomyocytes by combining single-cell transcriptomic data sets from failing cardiac tissue from both humans and mice. METHODS AND RESULTS Our bioinformatic analysis identified SORBS2 as conserved NPPA-correlated gene. Using mouse models and cardiac tissue from human heart failure patients, we demonstrated that SORBS2 expression is consistently increased during pathological remodelling, correlates to disease severity, and is regulated by GATA4. By affinity purification mass spectrometry, we showed SORBS2 to interact with the integrin-cytoskeleton connections. Cardiomyocyte-specific genetic loss of Sorbs2 in adult mice changed integrin interactions, indicated by the increased expression of several integrins and altered extracellular matrix components connecting to these integrins, leading to an exacerbated fibrotic response during pathological remodelling. CONCLUSION Sorbs2 is a cardiomyocyte-enriched gene that is increased during progression to heart failure in a GATA4-dependent manner and correlates to phenotypical hallmarks of cardiac failure. Our data indicate SORBS2 to function as a crucial regulator of integrin interactions and cardiac fibrosis.
Collapse
Affiliation(s)
- Louk T Timmer
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Elvira den Hertog
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Danielle Versteeg
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
- Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Harm Post
- Biomolecular Mass Spectrometry and Proteomics Group, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Netherlands Proteomics Center, Utrecht, The Netherlands
| | - Job A J Verdonschot
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Cardiology, Cardiovascular Research Institute (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
- European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart)
| | - Jantine Monshouwer-Kloots
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Eirini Kyriakopoulou
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Ilaria Perini
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Tim Koopmans
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Petra van der Kraak
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lorena Zentilin
- Molecular Medicine Laboratory, International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Stephane R B Heymans
- Department of Cardiology, Cardiovascular Research Institute (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Aryan Vink
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mauro Giacca
- Molecular Medicine Laboratory, International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- School of Cardiovascular and Metabolic Medicine & Sciences, British Heart Foundation Centre of Research Excellence, King’s College London, London, UK
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics Group, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Netherlands Proteomics Center, Utrecht, The Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
- Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
29
|
Zhao W, Zhang J, Guo Q, Wang Q, Zhao H, Xiao F, Han M, Cao Y, Ding R, Yang A, Xie W. Fibulin-1 deficiency alleviates liver fibrosis by inhibiting hepatic stellate cell activation via the p38 MAPK pathway. Cell Mol Life Sci 2025; 82:192. [PMID: 40323446 PMCID: PMC12052672 DOI: 10.1007/s00018-025-05647-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/16/2025] [Accepted: 03/04/2025] [Indexed: 05/08/2025]
Abstract
Elastin stabilization has been correlated with the reversibility of fibrosis. Fibulin-1 can participate in elastin assembly, which promotes its stabilization. However, the role of Fibulin-1 in liver fibrosis remains unknown. Here, we performed a proteomics analysis to identify notable changes in Fibulin-1 expression during continuous fibrosis progression and regression. Fibulin-1 expression was dramatically increased in the plasma of patients with cirrhosis as well as in liver fibrosis models and hepatic stellate cells (HSCs) treated with TGF-β1, and significant accumulation of Fibulin-1 was observed in chronic hepatitis B (CHB)- and metabolic dysfunction-associated steatohepatitis (MASH)-related cirrhosis. Functional studies demonstrated that Fibulin-1 silencing inhibited HSC activation, while the opposite effects were observed for Fibulin-1 overexpression in vitro. Furthermore, transcriptomic analysis revealed that Fibulin-1 mediated p38 MAPK pathway activation, which was confirmed by the addition of a p38 MAPK inhibitor. More importantly, Fibulin-1 depletion in a CCl4-induced liver fibrosis model substantially ameliorated fibrosis progression, which was accompanied by decreased profibrogenic gene expression and decreased levels of insoluble elastin. Moreover, activation of the p38 MAPK pathway was inhibited in vivo. The expression of Fibulin-1D, rather than Fibulin-1C, was elevated during liver fibrogenesis, which suggested a major role for Fibulin-1D in liver fibrosis. Next, we established Fibulin-1D/elastin-coated culture models with LX-2 cells. LX-2 cells with extracellular elastin and Fibulin-1D deposition showed more significant profibrotic phenotypic alterations than those with elastin alone. Fibulin-1 deficiency alleviated liver fibrosis by reducing insoluble elastin and HSC activation, and finally, the p38 MAPK pathway might be involved in the effect of Fibulin-1 on HSCs.
Collapse
Affiliation(s)
- Wenshan Zhao
- Center of Liver Diseases, National Medical Centre for Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People's Republic of China
| | - Jingyu Zhang
- Center of Liver Diseases, National Medical Centre for Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People's Republic of China
| | - Qingdong Guo
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, People's Republic of China
| | - Qi Wang
- Center of Liver Diseases, National Medical Centre for Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People's Republic of China
| | - Hong Zhao
- Center of Liver Diseases, National Medical Centre for Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People's Republic of China
| | - Fan Xiao
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People's Republic of China
| | - Ming Han
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People's Republic of China
| | - Ying Cao
- Center of Liver Diseases, National Medical Centre for Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People's Republic of China
| | - Rui Ding
- Center of Liver Diseases, National Medical Centre for Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People's Republic of China
| | - Aiting Yang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China.
| | - Wen Xie
- Center of Liver Diseases, National Medical Centre for Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People's Republic of China.
| |
Collapse
|
30
|
Liu Y, Li B, Zhang J, Zhao B, Chen L, Chen B. Temporal Proteome Profiling of Anterior Cruciate Ligament Tear Remnants: Secretory Proteins in the Acute Phase Potentially Promote Tissue Repair. J Proteome Res 2025; 24:2302-2313. [PMID: 40192091 DOI: 10.1021/acs.jproteome.4c00792] [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] [Indexed: 05/03/2025]
Abstract
Previous studies reported that preserving the anterior cruciate ligament (ACL) remnants following ACL rupture during reconstruction surgery could promote graft healing. However, the temporal proteomic expression of ACL remnants remains unclear. Based on previous reports, we have redefined the initial 6 weeks following ACL rupture as the acute phase and the subsequent 6 weeks to 6 months as the subacute phase. High-throughput proteomic sequencing on ACL remnants from the two groups was utilized. Our study unveiled a total of 381 differential expression proteins (DEPs), with 136 upregulated and 245 downregulated proteins in the acute phase. By intersecting these findings with secretory protein databases, we identified 26 upregulated secretory proteins and 19 downregulated in the acute phase. The upregulation of MMP9 and VTN and the downregulation of COL1A1 and POSTN in the acute phase were further confirmed by immunohistochemistry. These findings suggest that the elevated expression of secretory proteins in the acute phase may play crucial roles in promoting cell proliferation, angiogenesis, and tissue repair of the graft. This study not only enhances our understanding of repair mechanisms in ACL remnant preservation but also provides a theoretical foundation for guiding rational clinical surgical timing.
Collapse
Affiliation(s)
- Yiming Liu
- Division of Joint Surgery and Sports Medicine, Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430000, China
| | - Bin Li
- Division of Joint Surgery and Sports Medicine, Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430000, China
| | - Jun Zhang
- Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China
| | - Boming Zhao
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shannxi 710000, China
| | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430000, China
| | - Biao Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430000, China
| |
Collapse
|
31
|
Zhu L, Shi W, Tuoheti Y, Gong GJ, Chen M, Liang ZH, Abudureheman A, Gao WG. Long noncoding RNA LINC01811 sponges miR-214-3p and upregulates YAP1 thereby promoting the migration and invasion of colorectal cancer. 3 Biotech 2025; 15:123. [PMID: 40225417 PMCID: PMC11985869 DOI: 10.1007/s13205-025-04292-8] [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: 08/14/2024] [Accepted: 03/22/2025] [Indexed: 04/15/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) exert significant influence on the development of cancer. However, their role in colorectal cancer (CRC) is not fully clarified. The expression levels of LINC01811 in CRC samples were determined using differential expression analysis and validated by RT-qPCR assays. Transwell assays were conducted to investigate the biological function of LINC01811 in CRC. To elucidate the mechanism by which LINC01811 acts as a molecular sponge for miR-214-3p and regulates YAP1 expression, binding site analysis, Luciferase reporter assay, RT-qPCR, and Western blotting were employed. We identified a novel oncogenic lncRNA LINC01811 in CRC tissues and cell lines. Our results showed that the suppression of LINC01811 significantly reduced CRC cell invasion and migration by regulating epithelial-mesenchymal transition-related markers, including MMP2, MMP9, vimentin, and E-cadherin in vitro. Furthermore, LINCO1811 modulated YAP1 expression by sequestering miR-214-3p, thereby promoting CRC progression by suppressing its activity. In summary, this study identified a novel lncRNA LINC01811 involved in CRC progression through the miR-214-3p/YAP1 axis. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-025-04292-8.
Collapse
Affiliation(s)
- Li Zhu
- Department of Colorectal Surgery Ward, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Urumqi, 830000 China
| | - Wen Shi
- Department of Colorectal Surgery Ward, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Urumqi, 830000 China
| | - Yiminjiang Tuoheti
- Department of Colorectal Surgery Ward, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Urumqi, 830000 China
| | - Guo-jie Gong
- Department of Colorectal Surgery Ward, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Urumqi, 830000 China
| | - Min Chen
- Department of Colorectal Surgery Ward, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Urumqi, 830000 China
| | - Zong-hua Liang
- Department of Colorectal Surgery Ward, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Urumqi, 830000 China
| | - Abuduweili Abudureheman
- Department of Colorectal Surgery Ward, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Urumqi, 830000 China
| | - Wei-ge Gao
- Department of Colorectal Surgery Ward, People’s Hospital of Xinjiang Uygur Autonomous Region, No. 91 Tianchi Road, Urumqi, 830000 China
| |
Collapse
|
32
|
Long Q, Liu C, Zheng H, Wang M, Liu H, Liu Y, Cao Z, Sun Y, Mo Q, Backman LJ, Zhu J, Hu L, Huang J, Zhang W, Chen J. Enhancing Tendon Regeneration: Investigating the Impact of Topography on the Secretome of Adipose-Derived Stem Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417447. [PMID: 40091553 PMCID: PMC12079404 DOI: 10.1002/advs.202417447] [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] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Indexed: 03/19/2025]
Abstract
Tendons are vital for maintaining integrity and movement, but current treatment options are insufficient for their regeneration after injuries. Previous studies have shown that the secretome from mesenchymal stem cells (MSCs) promoted tendon regeneration. However, limited studies have explored the impact of the physical microenvironment on the secretome's efficacy of MSCs. In this study, it is shown that the topographic orientation regulates the secretome of human adipose-derived stem cells (ADSCs) and promotes tendon regeneration. Conditioned medium (CM) is collected from ADSCs cultured on the scaffolds with different topography. The results show that CM generated from aligned structure group has a potent effect in promoting cell migration and proliferation, tenogenic differentiation, macrophage polarization toward M2 phenotype, tendon structure and mechanical function recovery. Proteomic analysis revealed that the aligned structure can up-regulate the secretion of Extracellular matrix (ECM) proteins while down-regulate proinflammatory factors. This modulation activates the MAPK, GPCR and Integrin signaling pathways which may account for the enhanced effect on tendon regeneration. This study offers a promising and safer non-cell-based treatment option for tendon repair.
Collapse
Affiliation(s)
- Qiuzi Long
- Nanjing University of Chinese MedicineNanjing210029China
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- Nanjing Second HospitalNanjing Hospital affiliated to Nanjing University of Chinese MedicineNanjing210003China
| | - Chuanquan Liu
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
| | - Haotian Zheng
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
| | - Mingyue Wang
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
| | - Hanmei Liu
- Nanjing University of Chinese MedicineNanjing210029China
| | - Yue Liu
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
| | - Zhicheng Cao
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
- Department of Orthopaedic SurgeryInstitute of Digital MedicineNanjing First HospitalNanjing Medical UniversityNanjing210006China
| | - Yuzhi Sun
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
- Department of Orthopaedic SurgeryInstitute of Digital MedicineNanjing First HospitalNanjing Medical UniversityNanjing210006China
| | - Qingyun Mo
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
| | - Ludvig J. Backman
- Department of Medical and Translational Biology, AnatomyUmeå UniversityUmeå90187Sweden
- Department of Community Medicine and RehabilitationUmeå UniversityUmeå90187Sweden
| | - Jialin Zhu
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
| | - Lizhi Hu
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
| | - Jinlong Huang
- Nanjing University of Chinese MedicineNanjing210029China
| | - Wei Zhang
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
- Jiangsu Key Laboratory for Biomaterials and DevicesSoutheast UniversityNanjing210096China
- China Orthopedic Regenerative Medicine Group (CORMed)Hangzhou310058China
| | - Jialin Chen
- Center for Stem Cell and Regenerative MedicineSoutheast UniversityNanjing210009China
- School of MedicineSoutheast UniversityNanjing210009China
- Jiangsu Key Laboratory for Biomaterials and DevicesSoutheast UniversityNanjing210096China
- Department of OphthalmologyZhongda HospitalSoutheast UniversityNanjing210009China
| |
Collapse
|
33
|
Vicanolo T, Özcan A, Li JL, Huerta-López C, Ballesteros I, Rubio-Ponce A, Dumitru AC, Nicolás-Ávila JÁ, Molina-Moreno M, Reyes-Gutierrez P, Johnston AD, Martone C, Greto E, Quílez-Alvarez A, Calvo E, Bonzon-Kulichenko E, Álvarez-Velez R, Chooi MY, Kwok I, González-Bermúdez B, Malleret B, Espinosa FM, Zhang M, Wang YL, Sun D, Zhen Chong S, El-Armouche A, Kim KK, Udalova IA, Greco V, Garcia R, Vázquez J, Dopazo A, Plaza GR, Alegre-Cebollada J, Uderhardt S, Ng LG, Hidalgo A. Matrix-producing neutrophils populate and shield the skin. Nature 2025; 641:740-748. [PMID: 40108463 PMCID: PMC12074881 DOI: 10.1038/s41586-025-08741-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/04/2025] [Indexed: 03/22/2025]
Abstract
Defence from environmental threats is provided by physical barriers that confer mechanical protection and prevent the entry of microorganisms1. If microorganisms overcome those barriers, however, innate immune cells use toxic chemicals to kill the invading cells2,3. Here we examine immune diversity across tissues and identify a population of neutrophils in the skin that expresses a broad repertoire of proteins and enzymes needed to build the extracellular matrix. In the naive skin, these matrix-producing neutrophils contribute to the composition and structure of the extracellular matrix, reinforce its mechanical properties and promote barrier function. After injury, these neutrophils build 'rings' of matrix around wounds, which shield against foreign molecules and bacteria. This structural program relies on TGFβ signalling; disabling the TGFβ receptor in neutrophils impaired ring formation around wounds and facilitated bacterial invasion. We infer that the innate immune system has evolved diverse strategies for defence, including one that physically shields the host from the outside world.
Collapse
Affiliation(s)
- Tommaso Vicanolo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Alaz Özcan
- Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jackson LiangYao Li
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Carla Huerta-López
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Iván Ballesteros
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- Department of Neuroscience and Biomedical Sciences, Universidad Carlos III de Madrid, Madrid, Spain
| | - Andrea Rubio-Ponce
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Andra C Dumitru
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | | | - Miguel Molina-Moreno
- Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Pablo Reyes-Gutierrez
- Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Andrew D Johnston
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Catherine Martone
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Eric Greto
- Department of Internal Medicine 3-Rheumatology and Immunology, Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Enrique Calvo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Elena Bonzon-Kulichenko
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- Biochemistry Area, Faculty of Environmental Sciences and Biochemistry, University of Castilla-La Mancha, Toledo, Spain
| | | | - Ming Yao Chooi
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
- Department of Microbiology and Immunology, Immunology Translational Research Programme, NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Immanuel Kwok
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Blanca González-Bermúdez
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI de Caminos, Canales y Puertos, Universidad Politécnica de Madrid, Madrid, Spain
| | - Benoit Malleret
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
- Department of Microbiology and Immunology, Immunology Translational Research Programme, NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Ming Zhang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yu-Long Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Dasheng Sun
- OPO and Organ Transplantation Leading Group, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shu Zhen Chong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
- Department of Microbiology and Immunology, Immunology Translational Research Programme, NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ali El-Armouche
- Institute of Pharmacology and Toxicology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Kevin K Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Irina A Udalova
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Valentina Greco
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Ricardo Garcia
- Instituto de Ciencia de Materiales de Madrid, CSIC, Madrid, Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Ana Dopazo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Gustavo R Plaza
- Instituto de Ciencia de Materiales de Madrid, CSIC, Madrid, Spain
| | | | - Stefan Uderhardt
- Department of Internal Medicine 3-Rheumatology and Immunology, Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.
| | - Lai Guan Ng
- Shanghai Immune Therapy Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Andrés Hidalgo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.
- Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
34
|
Lin Q, Guan S, Peng M, Zhang K, Zhang H, Mo T, Yu H. Comprehensive analysis of SQOR involvement in ferroptosis resistance of pancreatic ductal adenocarcinoma in hypoxic environments. Front Immunol 2025; 16:1513589. [PMID: 40375994 PMCID: PMC12078260 DOI: 10.3389/fimmu.2025.1513589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 04/07/2025] [Indexed: 05/18/2025] Open
Abstract
Introduction Pancreatic ductal adenocarcinoma (PDAC) exhibits higher hypoxia level than most solid tumors, and the presence of intratumoral hypoxia is associated with a poor prognosis. However, the identification of hypoxia levels based on pathological images, and the mechanisms regulating ferroptosis resistance, remain to be elucidated. The objective of this study was to construct a deep learning model to evaluate the hypoxia characteristics of PDAC and to explore the role of Sulfide quinone oxidoreductase (SQOR) in hypoxia-mediated ferroptosis resistance. Methods Multi-omics data were integrated to analyze the correlation between hypoxia score of PDAC, SQOR expression and prognosis, and ferroptosis resistance level. A deep learning model of Whole Slide Images (WSIs) were constructed to predict the hypoxia level of patients. In vitro hypoxia cell models, SQOR knockdown experiments and nude mouse xenograft models were used to verify the regulatory function of SQOR on ferroptosis. Results PDAC exhibited significantly higher hypoxia levels than normal tissues, correlating with reduced overall survival in patients. In slide level, our deep learning model can effectively identify PDAC hypoxia levels with good performance. SQOR was upregulated in tumor tissues and positively associated with both hypoxia score and ferroptosis resistance. SQOR promotes the malignant progression of PDAC in hypoxic environment by enhancing the resistance of tumor cells to ferroptosis. SQOR knockdown resulted in decreased cell viability, decreased migration ability and increased MDA level under hypoxic Ersatin induced conditions. Furthermore, SQOR inhibitor in combination with ferroptosis inducer has the potential to inhibit tumor growth in vivo in a synergistic manner. Discussion This study has established a hypoxia detection model of PDAC based on WSIs, providing a new tool for clinical evaluation. The study revealed a new mechanism of SQOR mediating ferroptosis resistance under hypoxia and provided a basis for targeted therapy.
Collapse
Affiliation(s)
- Quan Lin
- Department of Hepatobiliary Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shiwei Guan
- Department of Hepatobiliary Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Minghui Peng
- Department of Hepatobiliary Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kailun Zhang
- Department of Hepatobiliary Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hewei Zhang
- Department of Hepatobiliary Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Taoming Mo
- Department of Pathology, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Haibo Yu
- Department of Hepatobiliary Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
35
|
Nasti A, Inagaki S, Ho TTB, Seki A, Yoshida K, Satomura K, Sakai Y, Kaneko S, Yamashita T. Cystatin A promotes the antitumor activity of T helper type 1 cells and dendritic cells in murine models of pancreatic cancer. Mol Oncol 2025; 19:1452-1470. [PMID: 39792573 PMCID: PMC12077287 DOI: 10.1002/1878-0261.13796] [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: 05/29/2024] [Revised: 11/21/2024] [Accepted: 12/12/2024] [Indexed: 01/12/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a disease with poor prognosis due to diagnostic and therapeutic limitations. We previously identified cystatin A (CSTA) as a PDAC biomarker and have conducted the present study to investigate the antitumor effects of CSTA. PDAC murine models were established with genetically modified PAN02 tumor cell lines to evaluate the antitumor immune response. PDAC mouse survival was significantly longer with CSTA, and its antitumor effect was mediated mainly by CD4+ cells and partly by CD8+ cells. We also observed an increased infiltration of CD4+ and CD8+ cells in tumors of mice overexpressing CSTA. Phenotypically, we confirmed higher T helper type 1 (Th1) cell activity and increased frequency and activity of M1 macrophages and dendritic cells (DCs) in CSTA-overexpressing mice. Gene expression analysis highlighted pathways related to interferon gamma (IFN-γ) induction and Th1 lymphocyte activation that were induced by CSTA. Macrophages and DCs shifted toward proinflammatory antitumor phenotypes. Furthermore, activated splenocytes of PDAC model mice expressing CSTA had increased proapoptotic activity. CSTA also promoted the selective migration of CD4+ and CD11c+ immune cells in an in vitro migration assay. In conclusion, CSTA exerts antitumor effects by enhancing Th1-mediated antitumor effects through promotion of DC and M1 macrophage activity, thereby increasing immune cell chemotaxis. CSTA could be a novel therapeutic candidate for PDAC.
Collapse
Affiliation(s)
- Alessandro Nasti
- Information‐Based Medicine DevelopmentGraduate School of Medical Sciences, Kanazawa UniversityJapan
| | - Shingo Inagaki
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
| | - Tuyen Thuy Bich Ho
- Information‐Based Medicine DevelopmentGraduate School of Medical Sciences, Kanazawa UniversityJapan
| | - Akihiro Seki
- Department of GastroenterologyKanazawa University HospitalJapan
| | - Keiko Yoshida
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
| | - Kosuke Satomura
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
| | - Yoshio Sakai
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
| | - Shuichi Kaneko
- Information‐Based Medicine DevelopmentGraduate School of Medical Sciences, Kanazawa UniversityJapan
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
- Department of GastroenterologyKanazawa University HospitalJapan
| | - Taro Yamashita
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
- Department of GastroenterologyKanazawa University HospitalJapan
| |
Collapse
|
36
|
Mukherjee P, Mahanty M, Dutta B, Rahaman SG, Sankaran KR, Liu Z, Rahaman SO. Trpv4-mediated mechanotransduction regulates the differentiation of valvular interstitial cells to myofibroblasts: implications for aortic valve stenosis. Am J Physiol Cell Physiol 2025; 328:C1558-C1570. [PMID: 40203884 DOI: 10.1152/ajpcell.00977.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/10/2025] [Accepted: 03/26/2025] [Indexed: 04/11/2025]
Abstract
As aortic valve stenosis (AVS) progresses, the valve tissue also stiffens. This increase in tissue stiffness causes the valvular interstitial cells (VICs) to transform into myofibroblasts in response. VIC-to-myofibroblast differentiation is critically involved in the development of AVS. Herein, we investigated the role of mechanosensitive Ca2+-permeant transient receptor potential vanilloid 4 (Trpv4) channels in matrix stiffness- and transforming growth factor β1 (TGFβ1)-induced VIC-myofibroblast activation. We confirmed Trpv4 functionality in primary mouse wild-type VICs compared with Trpv4 null VICs using live Ca2+ influx detection during application of its selective agonist and antagonist. Using physiologically relevant hydrogels of varying stiffness that respectively mimic healthy or diseased aortic valve tissue stiffness, we found that genetic ablation of Trpv4 blocked matrix stiffness- and TGFβ1-induced VIC-myofibroblast activation as determined by changes in morphology, alterations of expression of α-smooth muscle actin, and modulations of F-actin generation. Our results showed that N-terminal residues 30-130 in Trpv4 were crucial for cellular force generation and VIC-myofibroblast activation, while deletion of residues 1-30 had no noticeable negative effect on these processes. Collectively, these data suggest a differential regulatory role for Trpv4 in stiffness/TGFβ1-induced VIC-myofibroblast activation. Our data further showed that Trpv4 regulates stiffness/TGFβ1-induced PI3K-AKT activity that is required for VIC-myofibroblast differentiation and cellular force generation, suggesting a mechanism by which Trpv4 activity regulates VIC-myofibroblast activation. Altogether, these data identify a novel role for Trpv4 mechanotransduction in regulating VIC-myofibroblast activation, implicating Trpv4 as a potential therapeutic target to slow and/or reverse AVS development.NEW & NOTEWORTHY Aortic valve stenosis (AVS) progression involves stiffened valve tissue, driving valvular interstitial cells (VICs) to transform into myofibroblasts. This study highlights the role of Trpv4 channels in VIC activation triggered by matrix stiffness and TGFß1. Using hydrogels mimicking healthy and diseased valves, researchers found that Trpv4 regulates cellular force generation and differentiation via PI3K-AKT activity. These findings identify Trpv4 as a potential therapeutic target to slow or reverse AVS progression.
Collapse
Affiliation(s)
- Pritha Mukherjee
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland, United States
| | - Manisha Mahanty
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland, United States
| | - Bidisha Dutta
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland, United States
| | - Suneha G Rahaman
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland, United States
| | - Karunakaran R Sankaran
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland, United States
| | - Zhenguo Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri, United States
| | - Shaik O Rahaman
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland, United States
| |
Collapse
|
37
|
Oh JM, Park Y, Lee J, Shen K. Microfabricated Organ-Specific Models of Tumor Microenvironments. Annu Rev Biomed Eng 2025; 27:307-333. [PMID: 40310890 DOI: 10.1146/annurev-bioeng-110222-103522] [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] [Indexed: 05/03/2025]
Abstract
Despite the advances in detection, diagnosis, and treatments, cancer remains a lethal disease, claiming the lives of more than 600,000 people in the United States alone in 2024. To accelerate the development of new therapeutic strategies with improved responses, significant efforts have been made to develop microfabricated in vitro models of tumor microenvironments (TMEs) that address the limitations of animal-based cancer models. These models incorporate several advanced tissue engineering techniques to better reflect the organ- and patient-specific TMEs. Additionally, microfabricated models integrated with next-generation single-cell omics technologies provide unprecedented insights into patient's cellular and molecular heterogeneity and complexity. This review provides an overview of the recent understanding of cancer development and outlines the key TME elements that can be captured in microfabricated models to enhance their physiological relevance. We highlight the recent advances in microfabricated cancer models that reflect the unique characteristics of their organs of origin or sites of dissemination.
Collapse
Affiliation(s)
- Jeong Min Oh
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA;
| | - Yongkuk Park
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA;
| | - Jungwoo Lee
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA;
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
| | - Keyue Shen
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA;
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
38
|
Zaradzki M, Rehberg F, Zwaans V, Hecker M, Karck M, Arif R, Soethoff JP, Wagner AH. Stabilisation of extracellular matrix is crucial to rapamycin-mediated life span increase in Marfan mgR/mgR mice. Biochem Pharmacol 2025; 235:116830. [PMID: 40021021 DOI: 10.1016/j.bcp.2025.116830] [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: 08/18/2024] [Revised: 12/13/2024] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Marfan syndrome is a hereditary connective tissue disorder caused by heterozygous mutations in the fibrillin-1 gene (FBN1) and altered TGF-β signalling. Life-threatening complications involve thoracic aortic aneurysms (TAA) and dissections due to the disruption of microfibrillar assembly in the aortic wall. We previously demonstrated that Rapamycin, a typical mTOR pathway inhibitor, limits the ascending aorta elastolysis and expansion, significantly increasing lifespan in an established murine model of Marfan syndrome (Zaradzki et al., Biochem Pharmacol 2022). This study aimed to investigate how mTOR inhibition stabilises the aorta in fibrillin-1 hypomorphic mgR/mgR mice and previously observed increased life expectancy. We used antibody microarrays to detect protein expression in the proximal thoracic aorta of sham or rapamycin-treated male and female mgR/mgR mice immediately after the two-week treatment. Immunofluorescence staining was performed to visualize and quantify protein expression in the ascending thoracic aorta and arch four weeks after the short-term rapamycin treatment was completed. We showed that rapamycin significantly increased the abundance of extracellular matrix (ECM) proteins like cytokeratin-18 and betaglycan, also known as the TGF-β type 3 receptor (TGFBR3). In addition, it raises the abundance of aggrecanase-2 (ADAMTS5) and xylosyltransferase-1 proteins, enzymes involved in ECM remodelling and homeostasis. In conclusion, rapamycin affects the composition and organization of key ECM components, which determine the structure-function relationships in the aorta, thereby maintaining the balance critical for the increase in life expectancy. Using mTOR modulators for targeted therapy may help to prevent aortic complications of MFS and improve clinical outcomes.
Collapse
Affiliation(s)
- Marcin Zaradzki
- Department of Cardiac Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| | - Franziska Rehberg
- Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Vanessa Zwaans
- Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Markus Hecker
- Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Matthias Karck
- Department of Cardiac Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| | - Rawa Arif
- Department of Cardiac Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| | - Jasmin P Soethoff
- Department of Cardiac Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany.
| | - Andreas H Wagner
- Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| |
Collapse
|
39
|
Yang M, Song X, Zhang F, Li M, Chang W, Wang Z, Li M, Shan H, Li D. Spatial proteomic landscape of primary and relapsed hepatocellular carcinoma reveals immune escape characteristics in early relapse. Hepatology 2025; 81:1452-1467. [PMID: 38900411 DOI: 10.1097/hep.0000000000000979] [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] [Received: 03/22/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND AND AIMS Surgical resection serves as the principal curative strategy for HCC, yet the incidence of postoperative recurrence remains alarmingly high. However, the spatial molecular structural alterations contributing to postoperative recurrence in HCC are still poorly understood. APPROACH AND RESULTS We employed imaging mass cytometry to profile the in situ expression of 33 proteins within 358,729 single cells of 92 clinically annotated surgical specimens from 46 patients who were treated with surgical resections for primary and relapsed tumors. We revealed the recurrence progression of HCC was governed by the dynamic spatial distribution and functional interplay of diverse cell types across adjacent normal, tumor margin, and intratumor regions. Our exhaustive analyses revealed an aggressive, immunosuppression-related spatial ecosystem in relapsed HCC. Additionally, we illustrated the prominent implications of the tumor microenvironment of tumor margins in association with relapse HCC. Moreover, we identified a novel subpopulation of dendritic cells (PDL1 + CD103 + DCs) enriched in the peritumoral area that correlated with early postoperative recurrence, which was further validated in an external cohort. Through the analysis of single-cell RNA sequencing data, we found the interaction of PDL1 + CD103 + DCs with regulatory T cells and exhausted T cells enhanced immunosuppression and immune escape through multiple ligand-receptor pathways. CONCLUSIONS We comprehensively depicted the spatial landscape of single-cell dynamics and multicellular architecture within primary and relapsed HCC. Our findings highlight spatial organization as a prominent determinant of HCC recurrence and provide valuable insight into the immune evasion mechanisms driving recurrence.
Collapse
Affiliation(s)
- Meilin Yang
- Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Xiaoyi Song
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Fan Zhang
- Department of Head and Neck Oncology, Cancer Center, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Mingan Li
- Department of Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wuguang Chang
- Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Zheyan Wang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Man Li
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Department of Information Technology and Data Center, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
- Biobank of the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Hong Shan
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Dan Li
- Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
40
|
McGee Talkington G, Ouvrier B, White AL, Hall G, Umar M, Bix GJ. Imaging Interstitial Fluids and Extracellular Matrix in Cerebrovascular Disorders: Current Perspectives and Clinical Applications. Neuroimaging Clin N Am 2025; 35:181-189. [PMID: 40210376 PMCID: PMC11995915 DOI: 10.1016/j.nic.2025.01.001] [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] [Indexed: 04/12/2025]
Abstract
This article provides a comprehensive review of current neuroimaging techniques for visualizing and quantifying extracellular matrix (ECM) components and interstitial fluid (ISF) dynamics in cerebrovascular disorders. It examines how alterations in ECM composition and ISF movement patterns correlate with various cerebrovascular pathologies, including ischemic stroke, frontotemporal dementia, cerebral small vessel disease, Alzhheimer's disease, and vascular dementia. The review emphasizes novel imaging markers specific to ECM/ISF alterations and their utility in differentiating various cerebrovascular pathologies. Special attention is given to the clinical applications of these imaging techniques for early disease detection, monitoring progression, and guiding therapeutic interventions.
Collapse
Affiliation(s)
- Grant McGee Talkington
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA.
| | - Blake Ouvrier
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA
| | - Amanda Louise White
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Gregory Hall
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Meenakshi Umar
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Gregory Jaye Bix
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA; Department of Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
41
|
Hanitrarimalala V, Prgomet Z, Hedhammar M, Tassidis H, Wingren AG. In vitro 3D modeling of colorectal cancer: the pivotal role of the extracellular matrix, stroma and immune modulation. Front Genet 2025; 16:1545017. [PMID: 40376304 PMCID: PMC12078225 DOI: 10.3389/fgene.2025.1545017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/23/2025] [Indexed: 05/18/2025] Open
Abstract
Colorectal cancer (CRC) is a leading global cancer with high mortality, especially in metastatic cases, with limited therapeutic options. The tumor microenvironment (TME), a network comprising various immune cells, stromal cells and extracellular (ECM) components plays a crucial role in influencing tumor progression and therapy outcome. The genetic heterogeneity of CRC and the complex TME complicates the development of effective, personalized treatment strategies. The prognosis has slowly improved during the past decades, but metastatic CRC (mCRC) is common among patients and is still associated with low survival. The therapeutic options for CRC differ from those for mCRC and include surgery (mostly for CRC), chemotherapy, growth factor receptor signaling pathway targeting, as well as immunotherapy. Malignant CRC cells are established in the TME, which varies depending on the primary or metastatic site. Herein, we review the role and interactions of several ECM components in 3D models of CRC and mCRC tumor cells, with an emphasis on how the TME affects tumor growth and treatment. This comprehensive summary provides support for the development of 3D models that mimic the interactions within the TME, which will be essential for the development of novel anticancer therapies.
Collapse
Affiliation(s)
- Veroniaina Hanitrarimalala
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
- Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - Zdenka Prgomet
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
- Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - My Hedhammar
- KTH Royal Institute of Technology, Division of Protein Technology, Stockholm, Sweden
| | - Helena Tassidis
- Department of Bioanalysis, Faculty of Natural Sciences, Kristianstad University, Kristianstad, Sweden
| | - Anette Gjörloff Wingren
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
- Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden
- Department of Bioanalysis, Faculty of Natural Sciences, Kristianstad University, Kristianstad, Sweden
| |
Collapse
|
42
|
Woods JP, Rackley A, Kwon HR, Olson LE. PDGFRα signaling regulates cartilage and fibrous tissue differentiation during synovial joint development. Nat Commun 2025; 16:4041. [PMID: 40301343 PMCID: PMC12041487 DOI: 10.1038/s41467-025-59207-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 04/14/2025] [Indexed: 05/01/2025] Open
Abstract
Synovial joints develop from mesenchymal structures called interzones, with progenitor cells differentiating into specialized cartilaginous and fibrous tissues of the joint. Platelet-derived growth factor receptor-α (PDGFRα) is a tyrosine kinase expressed by cells of the limb bud, but its role in limb development is unknown. To investigate PDGFRα function, we generated mice expressing mutant PDGFRα with a point mutation (D842V) that increases receptor signaling. Mutant hindlimbs are immobile with knee joints fused by cartilage and lacking ligaments and menisci. The interzone marker Gdf5 is initially expressed at E12.5 but is downregulated thereafter, suggesting a defect in interzone maintenance. Omics analysis of the joint tissues identifies ectopic cartilage matrix expressing genes for cartilage and fibrotic tissue. Thus, elevated PDGFRα signaling corrupts joint development by downregulating Gdf5 and redirecting interzone progenitors into a fibrocartilage fate. This suggests that tight regulation of tyrosine kinase activity is necessary for the development of the mouse knee joint.
Collapse
Affiliation(s)
- John P Woods
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Alex Rackley
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Hae Ryong Kwon
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
| | - Lorin E Olson
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
43
|
Wu Y, Song Y, Soto J, Hoffman T, Lin X, Zhang A, Chen S, Massad RN, Han X, Qi D, Yeh KW, Fang Z, Eoh J, Gu L, Rowat AC, Gu Z, Li S. Viscoelastic extracellular matrix enhances epigenetic remodeling and cellular plasticity. Nat Commun 2025; 16:4054. [PMID: 40307238 PMCID: PMC12043949 DOI: 10.1038/s41467-025-59190-7] [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: 02/14/2025] [Accepted: 04/14/2025] [Indexed: 05/02/2025] Open
Abstract
Extracellular matrices of living tissues exhibit viscoelastic properties, yet how these properties regulate chromatin and the epigenome remains unclear. Here, we show that viscoelastic substrates induce changes in nuclear architecture and epigenome, with more pronounced effects on softer surfaces. Fibroblasts on viscoelastic substrates display larger nuclei, lower chromatin compaction, and differential expression of distinct sets of genes related to the cytoskeleton and nuclear function, compared to those on elastic surfaces. Slow-relaxing viscoelastic substrates reduce lamin A/C expression and enhance nuclear remodeling. These structural changes are accompanied by a global increase in euchromatin marks and local increase in chromatin accessibility at cis-regulatory elements associated with neuronal and pluripotent genes. Consequently, viscoelastic substrates improve the reprogramming efficiency from fibroblasts into neurons and induced pluripotent stem cells. Collectively, our findings unravel the roles of matrix viscoelasticity in epigenetic regulation and cell reprogramming, with implications for designing smart materials for cell fate engineering.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Yang Song
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, 610041, Chengdu, China
| | - Jennifer Soto
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Tyler Hoffman
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Xiao Lin
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Aaron Zhang
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Siyu Chen
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Ramzi N Massad
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Xiao Han
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Dongping Qi
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Kun-Wei Yeh
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Zhiwei Fang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Joon Eoh
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Luo Gu
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Amy C Rowat
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, 90095, USA
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Zhen Gu
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, 310027, Hangzhou, China
| | - Song Li
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
- Broad Stem Cell Research Center, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
44
|
Peng L, Fan Y, Wang L, Han C, Hao Z. Luteolin 7-Glucuronide in Artemisia rupestris L. Extract Attenuates Pulmonary Fibrosis by Inhibiting Fibroblast Activation and FMT via Targeting of TGF-β1. Antioxidants (Basel) 2025; 14:533. [PMID: 40427415 PMCID: PMC12108481 DOI: 10.3390/antiox14050533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/29/2025] Open
Abstract
Pulmonary fibrosis (PF) is a chronic pulmonary disease characterized by excessive extracellular matrix (ECM) deposition, with cigarette smoking being a major risk factor and no effective treatment at present. Transforming growth factor beta 1 (TGF-β1) plays a key role in PF and regulating oxidative stress. This study investigated the effects and mechanisms of Artemisia rupestris L. ethanol extract (ER) on cigarette smoke (CS)-induced PF. We used pull-down and LC-MS analyses to screen and identify compounds that bind to TGF-β1 in ER. We demonstrated that ER inhibits CS-induced PF, lung inflammation, and oxidative stress, thereby improving pulmonary structural injury. The ER inhibits fibroblast activation and fibroblast-to-myofibroblast transition (FMT), reducing collagen deposition for the treatment of PF. We identified the active ingredient in ER that binds to TGF-β1, namely, Luteolin 7-glucuronide (LG). LG inhibits the TGF-β1 signaling pathway through targeted binding to TGF-β1, downregulates the expression of downstream proteins (including collagen I, α-SMA, MMP-2, and MMP-9), and inhibits fibronectin expression. It also inhibits fibroblast activation and FMT, enhances E-cadherin expression to promote fibroblast adhesion, and suppresses collagen deposition, alleviating PF. Based on these findings, we propose that LG might be a promising therapeutic drug candidate for treating PF.
Collapse
Affiliation(s)
- Lingfeng Peng
- Chinese Veterinary Medicine Innovation Center, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (L.P.); (Y.F.); (L.W.); (C.H.)
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
- National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing 100193, China
| | - Yimeng Fan
- Chinese Veterinary Medicine Innovation Center, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (L.P.); (Y.F.); (L.W.); (C.H.)
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
- National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing 100193, China
| | - Luyao Wang
- Chinese Veterinary Medicine Innovation Center, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (L.P.); (Y.F.); (L.W.); (C.H.)
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
- National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing 100193, China
| | - Chao Han
- Chinese Veterinary Medicine Innovation Center, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (L.P.); (Y.F.); (L.W.); (C.H.)
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
- National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing 100193, China
| | - Zhihui Hao
- Chinese Veterinary Medicine Innovation Center, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (L.P.); (Y.F.); (L.W.); (C.H.)
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
- National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing 100193, China
| |
Collapse
|
45
|
Guizzetti M, Mangieri RA, Ezerskiy LA, Hashimoto JG, Bajo M, Farris SP, Homanics GE, Lasek AW, Mayfield RD, Messing RO, Roberto M. ASTROCYTES AND ALCOHOL THROUGHOUT THE LIFESPAN. Biol Psychiatry 2025:S0006-3223(25)01147-3. [PMID: 40311830 DOI: 10.1016/j.biopsych.2025.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/31/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025]
Abstract
Evidence for involvement of astrocytes in several neurodegenerative disorders and in drug addiction has been emerging over the last two decades, but only in recent years have astrocytes been investigated for their roles in alcohol use disorder (AUD). As a result, there is a need to evaluate existing preclinical literature supporting involvement of astrocytes in the effects of alcohol exposure. Here we review emerging evidence about responses of astrocytes to alcohol, and the contributions of astrocytes to the development of AUD. We review studies of single-cell RNA sequencing with a focus on alcohol and astrocyte heterogeneity, astrocyte reactivity, and the role of astrocytes in remodeling the extracellular matrix. Effects of alcohol on astrocyte-modulated synaptic transmission are also discussed emphasizing studies never reviewed before. Since astrocytes play essential roles in brain development, we review recent research on the role of astrocytes in fetal alcohol spectrum disorders (FASD) which may also shed light on fetal development of psychiatric disorders that have a high prevalence in individuals affected by FASD. Finally, this review highlights gaps in knowledge about astrocyte biology and alcohol that need further research. Particularly, the dire need to identify astrocyte subpopulations and molecules that are susceptible to alcohol exposure and may be targets for therapeutic intervention.
Collapse
Affiliation(s)
- Marina Guizzetti
- Oregon Health & Science University and Portland VA Health Care System, Portland, OR.
| | | | | | - Joel G Hashimoto
- Oregon Health & Science University and Portland VA Health Care System, Portland, OR
| | - Michal Bajo
- The Scripps Research Institute, La Jolla, CA
| | | | | | - Amy W Lasek
- Virginia Commonwealth University, Richmond, VA
| | | | | | | |
Collapse
|
46
|
Ding X, Liang Y, Zhou S, Wu Y, Sałata P, Mikolajczk-Martinez A, Khosrawipour V, Zhang Z. Targeting tumor extracellular matrix with nanoparticles to circumvent therapeutic resistance. J Control Release 2025; 383:113786. [PMID: 40306575 DOI: 10.1016/j.jconrel.2025.113786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
Each stage of tumor development is intrinsically linked to the tumor microenvironment (TME), wherein the extracellular matrix (ECM) serves as a vital and abundant component in tumor tissues. The ECM is a non-cellular, three-dimensional macromolecular network scaffold that provides structural support to cells, stores bioactive molecules, and mediates signaling pathways through specific binding to cell surface receptors. Moreover, the ECM in tumor tissues plays a crucial role in impeding drug diffusion and resisting apoptosis induced by conventional anti-cancer therapies that primarily target cancer cells. Therefore, directing attentions towards the tumor ECM can facilitate the identification of novel targets and the development of new therapies. This review aims to summarize the composition, structure, remodeling, and function of tumor ECM, its association with drug resistance, and current targeting strategies, with a specific emphasis on nanoparticles (NPs).
Collapse
Affiliation(s)
- Xinyue Ding
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Yiyu Liang
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Siyuan Zhou
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Yao Wu
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Patricia Sałata
- Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | | | | | - Zhiwen Zhang
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China.
| |
Collapse
|
47
|
Naidoo P, Naicker T. A Disintegrin and Metalloprotease with Thrombospondin Motif, Member 13, and Von Willebrand Factor in Relation to the Duality of Preeclampsia and HIV Infection. Int J Mol Sci 2025; 26:4103. [PMID: 40362344 PMCID: PMC12071684 DOI: 10.3390/ijms26094103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/17/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Normal pregnancy is associated with multiple changes in the coagulation and the fibrinolytic system. In contrast to a non-pregnant state, pregnancy is a hypercoagulable state where the level of VWF increases by 200-375%, affecting coagulation activity. Moreover, in this hypercoagulable state of pregnancy, preeclampsia is exacerbated. ADAMTS13 cleaves the bond between Tyr1605 and Met1606 in the A2 domain of VWF, thereby reducing its molecular weight. A deficiency of ADAMTS13 originates from mutations in gene or autoantibodies formed against the protease, leading to defective enzyme production. Von Willebrand protein is critical for hemostasis and thrombosis, promoting thrombus formation by mediating the adhesion of platelets and aggregation at high shear stress conditions within the vessel wall. Mutations in VWF disrupts multimer assembly, secretion and/or catabolism, thereby influencing bleeding. VWF is the primary regulator of plasma ADAMTS13 levels since even minute amounts of active ADAMTS13 protease have a significant inhibitory effect on inflammation and thrombosis. VWF is released as a result of endothelial activation brought on by HIV infection. The SARS-CoV-2 infection promotes circulating proinflammatory cytokines, increasing endothelial secretion of ultra large VWF that causes an imbalance in VWF/ADAMTS13. Raised VWF levels corresponds with greater platelet adhesiveness, promoting a thrombotic tendency in stenotic vessels, leading to increased shear stress conditions.
Collapse
Affiliation(s)
| | - Thajasvarie Naicker
- Optics & Imaging Centre, Doris Duke Medical Research Institute, University of KwaZulu-Natal, 719 Umbilo Road, Congella, Durban 4013, South Africa
| |
Collapse
|
48
|
Chen J, Luo Y, Xie L, Meng N, Li S, Xiao S, Li X. Long-term alterations of collagen reconstruction and basement membrane regeneration after corneal full-thickness penetrating injury in rabbits. PLoS One 2025; 20:e0320802. [PMID: 40273158 PMCID: PMC12021247 DOI: 10.1371/journal.pone.0320802] [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: 08/14/2024] [Accepted: 02/25/2025] [Indexed: 04/26/2025] Open
Abstract
PURPOSE To investigate the long-term alterations of collagen reconstruction and basement membrane (BM) regeneration after corneal full-thickness penetrating injury in rabbits. METHODS The corneal full-thickness penetrating injury model was established in the left eye of New Zealand White rabbits using a 2.0 mm trephine. All corneas were evaluated using slit-lamp photography, hematoxylin and eosin staining, immunofluorescent staining for collagen types I and III (Col I, III), and transmission electron microscopy for collagen fibers and basement membrane. RESULTS Between 3 days and 3 weeks, Col I and III expression were documented, exhibiting a largely disorganized distribution throughout the stromal thickness. At 3 weeks, the epithelial basement membrane (EBM) partially regenerated. From 3 weeks to 2 months, Col III was undetectable in the anterior stroma but present in the posterior stroma; Col I was disorganized in the posterior stroma. At 2 months, Descemet's membrane (DM) exhibited incomplete regeneration. From 3 to 4 months, Col I was disorganized in only a small part of the posterior stroma; Col III persisted in the posterior stroma; the EBM fully regenerated while DM exhibited incomplete regeneration. CONCLUSIONS Following full-thickness corneal injury, persistent fibrosis within the posterior stroma appears to be primarily responsible for the persistence of corneal scarring. Notably, regeneration of the EBM coincides with remodeling of the anterior stroma, whereas incomplete regeneration of DM is associated with posterior stromal fibrosis.
Collapse
Affiliation(s)
- Jingjing Chen
- Department of Ophthalmology, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yuqing Luo
- Department of Ophthalmology, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Luting Xie
- Department of Ophthalmology, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Na Meng
- Department of Ophthalmology, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Sumei Li
- Department of Ophthalmology, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shifang Xiao
- Department of Ophthalmology, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xia Li
- Department of Ophthalmology, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
49
|
Goux Corredera I, Amato G, Moya-Rull D, Garreta E, Montserrat N. Unlocking the full potential of human pluripotent stem cell-derived kidney organoids through bioengineering. Kidney Int 2025:S0085-2538(25)00327-8. [PMID: 40280411 DOI: 10.1016/j.kint.2025.01.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/17/2025] [Accepted: 01/28/2025] [Indexed: 04/29/2025]
Abstract
Human pluripotent stem cells hold inherent properties, allowing researchers to recapitulate key morphogenetic processes. These characteristics, coupled with bioengineering techniques, have led to the definition of early procedures to derive organ-like cell cultures, the so-called organoids. With regard to kidney organoids, challenges stand ahead, such as the need to enhance cellular composition, maturation, and function to that found in the native organ. To this end, the kidney organoid field has begun to nourish from innovative engineering approaches aiming to gain control on the externally imposed biochemical and biophysical cues. In this review, we first introduce how previous research in kidney development and human pluripotent stem cells has informed the establishment of current kidney organoid procedures. We then discuss recent engineering approaches to guide kidney organoid self-organization, differentiation, and maturation. In addition, we present current strategies to engineer vascularization and promote in vivo-like physiological microenvironments as potential solutions to increase kidney organoid lifespan and functionality. We finally emphasize how working at the cusp of cell mechanics and computational biology will set the ground for successful translational applications of kidney organoids.
Collapse
Affiliation(s)
- Iphigénie Goux Corredera
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Gaia Amato
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Daniel Moya-Rull
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Elena Garreta
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; University of Barcelona, Barcelona, Spain.
| | - Nuria Montserrat
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
50
|
Zhou X, Li R, Lai M, Lai C. Exploring molecular and cellular mechanisms of Pre-Metastatic niche in renal cell carcinoma. Mol Cancer 2025; 24:121. [PMID: 40264130 PMCID: PMC12012986 DOI: 10.1186/s12943-025-02315-9] [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: 12/21/2024] [Accepted: 03/25/2025] [Indexed: 04/24/2025] Open
Abstract
Renal cell carcinoma (RCC) is among the most frequently occurring types of cancer, and its metastasis is a major contributor to its elevated mortality. Before the primary tumor metastasizes to secondary or distant organs, it remodels the microenvironment of these sites, creating a pre-metastatic niche (PMN) conducive to the colonization and growth of metastatic tumors. RCC releases a variety of biomolecules that induce angiogenesis, alter vascular permeability, modulate immune cells to create an immunosuppressive microenvironment, affect extracellular matrix remodeling and metabolic reprogramming, and determine the organotropism of metastasis through different signaling pathways. This review summarizes the principal processes and mechanisms underlying the formation of the premetastatic niche in RCC. Additionally, we emphasize the significance and potential of targeting PMNs for the prevention and treatment of tumor metastasis in future therapeutic approaches. Finally, we summarized the currently potential targeted strategies for detecting and treating PMN in RCC and provide a roadmap for further in-depth studies on PMN in RCC.
Collapse
Affiliation(s)
- Xiao Zhou
- Department of Pathology, and Department of Pathology Sir Run Run Shaw Hospital, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Science (2019RU042), Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Ruirui Li
- Institute of Immunology, Department of Respiratory Disease of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Maode Lai
- Department of Pathology, and Department of Pathology Sir Run Run Shaw Hospital, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Science (2019RU042), Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
| | - Chong Lai
- Department of Urology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| |
Collapse
|