1
|
Liu J, Chen Y, Sun B, Xu D, Wang J, Sun Z, Liu P, Jing F, Song Y, Xia B. Toxicological effects of micro/nanoplastics and benzo[a]pyrene on cellular and molecular responses of Apostichopus japonicus (Selenka, 1867) during intestinal regeneration. JOURNAL OF HAZARDOUS MATERIALS 2025; 491:138003. [PMID: 40120257 DOI: 10.1016/j.jhazmat.2025.138003] [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: 10/15/2024] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Micro(nano)plastics (M/NPs) are pervasive in marine environments. Benzo[a]pyrene (B[a]P), a typical polycyclic aromatic hydrocarbon (PAH), possesses teratogenic, mutagenic, and carcinogenic properties. B[a]P can accumulate on M/NPs, altering their toxicity. This study investigated individual and combined effects of M/NPs and B[a]P on the intestinal regeneration of the benthic invertebrate Apostichopus japonicus. Eviscerated sea cucumbers were exposed to 0.1 mg L-1 M/NPs (80 nm [NP80] or 20 μm [MP20]) and/or 0.03 μg L-1 B[a]P for 28 days. Cell proliferation, antioxidant and immunoenzyme activity, gene expression, and microbial community in the regenerated intestine were assessed. It demonstrated that combined exposure prolonged regeneration process, leading to increased oxidative stress and intestinal damage. Differential gene expression analysis revealed that co-exposure and single NP80 exposure both significantly changed translation-related processes, while single MP20 exposure primarily affected lipid metabolism. All treatments significantly altered the intestinal microbiota. Under the MP20+B[a]P treatment, Ralstonia abundance significantly increased, while Cobetia and Paracoccus abundances decreased. In general, co-exposure exerted more detrimental effects on intestinal regeneration than any single exposure, with MP20+B[a]P demonstrating more severe impacts. This study provides novel insights into the biotoxicity of M/NPs and B[a]P, contributing to better understanding of the detriments of microplastics and PAHs on marine benthic invertebrates.
Collapse
Affiliation(s)
- Ji Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, China
| | - Yanru Chen
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, China
| | - Baiqin Sun
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, China
| | - Dongxue Xu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, China
| | - Jinye Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, China
| | - Zhenlong Sun
- Jiangsu Zhongyang Group Co., Ltd., Nantong, Jiangsu Province 226600, China.
| | - Peng Liu
- Shandong Fisheries Development and Resources Conservation Center, Jinan, Shandong Province 250013, China
| | - Futao Jing
- Shandong Fisheries Development and Resources Conservation Center, Jinan, Shandong Province 250013, China
| | - Yize Song
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, China
| | - Bin Xia
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, China.
| |
Collapse
|
2
|
Macdonald JK, Zambrzycki SC, Taylor HB, Dunne JB, Quick M, Mehta AS, Drake RR, Angel PM. Optimization of Collagenase Proteomics for Improved Mass Spectrometry Imaging Peptide Identification. Anal Chem 2025; 97:7672-7681. [PMID: 40154494 PMCID: PMC12005190 DOI: 10.1021/acs.analchem.4c04818] [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: 09/06/2024] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 04/01/2025]
Abstract
The extracellular matrix (ECM) is composed of a dynamically regulated collagenous scaffold that provides structure, conveys cellular and environmental communication, and contributes to disease progression. Collagen proteins derived from clinically archived formalin-fixed, paraffin-embedded (FFPE) tissues are analytically challenging due to dense post-translational modifications, high proline content, and insolubility. A recent advancement in ECM proteomics is the use of collagenase type III, an ECM-specific bacterial protease, to target native collagenous structures on-tissue for peptide imaging. The resulting collagenase-generated peptides have biochemical differences compared to tryptic peptides, creating analytical challenges in elucidating peptide sequence information. In this study, we characterize collagenase as a proteomic enzyme for ECM-targeted liquid chromatography trapped ion mobility spectrometry tandem mass spectrometry (LC-TIMS-MS/MS) and matrix-assisted laser/desorption ionization mass spectrometry imaging (MALDI-MSI) proteomic workflows. We then optimized collagenase-generated peptide sequencing for MALDI-MSI peptide identification from clinically archived FFPE tissue sections. Soluble rat tail collagen solution is used as a collagen standard to elucidate tryptic and collagenase cleavage sites within collagen. Proteomic readouts of FFPE tissue are compared across trypsin and collagenase digests to assess for ECM enrichment by collagenase in biologically complex samples. Optimized methods for MALDI-MSI peptide identification are comprehensively detailed from sample preparation to MS data acquisition and MS data analysis for reproducible implementation. On-tissue digestion followed by liquid surface extraction (LSE), inclusion of singly charged peptides during data acquisition, and implementation of nonspecific cleavage during database searching resulted in the most collagenase-generated peptide spectrum matches as well as MALDI-MSI peptide identifications. This research establishes parameters for the optimal identification of peptides from collagenase-directed ECM proteomic workflows for targeted spatial analysis of the ECM.
Collapse
Affiliation(s)
- Jade K. Macdonald
- Department
of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Stephen C. Zambrzycki
- Department
of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Harrison B. Taylor
- Department
of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Jaclyn B. Dunne
- Department
of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Montana Quick
- Bruker
Daltonics, Billerica, Massachusetts 01821, United States
| | - Anand S. Mehta
- Department
of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Richard R. Drake
- Department
of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Peggi M. Angel
- Department
of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| |
Collapse
|
3
|
Lee F, Shao X, Considine JM, Gao Y(T, Naba A. Time-lapse tryptic digestion: a proteomic approach to improve sequence coverage of extracellular matrix proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645502. [PMID: 40196545 PMCID: PMC11974830 DOI: 10.1101/2025.03.26.645502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The extracellular matrix (ECM) is a complex and dynamic meshwork of proteins providing structural support to cells. It also provides biochemical signals governing cellular processes, including proliferation, adhesion, and migration. Alterations of ECM structure and/or composition have been linked to many pathological processes, including cancer and fibrosis. Over the past decade, mass-spectrometry-based proteomics has become the state-of-the-art method to profile the protein composition of ECMs. However, existing methods do not fully capture the broad dynamic range of protein abundances in the ECM. They also do not permit to achieve the high coverage needed to gain finer biochemical on ECM proteoforms (e.g., isoforms, post-translational modifications) and topographical information critical to better understand ECM protein functions. Here, we present the development of a time-lapsed proteomic pipeline using limited tryptic proteolysis and sequential release of peptides over time. This experimental pipeline was combined with data-independent acquisition mass spectrometry and the assembly of a custom matrisome spectral library to enhance peptide-to-spectrum matching. This pipeline shows superior protein identification, peptide-to-spectrum matching, and significantly increased sequence coverage against standard ECM proteomic pipelines. Exploiting the spatio-temporal resolution of this method, we further demonstrate how time-resolved 3-dimensional peptide mapping can identify protein regions differentially susceptible to trypsin, which may aid in identifying protein-protein interaction sites.
Collapse
Affiliation(s)
- Fred Lee
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, U.S.A
| | - Xinhao Shao
- College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, U.S.A
| | - James M Considine
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, U.S.A
| | - Yu (Tom) Gao
- College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, U.S.A
- University of Illinois Cancer Center, Chicago, IL 60612, U.S.A
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, U.S.A
- University of Illinois Cancer Center, Chicago, IL 60612, U.S.A
| |
Collapse
|
4
|
Roy A, Hoff A, Her TK, Ariyaratne G, Gutiérrez RL, Tahawi MHDN, Rajagopalan KS, Brown MR, Omori K, Lewis-Brinkman S, Nguyen T, Soto-González A, Peterson QP, Matveyenko AV, Javeed N. Lipotoxicity Induces β-cell Small Extracellular Vesicle-Mediated β-cell Dysfunction in Male Mice. Endocrinology 2025; 166:bqaf067. [PMID: 40179251 PMCID: PMC12006739 DOI: 10.1210/endocr/bqaf067] [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: 09/15/2024] [Revised: 12/19/2024] [Accepted: 04/01/2025] [Indexed: 04/05/2025]
Abstract
Chronically elevated circulating excess free fatty acids (ie, lipotoxicity) is a pathological process implicated in several metabolic disorders, including obesity-driven type 2 diabetes (T2D). Lipotoxicity exerts detrimental effects on pancreatic islet β-cells by reducing glucose-stimulated insulin secretion (GSIS), altering β-cell transcriptional identity, and promoting apoptosis. While β-cell-derived small extracellular vesicles (sEV) have been shown to contribute to β-cell failure in T2D, their specific role in lipotoxicity-mediated β-cell failure remains to be elucidated. In this work, we demonstrate that lipotoxicity enhances the release of sEVs from β-cells, which exhibit altered proteomic and lipidomic profiles. These palmitate (PAL)-exposed extracellular vesicles (EVs) induce β-cell dysfunction in healthy mouse and human islets and trigger significant islet transcriptional changes, including the upregulation of genes associated with the TGFβ/Smad3 pathway, as noted by RNA sequencing. Importantly, pharmacological inhibition of the TGFβI/II receptor improved PAL EV-induced β-cell dysfunction, underscoring their involvement in activating the TGFβ/Smad3 pathway during this process. We have comprehensively characterized lipotoxic β-cell sEVs and implicated their role in inducing β-cell functional failure in T2D. These findings highlight potential avenues for therapeutic interventions targeting sEV-mediated pathways to preserve β-cell health in metabolic disorders.
Collapse
Affiliation(s)
- Abhishek Roy
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Alexandra Hoff
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Tracy K Her
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Gallage Ariyaratne
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Roberto-León Gutiérrez
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - M H D Noor Tahawi
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Matthew R Brown
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kazuno Omori
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Sean Lewis-Brinkman
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Thanh Nguyen
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Quinn P Peterson
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Division of Endocrinology, Diabetes, and Metabolism, Mayo Clinic, Rochester, MN 55905, USA
| | - Aleksey V Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Division of Endocrinology, Diabetes, and Metabolism, Mayo Clinic, Rochester, MN 55905, USA
| | - Naureen Javeed
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Division of Endocrinology, Diabetes, and Metabolism, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
5
|
Amin A, Ferreira DV, Figueiredo LM. How pathogens drive adipose tissue loss in the host. Curr Opin Microbiol 2025; 85:102597. [PMID: 40080953 DOI: 10.1016/j.mib.2025.102597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/05/2025] [Accepted: 02/17/2025] [Indexed: 03/15/2025]
Abstract
Weight loss is a hallmark of many infections, including those caused by bacteria, fungi and parasites. This loss is often attributed to infection-induced anorexia and the need to mobilise energy from internal sources to cope with the pathogens. Weight loss during infection results from a significant reduction of muscle and fat mass, two organs that together account for approximately 60% of body mass in the healthy state. While muscle wasting is a well-documented aspect of infection-related weight loss, adipose tissue loss via lipolysis also plays a critical role and can determine disease outcomes. This review explores the regulators of adipose tissue depletion via excessive lipolysis during infection, the probable mechanisms, and the potential consequences for host survival and pathogen fitness.
Collapse
Affiliation(s)
- Abdulbasit Amin
- Gulbenkian Institute for Molecular Medicine, Edificio Egas Moniz, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal; Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Nigeria
| | - David V Ferreira
- Gulbenkian Institute for Molecular Medicine, Edificio Egas Moniz, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
| | - Luisa M Figueiredo
- Gulbenkian Institute for Molecular Medicine, Edificio Egas Moniz, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal.
| |
Collapse
|
6
|
O'Leary M, Greed E, Pritchard J, Struszczak L, Bozbaş E, Bowtell J. The skeletal muscle proteomic determinants of neuromuscular function in young and older women following 8 weeks of resistance training. Exp Physiol 2025; 110:438-453. [PMID: 39663727 PMCID: PMC11868017 DOI: 10.1113/ep092328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/05/2024] [Indexed: 12/13/2024]
Abstract
Resistance training (RT) is the gold standard intervention for ameliorating sarcopenia. Outstanding mechanistic questions remain regarding the malleability of the molecular determinants of skeletal muscle function in older age. Discovery of proteomics can expand such knowledge. We aimed to compare the effect of RT on the skeletal muscle proteome and neuromuscular function (NMF) in older and younger women. Seven young (22 ± 6 years) and eight older (63 ± 5 years) women completed 8 weeks' leg RT. Pre- and post-training, measures of leg and handgrip strength, NMF and vastus lateralis (VL) biopsies were obtained. Tandem-mass-tagged skeletal muscle proteomic analyses were performed. Data were analysed using differential expression and weighted gene co-expression network approaches. Proteins related to skeletal muscle contraction were lower in older skeletal muscle; this was not normalised by RT. Following RT, older women had higher expression of VL mitochondrial biogenesis proteins compared to the young, a reversal of pre-training observations. Seventy proteins were differentially expressed between age groups. VL expression of these proteins in older women was consistently and significantly associated with poorer leg strength/NMF. Conversely, VL expression of these proteins in older women was often associated with greater handgrip strength. This study has identified important differences in the molecular responses of young and old skeletal muscle to RT. We have demonstrated their close relationship with skeletal muscle function. Proteins that are refractory to RT may represent targets to ameliorate sarcopenia. We have described a 'proteomic-function' relationship that appears to be muscle-specific. Future research should further unpick these complex relationships.
Collapse
Affiliation(s)
- Mary O'Leary
- Faculty of Health and Life Sciences, Department of Public Health and Sport SciencesUniversity of ExeterExeterDevonUK
| | - Elsa Greed
- Faculty of Health and Life Sciences, Department of Public Health and Sport SciencesUniversity of ExeterExeterDevonUK
- School of Sport, Exercise and Rehabilitation SciencesUniversity of BirminghamEdgbastonBirminghamUK
| | - Jack Pritchard
- Faculty of Health and Life Sciences, Department of Public Health and Sport SciencesUniversity of ExeterExeterDevonUK
| | - Lauren Struszczak
- Faculty of Health and Life Sciences, Department of Public Health and Sport SciencesUniversity of ExeterExeterDevonUK
| | - Esra Bozbaş
- Faculty of Health and Life Sciences, Department of Public Health and Sport SciencesUniversity of ExeterExeterDevonUK
| | - Joanna Bowtell
- Faculty of Health and Life Sciences, Department of Public Health and Sport SciencesUniversity of ExeterExeterDevonUK
| |
Collapse
|
7
|
Handschin C, Shalhoub H, Mazet A, Guyon C, Dusserre N, Boutet-Robinet E, Oliveira H, Guillermet-Guibert J. Biotechnological advances in 3D modeling of cancer initiation. Examples from pancreatic cancer research and beyond. Biofabrication 2025; 17:022008. [PMID: 40018875 DOI: 10.1088/1758-5090/adb51c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 02/12/2025] [Indexed: 03/01/2025]
Abstract
In recent years, biofabrication technologies have garnered significant attention within the scientific community for their potential to create advancedin vitrocancer models. While these technologies have been predominantly applied to model advanced stages of cancer, there exists a pressing need to develop pertinent, reproducible, and sensitive 3D models that mimic cancer initiation lesions within their native tissue microenvironment. Such models hold profound relevance for comprehending the intricacies of cancer initiation, to devise novel strategies for early intervention, and/or to conduct sophisticated toxicology assessments of putative carcinogens. Here, we will explain the pivotal factors that must be faithfully recapitulated when constructing these models, with a specific focus on early pancreatic cancer lesions. By synthesizing the current state of research in this field, we will provide insights into recent advances and breakthroughs. Additionally, we will delineate the key technological and biological challenges that necessitate resolution in future endeavors, thereby paving the way for more accurate and insightfulin vitrocancer initiation models.
Collapse
Affiliation(s)
- C Handschin
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - H Shalhoub
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, 2 av Hubert Curien, Toulouse, France
- Labex Toucan, 2 av Hubert Curien, Toulouse, France
| | - A Mazet
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - C Guyon
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, 2 av Hubert Curien, Toulouse, France
- Labex Toucan, 2 av Hubert Curien, Toulouse, France
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| | - N Dusserre
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - E Boutet-Robinet
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| | - H Oliveira
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - J Guillermet-Guibert
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, 2 av Hubert Curien, Toulouse, France
- Labex Toucan, 2 av Hubert Curien, Toulouse, France
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| |
Collapse
|
8
|
Metzcar J, Duggan BS, Fischer B, Murphy M, Heiland R, Macklin P. A Simple Framework for Agent-Based Modeling with Extracellular Matrix. Bull Math Biol 2025; 87:43. [PMID: 39937344 PMCID: PMC11821717 DOI: 10.1007/s11538-024-01408-8] [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: 02/02/2023] [Accepted: 12/21/2024] [Indexed: 02/13/2025]
Abstract
Extracellular matrix (ECM) is a key component of the cellular microenvironment and critical in multiple disease and developmental processes. Representing ECM and cell-ECM interactions is a challenging multiscale problem as they span molecular-level details to tissue-level dynamics. While several computational frameworks exist for ECM modeling, they often focus on very detailed modeling of individual ECM fibers or represent only a single aspect of the ECM. Using the PhysiCell agent-based modeling platform, we developed a framework of intermediate detail with the ability to capture bidirectional cell-ECM interactions. We represent a small region of ECM, an ECM element, with three variables describing its local microstructure: anisotropy, density, and overall fiber orientation. To spatially model the ECM, we use an array of ECM elements. Cells remodel local ECM microstructure and in turn, local microstructure impacts cellular motility. We demonstrate the utility of this framework and reusability of its core cell-ECM interaction model through examples in cellular invasion, wound healing, basement membrane degradation, and leader-follower collective migration. Despite the relative simplicity of the framework, it is able to capture a broad range of cell-ECM interactions of interest to the modeling community. Furthermore, variables representing the ECM microstructure are accessible through simple programming interfaces. This allows them to impact cell behaviors, such as proliferation and death, without requiring custom code for each interaction, particularly through PhysiCell's modeling grammar, enabling rapid modeling of a diverse range of cell-matrix biology. We make this framework available as a free and open source software package at https://github.com/PhysiCell-Models/collective-invasion .
Collapse
Affiliation(s)
- John Metzcar
- Intelligent Systems Engineering, Indiana University, 700 N. Woodlawn, Bloomington, IN, 47408, USA
- Informatics, Indiana University, 901 E. Tenth Street, Bloomington, IN, 47408, USA
| | - Ben S Duggan
- Computer Science, Indiana University, 700 N. Woodlawn, Bloomington, IN, 47408, USA
| | - Brandon Fischer
- Intelligent Systems Engineering, Indiana University, 700 N. Woodlawn, Bloomington, IN, 47408, USA
| | - Matthew Murphy
- Informatics, Indiana University, 901 E. Tenth Street, Bloomington, IN, 47408, USA
| | - Randy Heiland
- Intelligent Systems Engineering, Indiana University, 700 N. Woodlawn, Bloomington, IN, 47408, USA
| | - Paul Macklin
- Intelligent Systems Engineering, Indiana University, 700 N. Woodlawn, Bloomington, IN, 47408, USA.
| |
Collapse
|
9
|
Huang T, Fakurazi S, Cheah PS, Ling KH. Dysregulation of REST and its target genes impacts the fate of neural progenitor cells in down syndrome. Sci Rep 2025; 15:2818. [PMID: 39843579 PMCID: PMC11754635 DOI: 10.1038/s41598-025-87314-y] [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: 09/19/2024] [Accepted: 01/17/2025] [Indexed: 01/30/2025] Open
Abstract
Increasing shreds of evidence suggest that neurogenic-to-gliogenic shift may be critical to the abnormal neurodevelopment observed in individuals with Down syndrome (DS). REST, the Repressor Element-1 Silencing Transcription factor, regulates the differentiation and development of neural cells. Downregulation of REST may lead to defects in post-differentiation neuronal morphology in the brain of the DS fetal. This study aims to elucidate the role of REST in DS-derived NPCs using bioinformatics analyses and laboratory validations. We identified and validated vital REST-targeted DEGs: CD44, TGFB1, FN1, ITGB1, and COL1A1. Interestingly, these genes are involved in neurogenesis and gliogenesis in DS-derived NPCs. Furthermore, we identified nuclear REST loss and the neuroblast marker, DCX, was downregulated in DS human trisomic induced pluripotent stem cells (hiPSCs)-derived NPCs, whereas the glioblast marker, NFIA, was upregulated. Our findings indicate that the loss of REST is critical in the neurogenic-to-gliogenic shift observed in DS-derived NPCs. REST and its target genes may collectively regulate the NPC phenotype.
Collapse
Affiliation(s)
- Tan Huang
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia
| | - Sharida Fakurazi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia
| | - Pike-See Cheah
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia
- Brain and Mental Health Research Advancement and Innovation Networks (PUTRA® BRAIN), Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia
- Malaysian Research Institute on Ageing (MyAgeing®), Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia
| | - King-Hwa Ling
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia.
- Brain and Mental Health Research Advancement and Innovation Networks (PUTRA® BRAIN), Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia.
- Malaysian Research Institute on Ageing (MyAgeing®), Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia.
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia.
| |
Collapse
|
10
|
McNicol GR, Dalby MJ, Stewart PS. A theoretical model for focal adhesion and cytoskeleton formation in non-motile cells. J Theor Biol 2025; 596:111965. [PMID: 39442686 DOI: 10.1016/j.jtbi.2024.111965] [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: 02/05/2024] [Revised: 07/25/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
To function and survive cells need to be able to sense and respond to their local environment through mechanotransduction. Crucially, mechanical and biochemical perturbations initiate cell signalling cascades, which can induce responses such as growth, apoptosis, proliferation and differentiation. At the heart of this process are actomyosin stress fibres (SFs), which form part of the cell cytoskeleton, and focal adhesions (FAs), which bind this cytoskeleton to the extra-cellular matrix (ECM). The formation and maturation of these structures (connected by a positive feedback loop) is pivotal in non-motile cells, where SFs are generally of ventral type, interconnecting FAs and producing isometric tension. In this study we formulate a one-dimensional bio-chemo-mechanical continuum model to describe the coupled formation and maturation of ventral SFs and FAs. We use a set of reaction-diffusion-advection equations to describe three sets of biochemical events: the polymerisation of actin and subsequent bundling into activated SFs; the formation and maturation of cell-substrate adhesions; and the activation of signalling proteins in response to FA and SF formation. The evolution of these key proteins is coupled to a Kelvin-Voigt viscoelastic description of the cell cytoplasm and the ECM. We employ this model to understand how cells respond to external and intracellular cues in vitro and are able to reproduce experimentally observed phenomena including non-uniform cell striation and cells forming weaker SFs and FAs on softer substrates.
Collapse
Affiliation(s)
- Gordon R McNicol
- School of Mathematics and Statistics, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G11 6EW, United Kingdom
| | - Peter S Stewart
- School of Mathematics and Statistics, University of Glasgow, Glasgow, G12 8QQ, United Kingdom.
| |
Collapse
|
11
|
Greed E, Pritchard J, Struszczak L, Bozbaş E, Ek G, Acheson J, Winney B, Qadir A, Wong KKL, Bowtell J, O’Leary M. Shatavari supplementation during eight weeks of resistance training increases training load, enhances skeletal muscle contractility and alters the skeletal muscle proteome in older women. Front Nutr 2025; 11:1498674. [PMID: 39834460 PMCID: PMC11743497 DOI: 10.3389/fnut.2024.1498674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Shatavari is a herbal dietary supplement that may increase skeletal muscle strength in younger and older adults. Shatavari contains compounds with both estradiol-like and antioxidant properties, which could enhance muscle function. Postmenopausal women may derive the greatest benefit, as estrogen deficiency adversely impacts skeletal muscle function. However, mechanistic insights are limited and the effects of shatavari on muscle function require further characterization. Methods In this randomized, double-blind trial, 17 young (23 ± 5 yr) and 22 older (63 ± 5 yr) women completed an 8-week leg resistance training programme. They consumed either a placebo or shatavari (1000 mg/d, equivalent to 26,500 mg/d fresh weight) supplement throughout. Pre and post training, measures of leg strength, neuromuscular function and vastus lateralis (VL) biopsies were obtained. Tandem-mass-tagged VL proteomic analyses were performed. Data were analyzed using a differential expression (Reactome) approach. Results Shatavari supplementation increased 8-week training load in older women (leg press repetitions completed, p = 0.049, η p 2 = 0.198; maximum weight lifted each week, p = 0.03, η p 2 = 0.386; ANCOVA). There was no effect of shatavari on muscle strength post-training. VL half relaxation time was shortened post-training in older women supplemented with shatavari (post-training change: shatavari -11.74 ± 11.93%, placebo 0.42 ± 14.73%, p = 0.021; ANCOVA). Shatavari supplementation diminished the expression of extracellular matrix proteins in both cohorts. Expression of proteins related to striated muscle contraction, transcription and translation were decreased by shatavari supplementation in older women. Discussion These novel observations support the notion that shatavari supplementation confers resistance to neuromuscular fatigue in older women. This could ameliorate sarcopenic declines in skeletal muscle function.
Collapse
Affiliation(s)
- Elsa Greed
- Faculty of Health and Life Sciences, Department of Public Health and Sport Sciences, University of Exeter, Exeter, United Kingdom
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jack Pritchard
- Faculty of Health and Life Sciences, Department of Public Health and Sport Sciences, University of Exeter, Exeter, United Kingdom
| | - Lauren Struszczak
- Faculty of Health and Life Sciences, Department of Public Health and Sport Sciences, University of Exeter, Exeter, United Kingdom
| | - Esra Bozbaş
- Faculty of Health and Life Sciences, Department of Public Health and Sport Sciences, University of Exeter, Exeter, United Kingdom
| | - Georgia Ek
- Faculty of Health and Life Sciences, Department of Public Health and Sport Sciences, University of Exeter, Exeter, United Kingdom
| | - Jordan Acheson
- Faculty of Health and Life Sciences, Department of Public Health and Sport Sciences, University of Exeter, Exeter, United Kingdom
- Department of Sport and Exercise Sciences, Institute of Sport, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Ben Winney
- Faculty of Health and Life Sciences, Department of Public Health and Sport Sciences, University of Exeter, Exeter, United Kingdom
| | - Aaliyah Qadir
- Faculty of Health and Life Sciences, Department of Public Health and Sport Sciences, University of Exeter, Exeter, United Kingdom
| | - Karl Ka-Lam Wong
- Faculty of Health and Life Sciences, Department of Public Health and Sport Sciences, University of Exeter, Exeter, United Kingdom
| | - Joanna Bowtell
- Faculty of Health and Life Sciences, Department of Public Health and Sport Sciences, University of Exeter, Exeter, United Kingdom
| | - Mary O’Leary
- Faculty of Health and Life Sciences, Department of Public Health and Sport Sciences, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
12
|
Gibert-Ramos A, Andrés-Rozas M, Pastó R, Alfaro-Retamero P, Guixé-Muntet S, Gracia-Sancho J. Sinusoidal communication in chronic liver disease. Clin Mol Hepatol 2025; 31:32-55. [PMID: 39355871 PMCID: PMC11791556 DOI: 10.3350/cmh.2024.0734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 10/03/2024] Open
Abstract
The liver sinusoid, mainly composed of liver sinusoidal endothelial cells, hepatic macrophages and hepatic stellate cells, shapes the hepatic vasculature and is key to maintaining liver homeostasis and function. During chronic liver disease (CLD), the function of sinusoidal cells is impaired, being directly involved in the progression of liver fibrosis, cirrhosis, and main clinical complications including portal hypertension and hepatocellular carcinoma. In addition to their roles in liver diseases pathobiology, sinusoidal cells' paracrine communication or cross-talk is being studied as a mechanism of disease but also as a remarkable target for treatment. The aim of this review is to gather current knowledge of intercellular signalling in the hepatic sinusoid during the progression of liver disease. We summarise studies developed in pre-clinical models of CLD, especially emphasizing those pathways characterized in human-based clinically relevant models. Finally, we describe pharmacological treatments targeting sinusoidal communication as promising options to treat CLD and its clinical complications.
Collapse
Affiliation(s)
- Albert Gibert-Ramos
- Liver Vascular Biology Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - María Andrés-Rozas
- Liver Vascular Biology Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Raül Pastó
- Liver Vascular Biology Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Pablo Alfaro-Retamero
- Liver Vascular Biology Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Sergi Guixé-Muntet
- Liver Vascular Biology Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Research Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
13
|
Lamba R, Paguntalan AM, Petrov PB, Naba A, Izzi V. MatriCom: a scRNA-Seq data mining tool to infer ECM-ECM and cell-ECM communication systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.10.627834. [PMID: 39763937 PMCID: PMC11702561 DOI: 10.1101/2024.12.10.627834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
The ECM is a complex and dynamic meshwork of proteins that forms the framework of all multicellular organisms. Protein interactions within the ECM are critical to building and remodeling the ECM meshwork, while interactions between ECM proteins and cell surface receptors are essential for the initiation of signal transduction and the orchestration of cellular behaviors. Here, we report the development of MatriCom, a web application (https://matrinet.shinyapps.io/matricom) and a companion R package (https://github.com/Izzilab/MatriCom), devised to mine scRNA-Seq datasets and infer communications between ECM components and between different cell populations and the ECM. To impute interactions from expression data, MatriCom relies on a unique database, MatriComDB, that includes over 25,000 curated interactions involving matrisome components, with data on 80% of the ~1,000 genes that compose the mammalian matrisome. MatriCom offers the option to query open-access datasets sourced from large sequencing efforts (Tabula Sapiens, The Human Protein Atlas, HuBMAP) or to process user-generated datasets. MatriCom is also tailored to account for the specific rules governing ECM protein interactions and offers options to customize the output through stringency filters. We illustrate the usability of MatriCom with the example of the human kidney matrisome communication network. Last, we demonstrate how the integration of 46 scRNA-Seq datasets led to the identification of both ubiquitous and tissue-specific ECM communication patterns. We envision that MatriCom will become a powerful resource to elucidate the roles of different cell populations in ECM-ECM and cell-ECM interactions and their dysregulations in the context of diseases such as cancer or fibrosis.
Collapse
Affiliation(s)
- Rijuta Lamba
- Faculty of Biochemistry and Molecular Medicine & Faculty of Medicine, BioIM Unit, University of Oulu, Oulu, FI-90014, Finland
| | - Asia M. Paguntalan
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Petar B. Petrov
- Infotech Institute, University of Oulu, Oulu, FI-90014, Finland
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Valerio Izzi
- Faculty of Biochemistry and Molecular Medicine & Faculty of Medicine, BioIM Unit, University of Oulu, Oulu, FI-90014, Finland
- Infotech Institute, University of Oulu, Oulu, FI-90014, Finland
| |
Collapse
|
14
|
Chan HW, Kuo DY, Shueng PW, Chuang HY. Visualizing the Tumor Microenvironment: Molecular Imaging Probes Target Extracellular Matrix, Vascular Networks, and Immunosuppressive Cells. Pharmaceuticals (Basel) 2024; 17:1663. [PMID: 39770505 PMCID: PMC11676442 DOI: 10.3390/ph17121663] [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: 11/20/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
The tumor microenvironment (TME) is a critical factor in cancer progression, driving tumor growth, immune evasion, therapeutic resistance, and metastasis. Understanding the dynamic interactions within the TME is essential for advancing cancer management. Molecular imaging provides a non-invasive, real-time, and longitudinal approach to studying the TME, with techniques such as positron emission tomography (PET), magnetic resonance imaging (MRI), and fluorescence imaging offering complementary strengths, including high sensitivity, spatial resolution, and intraoperative precision. Recent advances in imaging probe development have enhanced the ability to target and monitor specific components of the TME, facilitating early cancer diagnosis, therapeutic monitoring, and deeper insights into tumor biology. By integrating these innovations, molecular imaging offers transformative potential for precision oncology, improving diagnostic accuracy and treatment outcomes through a comprehensive assessment of TME dynamics.
Collapse
Affiliation(s)
- Hui-Wen Chan
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., Beitou Dist., Taipei City 112, Taiwan;
| | - Deng-Yu Kuo
- Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
| | - Pei-Wei Shueng
- Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei City 112, Taiwan
| | - Hui-Yen Chuang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., Beitou Dist., Taipei City 112, Taiwan;
| |
Collapse
|
15
|
Jarusintanakorn S, Mastrobattista E, Yamabhai M. Ectoine enhances recombinant antibody production in Chinese hamster ovary cells by promoting cell cycle arrest. N Biotechnol 2024; 83:56-65. [PMID: 38945523 DOI: 10.1016/j.nbt.2024.06.006] [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: 02/07/2024] [Revised: 06/08/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
Chinese hamster ovary (CHO) cells represent the most preferential host cell system for therapeutic monoclonal antibody (mAb) production. Enhancing mAb production in CHO cells can be achieved by adding chemical compounds that regulate the cell cycle and cell survival pathways. This study investigated the impact of ectoine supplementation on mAb production in CHO cells. The results showed that adding ectoine at a concentration of 100 mM on the 3rd day of cultivation improved mAb production by improving cell viability and extending the culture duration. RNA sequencing analysis revealed differentially expressed genes associated with cell cycle regulation, cell proliferation, and cellular homeostasis, in particular promotion of cell cycle arrest, which was then confirmed by flow cytometry analysis. Ectoine-treated CHO cells exhibited an increase in the number of cells in the G0/G1 phase. In addition, the cell diameter was also increased. These findings support the hypothesis that ectoine enhances mAb production in CHO cells through mechanisms involving cell cycle arrest and cellular homeostasis. Overall, this study highlights the potential of ectoine as a promising supplementation strategy to enhance mAb production not only in CHO cells but also in other cell lines.
Collapse
Affiliation(s)
- Salinthip Jarusintanakorn
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Department of Pharmaceutics, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, the Netherlands; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mahidol University, 447, Sri-Ayuthaya Road, Rachathevi, Bangkok 10400, Thailand
| | - Enrico Mastrobattista
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Department of Pharmaceutics, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, the Netherlands.
| | - Montarop Yamabhai
- Molecular Biotechnology Laboratory, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand.
| |
Collapse
|
16
|
Zhang L, Yan L, Fu X, Tao Z, Liu S, Li R, Wang T, Mao Y, Shang W, Gong M, Jia X, Wang F. PDK1 promotes epithelial ovarian cancer progression by upregulating BGN. Acta Biochim Biophys Sin (Shanghai) 2024; 57:712-726. [PMID: 39578715 DOI: 10.3724/abbs.2024186] [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: 11/24/2024] Open
Abstract
Pyruvate dehydrogenase kinase 1 (PDK1) is a new therapeutic target that is dysregulated in multiple tumors. This study aims to explore the potential role and regulatory mechanism of PDK1 in epithelial ovarian cancer (EOC). We detect PDK1 expression in EOC tissues and cells using qRT-PCR and western blot analysis, and the effects of PDK1 on EOC cell malignant behaviors are explored. RNA sequencing analyses are performed to explore the differentially expressed genes in PDK1-silenced EOC cells. Furthermore, tumor-bearing mouse models are established to assess the impacts of PDK1 and BGN on EOC tumor growth and metastasis in vivo. The results show that PDK1 is upregulated in EOC tissues and cell lines. Biglycan (BGN) is downregulated in PDK1-silenced EOC cells, and its expression is positively correlated with PDK1 levels in EOC tissues. PDK1 depletion inhibits EOC cell proliferation, migration and invasion. Mechanistically, PDK1 and BGN are colocalized in the cytoplasm of EOC cells and interact with each other. PDK1 positively regulates BGN expression by enhancing BGN mRNA stability. BGN overexpression partially reverses the anti-tumor effects of PDK1 depletion on EOC cell malignant behaviors. PDK1 has also been revealed to upregulate BGN to activate the NF-κB oncogenic pathway in EOC cells. Additionally, PDK1 accelerates tumor growth and metastasis by modulating BGN expression. In conclusion, PDK1 functions as an oncogene, facilitating EOC progression by upregulating BGN and activating the NF-κB pathway. These findings may provide valuable biomarkers for the diagnosis and treatment of EOC.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
- Department of Gynecology, the Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian 223300, China
| | - Lina Yan
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing 210004, China
| | - Xin Fu
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
- Clinical Laboratory, Baoshan People's Hospital, Baoshan 678000, China
| | - Ziqi Tao
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
| | - Shuna Liu
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
| | - Rong Li
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing 210004, China
| | - Ting Wang
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
| | - Yepeng Mao
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
| | - Wenwen Shang
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
| | - Mi Gong
- Department of Gynecology, the Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian 223300, China
| | - Xuemei Jia
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing 210004, China
| | - Fang Wang
- Department of Laboratory Medicine, the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing 210029, China
| |
Collapse
|
17
|
Abstract
The extracellular matrix (ECM) is the complex meshwork of proteins and glycans that forms the scaffold that surrounds and supports cells. It exerts key roles in all aspects of metazoan physiology, from conferring physical and mechanical properties on tissues and organs to modulating cellular processes such as proliferation, differentiation and migration. Understanding the mechanisms that orchestrate the assembly of the ECM scaffold is thus crucial to understand ECM functions in health and disease. This Review discusses novel insights into the compositional diversity of matrisome components and the mechanisms that lead to tissue-specific assemblies and architectures tailored to support specific functions. The Review then highlights recently discovered mechanisms, including post-translational modifications and metabolic pathways such as amino acid availability and the circadian clock, that modulate ECM secretion, assembly and remodelling in homeostasis and human diseases. Last, the Review explores the potential of 'matritherapies', that is, strategies to normalize ECM composition and architecture to achieve a therapeutic benefit.
Collapse
Affiliation(s)
- Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL, USA.
- University of Illinois Cancer Center, Chicago, IL, USA.
| |
Collapse
|
18
|
Han Q, Yang F, Chen M, Zhang M, Wang L, Wang H, Liu J, Cao Z. Coating Dormant Collagenase-Producing Bacteria with Metal-Anesthetic Networks for Precision Tumor Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407402. [PMID: 39291426 PMCID: PMC11558152 DOI: 10.1002/advs.202407402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/09/2024] [Indexed: 09/19/2024]
Abstract
Tumor malignancy highly depends on the stiffness of tumor matrix, which mainly consists of collagen. Despite the destruction of tumor matrix is conducive to tumor therapy, it causes the risk of tumor metastasis. Here, metal-anesthetic network-coated dormant collagenase-producing Clostridium is constructed to simultaneously destruct tumor matrix and inhibit tumor metastasis. By metal-phenolic complexation and π-π stacking interactions, a Fe3+-propofol network is formed on bacterial surface. Coated dormant Clostridium can selectively germinate and rapidly proliferate in tumor sites due to the ability of carried Fe3+ ions to promote bacterial multiplication. Intratumoral colonization of Clostridium produces sufficient collagenases to degrade tumor collagen mesh and the loaded propofol restrains tumor metastasis by inhibiting tumor cell migration and invasion. Meanwhile, the delivered Fe3+ ions are reduced to the Fe2+ form by intracellular glutathione, thereby inducing potent Fenton reaction to trigger lipid peroxidation and ultimate ferroptosis of tumor cells. In addition to a satisfactory safety, a single intratumoral injection of coated dormant Clostridium not only effectively retards the growth of established large primary tumors, but also significantly suppresses distal lung metastasis in two different orthotopic tumor models. This work proposes a strategy to develop advanced therapeutics for malignant tumor treatment and metastasis prevention.
Collapse
Affiliation(s)
- Qiuju Han
- Shanghai Key Laboratory for Nucleic Acid Chemistry and NanomedicineInstitute of Molecular MedicineState Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127China
- School of Chemistry and Chemical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Fengmin Yang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and NanomedicineInstitute of Molecular MedicineState Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127China
| | - Mian Chen
- Shanghai Key Laboratory for Nucleic Acid Chemistry and NanomedicineInstitute of Molecular MedicineState Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127China
- School of Chemistry and Chemical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Mengmeng Zhang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and NanomedicineInstitute of Molecular MedicineState Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127China
| | - Lu Wang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and NanomedicineInstitute of Molecular MedicineState Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127China
| | - Hongxia Wang
- Department of Medical OncologyFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and NanomedicineInstitute of Molecular MedicineState Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127China
- School of Chemistry and Chemical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Zhenping Cao
- Shanghai Key Laboratory for Nucleic Acid Chemistry and NanomedicineInstitute of Molecular MedicineState Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127China
| |
Collapse
|
19
|
Rodríguez-Quesada L, Ramírez-Sánchez K, Formosa-Dague C, Dague E, Sáenz-Arce G, García-González CA, Vásquez-Sancho F, Avendaño-Soto E, Starbird-Pérez R. Evaluation of Conductive Porous Biobased Composites with Tunable Mechanical Properties for Potential Biological Applications. ACS OMEGA 2024; 9:43426-43437. [PMID: 39493987 PMCID: PMC11525745 DOI: 10.1021/acsomega.4c04391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 11/05/2024]
Abstract
In this work, starch-based porous cryogels with controlled mechanical and electrical properties were prepared for tissue engineering applications. The starch cryogels were formulated using κ-carrageenan, poly(vinyl alcohol) (PVA), and styrylpyridinium-substituted PVA (SbQ) into the composite. A conductive cryogel was polymerized by chemical oxidation of 3,4-ethylenedioxythiophene (EDOT) using iron(III) p-toluenesulfonate as a strategy to control the electrical properties. The physical, thermal, and mechanical properties were evaluated for the obtained composites. Macro- and nanoscale results confirmed the capability of tuning the mechanical properties of the material by the addition of biopolymers in different contents. The presence of κ-carrageenan significantly increased the storage modulus and decreased the damping effect in the formulations. The presence of PVA showed a plasticizing effect in the formulations, confirmed by the buffering effect and an increase in storage modulus. PVA-SBQ improved the mechanical properties by cross-linking. The addition of PEDOT increased the mechanical and electrical properties of the obtained materials.
Collapse
Affiliation(s)
- Laria Rodríguez-Quesada
- Centro
de Investigación en Servicios Químicos y Microbiológicos
(CEQIATEC), Escuela de Química, Instituto
Tecnológico de Costa Rica, Cartago 159-7050, Costa Rica
- Master
Program in Medical Devices Engineering, Instituto Tecnológico de Costa Rica, Cartago 159-7050, Costa Rica
- Departamento
de Física, Facultad de Ciencias Exactas y Naturales, Universidad Nacional, Heredia 86-3000, Costa Rica
| | - Karla Ramírez-Sánchez
- Centro
de Investigación en Servicios Químicos y Microbiológicos
(CEQIATEC), Escuela de Química, Instituto
Tecnológico de Costa Rica, Cartago 159-7050, Costa Rica
| | | | - Etienne Dague
- LAAS-CNRS,
CNRS, Université de Toulouse, 31400Toulouse, France
| | - Giovanni Sáenz-Arce
- Departamento
de Física, Facultad de Ciencias Exactas y Naturales, Universidad Nacional, Heredia 86-3000, Costa Rica
- Centro de
Investigación en Óptica y Nanofísica, Departamento
de Física, Universidad de Murcia, 30100 Murcia, Spain
| | - Carlos A. García-González
- Departamento
de Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Universidad de Santiago de Compostela, 15782 Santiago
de Compostela, Spain
| | - Fabián Vásquez-Sancho
- Centro
de Investigación en Ciencia e Ingeniería de Materiales
(CICIMA), Universidad de Costa Rica, San José 11501-2060, Costa Rica
- School
of Physics, Universidad de Costa Rica, San José 11501-2060, Costa Rica
| | - Esteban Avendaño-Soto
- Centro
de Investigación en Ciencia e Ingeniería de Materiales
(CICIMA), Universidad de Costa Rica, San José 11501-2060, Costa Rica
- School
of Physics, Universidad de Costa Rica, San José 11501-2060, Costa Rica
| | - Ricardo Starbird-Pérez
- Centro
de Investigación en Servicios Químicos y Microbiológicos
(CEQIATEC), Escuela de Química, Instituto
Tecnológico de Costa Rica, Cartago 159-7050, Costa Rica
| |
Collapse
|
20
|
Bai Y, Lv YN, Zeng M, Yan ZY, Huang DY, Wen JZ, Lu HN, Zhang PY, Wang YF, Ban N, Yuan DW, Li S, Luan YX. E93 is indispensable for reproduction in ametabolous and hemimetabolous insects. Development 2024; 151:dev202518. [PMID: 38646855 DOI: 10.1242/dev.202518] [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: 11/07/2023] [Accepted: 04/15/2024] [Indexed: 04/23/2024]
Abstract
Ecdysone-induced protein 93 (E93), known as the 'adult-specifier' transcription factor in insects, triggers metamorphosis in both hemimetabolous and holometabolous insects. Although E93 is conserved in ametabolous insects, its spatiotemporal expression and physiological function remain poorly understood. In this study, we first discover that, in the ametabolous firebrat Thermobia domestica, the previtellogenic ovary exhibits cyclically high E93 expression, and E93 mRNA is broadly distributed in previtellogenic ovarioles. E93 homozygous mutant females of T. domestica exhibit severe fecundity deficiency due to impaired previtellogenic development of the ovarian follicles, likely because E93 induces the expression of genes involved in ECM (extracellular matrix)-receptor interactions during previtellogenesis. Moreover, we reveal that in the hemimetabolous cockroach Blattella germanica, E93 similarly promotes previtellogenic ovarian development. In addition, E93 is also essential for vitellogenesis that is necessary to guarantee ovarian maturation and promotes the vitellogenesis-previtellogenesis switch in the fat body of adult female cockroaches. Our findings deepen the understanding of the roles of E93 in controlling reproduction in insects, and of E93 expression and functional evolution, which are proposed to have made crucial contributions to the origin of insect metamorphosis.
Collapse
Affiliation(s)
- Yu Bai
- Guangdong Provincial Key Laboratory of Insect Development Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510000, China
- Guangmeiyuan R&D Center, Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, South China Normal University, Meizhou 514000, China
| | - Ya-Nan Lv
- Guangdong Provincial Key Laboratory of Insect Development Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510000, China
| | - Mei Zeng
- Guangdong Provincial Key Laboratory of Insect Development Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510000, China
| | - Zi-Yu Yan
- Guangdong Provincial Key Laboratory of Insect Development Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510000, China
| | - Dan-Yan Huang
- Guangdong Provincial Key Laboratory of Insect Development Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510000, China
| | - Jia-Zhen Wen
- Guangdong Provincial Key Laboratory of Insect Development Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510000, China
| | - Hu-Na Lu
- Guangdong Provincial Key Laboratory of Insect Development Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510000, China
| | - Pei-Yan Zhang
- Guangdong Provincial Key Laboratory of Insect Development Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510000, China
| | - Yi-Fan Wang
- Guangdong Provincial Key Laboratory of Insect Development Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510000, China
| | - Ning Ban
- Guangdong Provincial Key Laboratory of Insect Development Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510000, China
| | - Dong-Wei Yuan
- Guangdong Provincial Key Laboratory of Insect Development Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510000, China
| | - Sheng Li
- Guangdong Provincial Key Laboratory of Insect Development Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510000, China
- Guangmeiyuan R&D Center, Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, South China Normal University, Meizhou 514000, China
| | - Yun-Xia Luan
- Guangdong Provincial Key Laboratory of Insect Development Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510000, China
- Guangmeiyuan R&D Center, Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, South China Normal University, Meizhou 514000, China
| |
Collapse
|
21
|
Li W, Wei J, Cheng M, Liu M. Unveiling promising targets in gastric cancer therapy: A comprehensive review. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200857. [PMID: 39280587 PMCID: PMC11396074 DOI: 10.1016/j.omton.2024.200857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Gastric cancer (GC) poses a significant global health challenge, ranking fifth in incidence and third in mortality among all malignancies worldwide. Its insidious onset, aggressive growth, proclivity for metastasis, and limited treatment options have contributed to its high fatality rate. Traditional approaches for GC treatment primarily involve surgery and chemotherapy. However, there is growing interest in targeted therapies and immunotherapies. This comprehensive review highlights recent advancements in GC targeted therapy and immunotherapy. It delves into the mechanisms of various strategies, underscoring their potential in GC treatment. Additionally, the review evaluates the efficacy and safety of relevant clinical trials. Despite the benefits observed in numerous advanced GC patients with targeted therapies and immunotherapies, challenges persist. We discuss pertinent strategies to overcome these challenges, thereby providing a solid foundation for enhancing the clinical effectiveness of targeted therapies and immunotherapies.
Collapse
Affiliation(s)
- Wenke Li
- Gastric Cancer Center/Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Jing Wei
- Gastric Cancer Center/Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Mo Cheng
- Gastric Cancer Center/Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Ming Liu
- Gastric Cancer Center/Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| |
Collapse
|
22
|
Diallo A, Abbas M, Goodney G, Price E, Gaye A. Relationship between LDL-cholesterol, small and dense LDL particles, and mRNA expression in a cohort of African Americans. Am J Physiol Heart Circ Physiol 2024; 327:H690-H700. [PMID: 39028281 PMCID: PMC11901346 DOI: 10.1152/ajpheart.00332.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/20/2024]
Abstract
Understanding the characteristics and behavior of low-density lipoprotein (LDL) particles provides insights into the atherogenic risk of elevated LDL cholesterol in hypercholesterolemia, cardiovascular disease risks. Studying LDL particles helps identify specific LDL subtypes [e.g., small dense LDL particles (sdLDL)] that may be atherogenic and, consequently, potential targets for therapeutics. This study cohort consists of African Americans (AAs), a population disproportionately affected by cardiovascular diseases, thereby accentuating the importance of the investigation. Differential expression (DE) analysis was undertaken using a dataset comprising 17,947 protein-coding mRNAs from the whole blood transcriptomes of 416 samples to identify mRNAs associated with low-density lipoprotein cholesterol (LDL-C) and sdLDL plasma levels. Subsequently, mediation analyses were used to investigate the mediating role of sdLDL particles on the relationship between LDL-C levels and mRNA expression. Finally, pathway enrichment analysis was conducted to identify pathways involving mRNAs whose relationship with LDL-C is mediated by sdLDL. DE analysis revealed 1,048 and 284 mRNA transcripts differentially expressed by LDL-C and sdLDL levels, respectively. Mediation analysis revealed that the associations between LDL-C and 33 mRNAs were mediated by sdLDL. Of the 33 mRNAs mediated by sdLDL, 18 were mediated in both males and females. Nine mRNAs were mediated only in females, and six were mediated only in males. Pathway analysis showed that 33 mRNAs are involved in pathways associated with the immune system, inflammatory response, metabolism, and cardiovascular disease (CVD) risk. In conclusion, our study provides valuable insights into the complex interplay between LDL-C, sdLDL, and mRNA expression in a large sample of AAs. The results underscore the importance of incorporating sdLDL measurement alongside LDL-C levels to improve the accuracy of managing hypercholesterolemia and effectively stratify the risk of CVD. This is essential as differences in sdLDL modulate atherogenic properties at the transcriptome level.NEW & NOTEWORTHY The study investigated the interplay between LDL-C and mRNA expression, focusing on the role of small dense LDL (sdLDL) particles and sex differences. Differential expression analysis identified 1,048 and 284 mRNAs associated with LDL-C and sdLDL levels, respectively. Mediation analysis revealed that sdLDL mediates the relationship between LDL-C and 33 mRNAs involved in immune, inflammatory, and metabolic pathways. These findings highlight the significance of sdLDL in cardiovascular disease risk assessment and underscore sex-specific differences in lipid metabolism.
Collapse
Affiliation(s)
- Ana Diallo
- School of Nursing, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Malak Abbas
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Gabriel Goodney
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Elvin Price
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Amadou Gaye
- Department of Integrative Genomics and Epidemiology, School of Graduate Studies, Meharry Medical College, Nashville, Tennessee, United States
| |
Collapse
|
23
|
Fattahi AS, Jafari M, Farahavar G, Abolmaali SS, Tamaddon AM. Expanding horizons in cancer therapy by immunoconjugates targeting tumor microenvironments. Crit Rev Oncol Hematol 2024; 201:104437. [PMID: 38977144 DOI: 10.1016/j.critrevonc.2024.104437] [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: 03/20/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024] Open
Abstract
Immunoconjugates are promising molecules combining antibodies with different agents, such as toxins, drugs, radionuclides, or cytokines that primarily aim to target tumor cells. However, tumor microenvironment (TME), which comprises a complex network of various cells and molecular cues guiding tumor growth and progression, remains a major challenge for effective cancer therapy. Our review underscores the pivotal role of TME in cancer therapy with immunoconjugates, examining the intricate interactions with TME and recent advancements in TME-targeted immunoconjugates. We explore strategies for targeting TME components, utilizing diverse antibodies such as neutralizing, immunomodulatory, immune checkpoint inhibitors, immunostimulatory, and bispecific antibodies. Additionally, we discuss different immunoconjugates, elucidating their mechanisms of action, advantages, limitations, and applications in cancer immunotherapy. Furthermore, we highlight emerging technologies enhancing the safety and efficacy of immunoconjugates, such as antibody engineering, combination therapies, and nanotechnology. Finally, we summarize current advancements, perspectives, and future developments of TME-targeted immunoconjugates.
Collapse
Affiliation(s)
- Amir Saamaan Fattahi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mahboobeh Jafari
- Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| | - Ghazal Farahavar
- Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| |
Collapse
|
24
|
Gharib E, Robichaud GA. From Crypts to Cancer: A Holistic Perspective on Colorectal Carcinogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:9463. [PMID: 39273409 PMCID: PMC11395697 DOI: 10.3390/ijms25179463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Gilles A Robichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
25
|
Yang X, Gao J, Zhang T, Yang L, Jing C, Zhang Z, Tian D. Potential regulation and prognostic model of colorectal cancer with extracellular matrix genes. Heliyon 2024; 10:e36164. [PMID: 39247375 PMCID: PMC11379549 DOI: 10.1016/j.heliyon.2024.e36164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 08/10/2024] [Accepted: 08/11/2024] [Indexed: 09/10/2024] Open
Abstract
Background The tumor microenvironment (TME) of colorectal cancer (CRC) mainly comprises immune cells, stromal cells, tumor cells, as well as the extracellular matrix (ECM), which holds a pivotal position. The ECM affects cancer progression, but its regulatory roles and predictive potential in CRC are not fully understood. Methods We analyzed transcriptomes from CRC tumors and paired normal tissues to study ECM features. Up-regulated ECM components were examined through functional enrichment analysis, and single-cell sequencing identified cell types producing collagen, regulators, and secreted factors. Transcription factor analysis and cell-cell interaction studies were conducted to identify potential regulators of ECM changes. Additionally, a prognostic model was developed using TCGA-CRC cohort data, focusing on up-regulated core ECM components. Results Bulk RNA-seq analysis revealed a unique ECM pattern in tumors, with ECM abundance and composition significantly related to patient survival. Up-regulated ECM components were linked to various cancer-related pathways. Fibroblasts and non-fibroblasts interactions were crucial in forming the TME. Key potential regulators identified included ZNF469, PRRX2, TWIST1, and AEBP1. A prognostic model based on five ECM genes (THBS3, LAMB3, ESM1, SPRX, COL9A3) demonstrated strong associations with immune suppression and tumor angiogenesis. Conclusions The ECM components were involved in various cell-cell interactions and correlated with tumor development and poor survival outcomes. The ECM prognostic model components could be potential targets for novel therapeutic interventions in colorectal cancer.
Collapse
Affiliation(s)
- Xiaobao Yang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jiale Gao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Tianzhen Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Lu Yang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Chao Jing
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Dan Tian
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
26
|
Aydin H, Ozcelikkale A, Acar A. Exploiting Matrix Stiffness to Overcome Drug Resistance. ACS Biomater Sci Eng 2024; 10:4682-4700. [PMID: 38967485 PMCID: PMC11322920 DOI: 10.1021/acsbiomaterials.4c00445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
Drug resistance is arguably one of the biggest challenges facing cancer research today. Understanding the underlying mechanisms of drug resistance in tumor progression and metastasis are essential in developing better treatment modalities. Given the matrix stiffness affecting the mechanotransduction capabilities of cancer cells, characterization of the related signal transduction pathways can provide a better understanding for developing novel therapeutic strategies. In this review, we aimed to summarize the recent advancements in tumor matrix biology in parallel to therapeutic approaches targeting matrix stiffness and its consequences in cellular processes in tumor progression and metastasis. The cellular processes governed by signal transduction pathways and their aberrant activation may result in activating the epithelial-to-mesenchymal transition, cancer stemness, and autophagy, which can be attributed to drug resistance. Developing therapeutic strategies to target these cellular processes in cancer biology will offer novel therapeutic approaches to tailor better personalized treatment modalities for clinical studies.
Collapse
Affiliation(s)
- Hakan
Berk Aydin
- Department
of Biological Sciences, Middle East Technical
University, 06800, Ankara, Turkey
| | - Altug Ozcelikkale
- Department
of Mechanical Engineering, Middle East Technical
University, 06800, Ankara, Turkey
- Graduate
Program of Biomedical Engineering, Middle
East Technical University, 06800, Ankara, Turkey
| | - Ahmet Acar
- Department
of Biological Sciences, Middle East Technical
University, 06800, Ankara, Turkey
| |
Collapse
|
27
|
Gokyer D, Akinboro S, Zhou LT, Kleinhans A, Laronda MM, Duncan FE, Riley JK, Goldman KN, Babayev E. The oocyte microenvironment is altered in adolescents compared to oocyte donors. Hum Reprod Open 2024; 2024:hoae047. [PMID: 39211054 PMCID: PMC11361810 DOI: 10.1093/hropen/hoae047] [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: 03/01/2024] [Revised: 07/03/2024] [Indexed: 09/04/2024] Open
Abstract
STUDY QUESTION Do the molecular signatures of cumulus cells (CCs) and follicular fluid (FF) of adolescents undergoing fertility preservation differ from that of oocyte donors? SUMMARY ANSWER The microenvironment immediately surrounding the oocyte, including the CCs and FF, is altered in adolescents undergoing fertility preservation compared to oocyte donors. WHAT IS KNOWN ALREADY Adolescents experience a period of subfecundity following menarche. Recent evidence suggests that this may be at least partially due to increased oocyte aneuploidy. Reproductive juvenescence in mammals is associated with suboptimal oocyte quality. STUDY DESIGN SIZE DURATION This was a prospective cohort study. Adolescents (10-19 years old, n = 23) and oocyte donors (22-30 years old, n = 31) undergoing ovarian stimulation and oocyte retrieval at a single center between 1 November 2020 and 1 May 2023 were enrolled in this study. PARTICIPANTS/MATERIALS SETTING METHODS Patient demographics, ovarian stimulation, and oocyte retrieval outcomes were collected for all participants. The transcriptome of CCs associated with mature oocytes was compared between adolescents (10-19 years old, n = 19) and oocyte donors (22-30 years old, n = 19) using bulk RNA-sequencing. FF cytokine profiles (10-19 years old, n = 18 vs 25-30 years old, n = 16) were compared using cytokine arrays. MAIN RESULTS AND THE ROLE OF CHANCE RNA-seq analysis revealed 581 differentially expressed genes in CCs of adolescents relative to oocyte donors, with 361 genes downregulated and 220 upregulated. Genes enriched in pathways involved in cell cycle and cell division (e.g. GO: 1903047, P = 3.5 × 10-43; GO: 0051983, P = 4.1 × 10-30; GO: 0000281, P = 7.7 × 10-15; GO: 0044839, P = 5.3 × 10-13) were significantly downregulated, while genes enriched in several pathways involved in cellular and vesicle organization (e.g. GO: 0010256, P = 1.2 × 10-8; GO: 0051129, P = 6.8 × 10-7; GO: 0016050, P = 7.4 × 10-7; GO: 0051640, P = 8.1 × 10-7) were upregulated in CCs of adolescents compared to oocyte donors. The levels of nine cytokines were significantly increased in FF of adolescents compared to oocyte donors: IL-1 alpha (2-fold), IL-1 beta (1.7-fold), I-309 (2-fold), IL-15 (1.6-fold), TARC (1.9-fold), TPO (2.1-fold), IGFBP-4 (2-fold), IL-12-p40 (1.7-fold), and ENA-78 (1.4-fold). Interestingly, seven of these cytokines have known pro-inflammatory roles. Importantly, neither the CC transcriptomes nor FF cytokine profiles were different in adolescents with or without cancer. LARGE SCALE DATA Original high-throughput sequencing data have been deposited in Gene Expression Omnibus (GEO) database with the accession number GSE265995. LIMITATIONS REASONS FOR CAUTION This study aims to gain insights into the associated gamete quality by studying the immediate oocyte microenvironment. The direct study of oocytes is more challenging due to sample scarcity, as they are cryopreserved for future use, but would provide a more accurate assessment of oocyte reproductive potential. WIDER IMPLICATIONS OF THE FINDINGS Our findings have implications for the adolescent fertility preservation cycles. Understanding the expected quality of cryopreserved eggs in this age group will lead to better counseling of these patients about their reproductive potential and may help to determine the number of eggs that is recommended to be banked to achieve a reasonable chance of future live birth(s). STUDY FUNDING/COMPETING INTERESTS This project was supported by Friends of Prentice organization SP0061324 (M.M.L. and E.B.), Gesualdo Family Foundation (Research Scholar: M.M.L.), and NIH/NICHD K12 HD050121 (E.B.). The authors have declared that no conflict of interest exists.
Collapse
Affiliation(s)
- Dilan Gokyer
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sophia Akinboro
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Neuroscience, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA
| | - Luhan T Zhou
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Anna Kleinhans
- Department of Obstetrics and Gynecology, Northwestern Medicine Center for Fertility and Reproductive Medicine, Chicago, IL, USA
| | - Monica M Laronda
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Basic and Preclinical Science, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Joan K Riley
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Obstetrics and Gynecology, Northwestern Medicine Center for Fertility and Reproductive Medicine, Chicago, IL, USA
| | - Kara N Goldman
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Obstetrics and Gynecology, Northwestern Medicine Center for Fertility and Reproductive Medicine, Chicago, IL, USA
| | - Elnur Babayev
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Obstetrics and Gynecology, Northwestern Medicine Center for Fertility and Reproductive Medicine, Chicago, IL, USA
| |
Collapse
|
28
|
Urciuolo F, Imparato G, Netti PA. Engineering Cell Instructive Microenvironments for In Vitro Replication of Functional Barrier Organs. Adv Healthc Mater 2024; 13:e2400357. [PMID: 38695274 DOI: 10.1002/adhm.202400357] [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: 01/29/2024] [Revised: 04/02/2024] [Indexed: 05/14/2024]
Abstract
Multicellular organisms exhibit synergistic effects among their components, giving rise to emergent properties crucial for their genesis and overall functionality and survival. Morphogenesis involves and relies upon intricate and biunivocal interactions among cells and their environment, that is, the extracellular matrix (ECM). Cells secrete their own ECM, which in turn, regulates their morphogenetic program by controlling time and space presentation of matricellular signals. The ECM, once considered passive, is now recognized as an informative space where both biochemical and biophysical signals are tightly orchestrated. Replicating this sophisticated and highly interconnected informative media in a synthetic scaffold for tissue engineering is unattainable with current technology and this limits the capability to engineer functional human organs in vitro and in vivo. This review explores current limitations to in vitro organ morphogenesis, emphasizing the interplay of gene regulatory networks, mechanical factors, and tissue microenvironment cues. In vitro efforts to replicate biological processes for barrier organs such as the lung and intestine, are examined. The importance of maintaining cells within their native microenvironmental context is highlighted to accurately replicate organ-specific properties. The review underscores the necessity for microphysiological systems that faithfully reproduce cell-native interactions, for advancing the understanding of developmental disorders and disease progression.
Collapse
Affiliation(s)
- Francesco Urciuolo
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Piazzale Tecchio 80, Napoli, 80125, Italy
| | - Giorgia Imparato
- Centre for Advanced Biomaterials for Health Care (IIT@CRIB), Istituto Italiano di Tecnologia, L.go Barsanti e Matteucci, Napoli, 80125, Italy
| | - Paolo Antonio Netti
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) and Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Piazzale Tecchio 80, Napoli, 80125, Italy
- Centre for Advanced Biomaterials for Health Care (IIT@CRIB), Istituto Italiano di Tecnologia, L.go Barsanti e Matteucci, Napoli, 80125, Italy
| |
Collapse
|
29
|
Yau JNN, Yempala T, Muthuramalingam RPK, Giustarini G, Teng G, Ang WH, Gibson D, Adriani G, Pastorin G. Fluorescence-Guided Spatial Drug Screening in 3D Colorectal Cancer Spheroids. Adv Healthc Mater 2024; 13:e2400203. [PMID: 38774999 DOI: 10.1002/adhm.202400203] [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: 01/18/2024] [Revised: 05/06/2024] [Indexed: 06/04/2024]
Abstract
The limited recapitulation of critical cancer features in 2D cultures causes poor translatability of preclinical results from in vitro assays to in vivo tumor models. This contributes to slow drug development with a low success rate. 3D cultures better recapitulate the tumor microenvironment, enabling more accurate predictions when screening drug candidates and improving the development of chemotherapeutics. Platinum (Pt) (IV) compounds are promising prodrugs designed to reduce the severe systemic toxicity of widely used Food and Drug Administration (FDA)-approved Pt(II) drugs such as cisplatin. Here, this work presents spatiotemporal evaluations in 3D colorectal cancer (CRC) spheroids of mitochondria-targeting Pt(IV) complexes. CRC spheroids provide a greater pathophysiological recapitulation of in vivo tumors than 2D cultures by a marked upregulation of the ABCG2 chemoresistance marker expression. Furthermore, new 3D-staining protocols are introduced to evaluate the real-time decrease in mitochondria membrane potential (ΔΨ) in CRC spheroids, and a Pt-sensing dye to quantify the Pt mitochondrial accumulation. Finally, this work demonstrates a correlation between in vitro results and the efficacy of the compounds in vivo. Overall, the CRC spheroids represent a fast and cost-effective model to assess the behavior of Pt compounds in vitro and predict their translational potential in CRC treatment.
Collapse
Affiliation(s)
- Jia Ning Nicolette Yau
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, 117544, Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, 119077, Singapore
| | - Thirumal Yempala
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, 117544, Singapore
| | - Ram Pravin Kumar Muthuramalingam
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, 117544, Singapore
| | - Giulio Giustarini
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, 138648, Singapore
| | - Germaine Teng
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, 138648, Singapore
| | - Wee Han Ang
- Department of Chemistry, Faculty of Science, National University of Singapore, Singapore, 117544, Singapore
| | - Dan Gibson
- Institute for Drug Research, School of Pharmacy, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Giulia Adriani
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, 138648, Singapore
- Department of Biomedical Engineering, Faculty of Engineerin, National University of Singapore, Singapore, 117578, Singapore
| | - Giorgia Pastorin
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, 117544, Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, 119077, Singapore
| |
Collapse
|
30
|
Manucci AC, Fiore APZP, Genesi GL, Bruni-Cardoso A. The basement membrane regulates the cellular localization and the cytoplasmic interactome of Yes-Associated Protein (YAP) in mammary epithelial cells. J Cell Biochem 2024; 125:e30606. [PMID: 38779980 DOI: 10.1002/jcb.30606] [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: 10/23/2023] [Revised: 04/16/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
The Hippo pathway, a signaling cascade involved in the regulation of organ size and several other processes, acts as a conduit between extracellular matrix (ECM) cues and cellular responses. We asked whether the basement membrane (BM), a specialized ECM component known to induce quiescence and differentiation in mammary epithelial cells, would regulate the localization, activity, and interactome of YAP, a Hippo pathway effector. To address this question, we used a broad range of experimental approaches, including 2D and 3D cultures of both mouse and human mammary epithelial cells, as well as the developing mouse mammary gland. In contrast to malignant cells, nontumoral cells cultured with a reconstituted BM (rBM) displayed higher concentrations of YAP in the cytoplasm. Incidentally, when in the nucleus of rBM-treated cells, YAP resided preferentially at the nuclear periphery. In agreement with our cell culture experiments, YAP exhibited cytoplasmic predominance in ductal cells of developing mammary epithelia, where a denser BM is found. Conversely, terminal end bud (TEB) cells with a thinner BM displayed higher nucleus-to-cytoplasm ratios of YAP. Bioinformatic analysis revealed that genes regulated by YAP were overrepresented in the transcriptomes of microdissected TEBs. Consistently, mouse epithelial cells exposed to the rBM expressed lower levels of YAP-regulated genes, although the protein level of YAP and Hippo components were slightly altered by the treatment. Mass spectrometry analysis identified a differential set of proteins interacting with YAP in cytoplasmic fractions of mouse epithelial cells in the absence or presence of rBM. In untreated cells, YAP interactants were enriched in processes related to ubiquitin-mediated proteolysis, whereas in cells exposed to rBM YAP interactants were mainly key proteins related to amino acid, amino sugar, and carbohydrate metabolism. Collectively, we unraveled that the BM induces YAP translocation or retention in the cytoplasm of nontumoral epithelial cells and that in the cytoplasm YAP seems to undertake novel functions in metabolic pathways.
Collapse
Affiliation(s)
- Antonio Carlos Manucci
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | | | - Giovani Luiz Genesi
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Alexandre Bruni-Cardoso
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
31
|
Chi Z, Wang Q, Wang X, Li D, Tong L, Shi Y, Yang F, Guo Q, Zheng J, Chen Z. P4HA2 promotes proliferation, invasion, and metastasis through regulation of the PI3K/AKT signaling pathway in oral squamous cell carcinoma. Sci Rep 2024; 14:15023. [PMID: 38951593 PMCID: PMC11217378 DOI: 10.1038/s41598-024-64264-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 06/06/2024] [Indexed: 07/03/2024] Open
Abstract
Proline 4-hydroxylase 2 (P4HA2) is known for its hydroxylase activity, primarily involved in hydroxylating collagen precursors and promoting collagen cross-linking under physiological conditions. Although its overexpression influences a wide variety of malignant tumors' occurrence and development, its specific effects and mechanisms in oral squamous cell carcinoma (OSCC) remain unclear. This study focused on investigating the expression patterns, carcinogenic functions, and underlying mechanisms of P4HA2 in OSCC cells. Various databases, including TCGA, TIMER, UALCAN, GEPIA, and K-M plotter, along with paraffin-embedded samples, were used to ascertain P4HA2 expression in cancer and its correlation with clinicopathological features. P4HA2 knockdown and overexpression cell models were developed to assess its oncogenic roles and mechanisms. The results indicated that P4HA2 was overexpressed in OSCC and inversely correlated with patient survival. Knockdown of P4HA2 suppressed invasion, migration, and proliferation of OSCC cells both in vitro and in vivo, whereas overexpression of P4HA2 had the opposite effects. Mechanistically, the phosphorylation levels of the PI3K/AKT pathway were reduced following P4HA2 silencing. The study reveals that P4HA2 acts as a promising biomarker for predicting prognosis in OSCC and significantly affects metastasis, invasion, and proliferation of OSCC cells through the regulation of the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Zengpeng Chi
- Department of Stomatology, Qingdao Huangdao District Central Hospital, Qingdao, 266555, China
| | - Qimin Wang
- Department of Stomatology, Qingdao Hospital, University of Health and Rehabilitation Sciences(Qingdao Municipal Hospital), No.5 Donghai Middle Road, Qingdao, 266071, China
| | - Xin Wang
- Acupuncture and Tuina Department, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
| | - Dagang Li
- Department of Stomatology, Qingdao Huangdao District Central Hospital, Qingdao, 266555, China
| | - Lei Tong
- Department of Stomatology, Qingdao Hospital, University of Health and Rehabilitation Sciences(Qingdao Municipal Hospital), No.5 Donghai Middle Road, Qingdao, 266071, China
| | - Yu Shi
- Department of Stomatology, Shenzhen-Shanwei Central Hospital, Sun Yat-sen University, Shanwei, 516699, China
| | - Fang Yang
- Department of Stomatology, Qingdao Hospital, University of Health and Rehabilitation Sciences(Qingdao Municipal Hospital), No.5 Donghai Middle Road, Qingdao, 266071, China
| | - Qingyuan Guo
- Department of Stomatology, Qingdao Hospital, University of Health and Rehabilitation Sciences(Qingdao Municipal Hospital), No.5 Donghai Middle Road, Qingdao, 266071, China
| | - Jiawei Zheng
- Department of Oromaxillofacial Head and Neck Oncology, College of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Manufacturing Bureau Road, Huangpu District, Shanghai, 200011, China.
| | - Zhenggang Chen
- Institute of Stomatology, Binzhou Medical University, 256600, Binzhou, China.
- The affiliated Yantai Stomatological Hospital, Binzhou Medical University, 264000, Binzhou, China.
| |
Collapse
|
32
|
Shen XT, Chen ZC, Wang XY, Wang XF, Xie SZ, Zheng X, Yang LY, Lu L. Establishment of homotrimer collagen type I signature and its association with clinical manifestation and tertiary lymphoid structures formation in liver cancer. Heliyon 2024; 10:e31320. [PMID: 38841477 PMCID: PMC11152946 DOI: 10.1016/j.heliyon.2024.e31320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/08/2024] [Accepted: 05/14/2024] [Indexed: 06/07/2024] Open
Abstract
Background collagen type I is a fundamental composition of extracellular matrix. Typically it exists in the form of a heterotrimer, consisting of two α1 chains encoded by COL1A1 and one α2 chain encoded by COL1A2. However, in cancer a homotrimeric form of collagen type I comprises three α1 chains encoded by COL1A1 was founded. There is still a lack of transcriptional and histologic methods for detecting homotrimeric collagen type I. Furthermore, a comprehensive analysis of the pan-cancer distribution pattern and clinical relevance of homotrimeric collagen type I is conspicuously absent. Method Using transcriptional and immunoflourance method, we established homocol signature, which is able to transcriptionally and histologically detect homotrimeric collagen type I. We investigated the diagnostic and prognostic potential of homocol as a novel cancer biomarker in a pan-cancer cohort. Furthermore, we assessed its association with clinical manifestations in a liver cancer cohort undergoing treatment at our institute. Result Homotrimer Collagen Type I is predominantly expressed by cancer cells and is linked to several critical cancer hallmarks, particularly inflammatory response and proliferation. Survival analyses have indicated that a high Homocol expression is correlated with poor outcomes in most types of cancer studied. In terms of cancer detection, Homocol demonstrated strong performance in Receiver Operating Characteristic (ROC) analysis, with an Area Under Curve (AUC) of 0.83 for pan-cancer detection and between 0.72 and 0.99 for individual cancers.In cohorts undergoing PD1 treatment, we noted a higher presence of Homocol in the response group. In a Hepatocellular Carcinoma (HCC) clinical set, high Homocol expression was associated with an increased formation of intra-tumor tertiary lymphoid structures (TLS), larger tumor sizes, more advanced Barcelona Clinic Liver Cancer (BCLC) stages, higher microvascular invasion (MVI) grades, absence of a capsule, and an enriched para-tumor collagen presence. Conclusion our research has led to the development of a novel gene signature that facilitates the detection of Homotrimer Collagen Type I. This may greatly assist efforts in cancer detection, prognosis, treatment response prediction, and further research into Homotrimer Collagen Type I.
Collapse
Affiliation(s)
- Xiao-Tian Shen
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
- Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Zhen-Chao Chen
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
- Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Xiang-Yu Wang
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
- Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Xu-Feng Wang
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
- Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Sun-Zhe Xie
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
- Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Xin Zheng
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
- Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Lu-Yu Yang
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
- Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Lu Lu
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
- Cancer Metastasis Institute, Fudan University, Shanghai, China
| |
Collapse
|
33
|
Palmer JA, Rosenthal N, Teichmann SA, Litvinukova M. Revisiting Cardiac Biology in the Era of Single Cell and Spatial Omics. Circ Res 2024; 134:1681-1702. [PMID: 38843288 PMCID: PMC11149945 DOI: 10.1161/circresaha.124.323672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 06/09/2024]
Abstract
Throughout our lifetime, each beat of the heart requires the coordinated action of multiple cardiac cell types. Understanding cardiac cell biology, its intricate microenvironments, and the mechanisms that govern their function in health and disease are crucial to designing novel therapeutical and behavioral interventions. Recent advances in single-cell and spatial omics technologies have significantly propelled this understanding, offering novel insights into the cellular diversity and function and the complex interactions of cardiac tissue. This review provides a comprehensive overview of the cellular landscape of the heart, bridging the gap between suspension-based and emerging in situ approaches, focusing on the experimental and computational challenges, comparative analyses of mouse and human cardiac systems, and the rising contextualization of cardiac cells within their niches. As we explore the heart at this unprecedented resolution, integrating insights from both mouse and human studies will pave the way for novel diagnostic tools and therapeutic interventions, ultimately improving outcomes for patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Jack A. Palmer
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom (J.A.P., S.A.T.)
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus (J.A.P., S.A.T.), University of Cambridge, United Kingdom
| | - Nadia Rosenthal
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME (N.R.)
- National Heart and Lung Institute, Imperial College London, United Kingdom (N.R.)
| | - Sarah A. Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom (J.A.P., S.A.T.)
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus (J.A.P., S.A.T.), University of Cambridge, United Kingdom
- Theory of Condensed Matter Group, Department of Physics, Cavendish Laboratory (S.A.T.), University of Cambridge, United Kingdom
| | - Monika Litvinukova
- University Hospital Würzburg, Germany (M.L.)
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, Germany (M.L.)
- Helmholtz Pioneer Campus, Helmholtz Munich, Germany (M.L.)
| |
Collapse
|
34
|
Tung Y, Chen Y, Derr K, Wilson K, Song MJ, Ferrer M. A 3D Bioprinted Human Neurovascular Unit Model of Glioblastoma Tumor Growth. Adv Healthc Mater 2024; 13:e2302831. [PMID: 38394389 PMCID: PMC11176035 DOI: 10.1002/adhm.202302831] [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: 10/01/2023] [Revised: 01/29/2024] [Indexed: 02/25/2024]
Abstract
A 3D bioprinted neurovascular unit (NVU) model is developed to study glioblastoma (GBM) tumor growth in a brain-like microenvironment. The NVU model includes human primary astrocytes, pericytes and brain microvascular endothelial cells, and patient-derived glioblastoma cells (JHH-520) are used for this study. Fluorescence reporters are used with confocal high content imaging to quantitate real-time microvascular network formation and tumor growth. Extensive validation of the NVU-GBM model includes immunostaining for brain relevant cellular markers and extracellular matrix components; single cell RNA sequencing (scRNAseq) to establish physiologically relevant transcriptomics changes; and secretion of NVU and GBM-relevant cytokines. The scRNAseq reveals changes in gene expression and cytokines secretion associated with wound healing/angiogenesis, including the appearance of an endothelial mesenchymal transition cell population. The NVU-GBM model is used to test 18 chemotherapeutics and anti-cancer drugs to assess the pharmacological relevance of the model and robustness for high throughput screening.
Collapse
Affiliation(s)
- Yen‐Ting Tung
- National Center for Advancing Translational Sciences (NCATS)National Institutes of Health (NIH)RockvilleMD20850USA
| | - Yu‐Chi Chen
- National Center for Advancing Translational Sciences (NCATS)National Institutes of Health (NIH)RockvilleMD20850USA
| | - Kristy Derr
- National Center for Advancing Translational Sciences (NCATS)National Institutes of Health (NIH)RockvilleMD20850USA
| | - Kelli Wilson
- National Center for Advancing Translational Sciences (NCATS)National Institutes of Health (NIH)RockvilleMD20850USA
| | - Min Jae Song
- National Center for Advancing Translational Sciences (NCATS)National Institutes of Health (NIH)RockvilleMD20850USA
| | - Marc Ferrer
- National Center for Advancing Translational Sciences (NCATS)National Institutes of Health (NIH)RockvilleMD20850USA
| |
Collapse
|
35
|
Meder L, Orschel CI, Otto CJ, Koker M, Brägelmann J, Ercanoglu MS, Dähling S, Compes A, Selenz C, Nill M, Dietlein F, Florin A, Eich ML, Borchmann S, Odenthal M, Blazquez R, Hilberg F, Klein F, Hallek M, Büttner R, Reinhardt HC, Ullrich RT. Blocking the angiopoietin-2-dependent integrin β-1 signaling axis abrogates small cell lung cancer invasion and metastasis. JCI Insight 2024; 9:e166402. [PMID: 38775153 PMCID: PMC11141935 DOI: 10.1172/jci.insight.166402] [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: 10/18/2022] [Accepted: 04/05/2024] [Indexed: 06/02/2024] Open
Abstract
Small cell lung cancer (SCLC) is the most aggressive lung cancer entity with an extremely limited therapeutic outcome. Most patients are diagnosed at an extensive stage. However, the molecular mechanisms driving SCLC invasion and metastasis remain largely elusive. We used an autochthonous SCLC mouse model and matched samples from patients with primary and metastatic SCLC to investigate the molecular characteristics of tumor metastasis. We demonstrate that tumor cell invasion and liver metastasis in SCLC are triggered by an Angiopoietin-2 (ANG-2)/Integrin β-1-dependent pathway in tumor cells, mediated by focal adhesion kinase/Src kinase signaling. Strikingly, CRISPR-Cas9 KO of Integrin β-1 or blocking Integrin β-1 signaling by an anti-ANG-2 treatment abrogates liver metastasis formation in vivo. Interestingly, analysis of a unique collection of matched samples from patients with primary and metastatic SCLC confirmed a strong increase of Integrin β-1 in liver metastasis in comparison with the primary tumor. We further show that ANG-2 blockade combined with PD-1-targeted by anti-PD-1 treatment displays synergistic treatment effects in SCLC. Together, our data demonstrate a fundamental role of ANG-2/Integrin β-1 signaling in SCLC cells for tumor cell invasion and liver metastasis and provide a potentially new effective treatment strategy for patients with SCLC.
Collapse
Affiliation(s)
- Lydia Meder
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Faculty of Medicine, Department of Experimental Medicine 1, Erlangen, Germany
- Mildred Scheel School of Oncology and
| | - Charlotte Isabelle Orschel
- Mildred Scheel School of Oncology and
- Department I of Internal Medicine, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Christoph Julius Otto
- Mildred Scheel School of Oncology and
- Department I of Internal Medicine, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Mirjam Koker
- Department I of Internal Medicine, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Johannes Brägelmann
- Mildred Scheel School of Oncology and
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Department of Translational Genomics and
| | - Meryem S. Ercanoglu
- Institute of Virology, Laboratory of Experimental Immunology, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
| | - Sabrina Dähling
- Institute of Virology, Laboratory of Experimental Immunology, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
| | - Anik Compes
- Mildred Scheel School of Oncology and
- Department I of Internal Medicine, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Carolin Selenz
- Department I of Internal Medicine, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Marieke Nill
- Department I of Internal Medicine, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Felix Dietlein
- Department of Medical Oncology, Dana-Faber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Alexandra Florin
- Institute for Pathology, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
| | - Marie-Lisa Eich
- Institute for Pathology, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
| | - Sven Borchmann
- Department I of Internal Medicine, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
- Else Kröner Forschungskolleg Clonal Evolution in Cancer, University Hospital Cologne, Cologne, Germany
- German Hodgkin Study Group, Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
| | - Margarete Odenthal
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Institute for Pathology, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
| | - Raquel Blazquez
- University Hospital Regensburg, Department of Internal Medicine III, Hematology and Medical Oncology, Regensburg, Germany
| | - Frank Hilberg
- Department of Pharmacology, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Florian Klein
- Institute of Virology, Laboratory of Experimental Immunology, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
| | - Michael Hallek
- Department I of Internal Medicine, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
| | - Reinhard Büttner
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Institute for Pathology, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
| | - H. Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, German Cancer Consortium (DKTK), Essen, Germany
| | - Roland T. Ullrich
- Mildred Scheel School of Oncology and
- Department I of Internal Medicine, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Faculty of Medicine at the University Hospital Cologne, Cologne, Germany
| |
Collapse
|
36
|
Ahmad SS, Ahmad K, Lim JH, Shaikh S, Lee EJ, Choi I. Therapeutic applications of biological macromolecules and scaffolds for skeletal muscle regeneration: A review. Int J Biol Macromol 2024; 267:131411. [PMID: 38588841 DOI: 10.1016/j.ijbiomac.2024.131411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 04/10/2024]
Abstract
Skeletal muscle (SM) mass and strength maintenance are important requirements for human well-being. SM regeneration to repair minor injuries depends upon the myogenic activities of muscle satellite (stem) cells. However, losses of regenerative properties following volumetric muscle loss or severe trauma or due to congenital muscular abnormalities are not self-restorable, and thus, these conditions have major healthcare implications and pose clinical challenges. In this context, tissue engineering based on different types of biomaterials and scaffolds provides an encouraging means of structural and functional SM reconstruction. In particular, biomimetic (able to transmit biological signals) and several porous scaffolds are rapidly evolving. Several biological macromolecules/biomaterials (collagen, gelatin, alginate, chitosan, and fibrin etc.) are being widely used for SM regeneration. However, available alternatives for SM regeneration must be redesigned to make them more user-friendly and economically feasible with longer shelf lives. This review aimed to explore the biological aspects of SM regeneration and the roles played by several biological macromolecules and scaffolds in SM regeneration in cases of volumetric muscle loss.
Collapse
Affiliation(s)
- Syed Sayeed Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Jeong Ho Lim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Sibhghatulla Shaikh
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea.
| |
Collapse
|
37
|
Lin CC, Suen KM, Lidster J, Ladbury JE. The emerging role of receptor tyrosine kinase phase separation in cancer. Trends Cell Biol 2024; 34:371-379. [PMID: 37777392 DOI: 10.1016/j.tcb.2023.09.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/17/2023] [Accepted: 09/06/2023] [Indexed: 10/02/2023]
Abstract
Receptor tyrosine kinase (RTK)-mediated signal transduction is fundamental to cell function and drives important cellular outcomes which, when dysregulated, can lead to malignant tumour growth and metastasis. The initiation of signals from plasma membrane-bound RTKs is subjected to multiple regulatory mechanisms that control downstream effector protein recruitment and function. The high propensity of RTKs to condense via liquid-liquid phase separation (LLPS) into membraneless organelles with downstream effector proteins provides a further fundamental mechanism for signal regulation. Herein we highlight how this phenomenon contributes to cancer signalling and consider the potential impact of LLPS on outcomes for cancer patients.
Collapse
Affiliation(s)
- Chi-Chuan Lin
- School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK.
| | - Kin Man Suen
- School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Jessica Lidster
- School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - John E Ladbury
- School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
38
|
Olaizola-Rodrigo C, Castro-Abril H, Perisé-Badía I, Pancorbo L, Ochoa I, Monge R, Oliván S. Reducing Inert Materials for Optimal Cell-Cell and Cell-Matrix Interactions within Microphysiological Systems. Biomimetics (Basel) 2024; 9:262. [PMID: 38786472 PMCID: PMC11118140 DOI: 10.3390/biomimetics9050262] [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: 02/20/2024] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
In the pursuit of achieving a more realistic in vitro simulation of human biological tissues, microfluidics has emerged as a promising technology. Organ-on-a-chip (OoC) devices, a product of this technology, contain miniature tissues within microfluidic chips, aiming to closely mimic the in vivo environment. However, a notable drawback is the presence of inert material between compartments, hindering complete contact between biological tissues. Current membranes, often made of PDMS or plastic materials, prevent full interaction between cell types and nutrients. Furthermore, their non-physiological mechanical properties and composition may induce unexpected cell responses. Therefore, it is essential to minimize the contact area between cells and the inert materials while simultaneously maximizing the direct contact between cells and matrices in different compartments. The main objective of this work is to minimize inert materials within the microfluidic chip while preserving proper cellular distribution. Two microfluidic devices were designed, each with a specific focus on maximizing direct cell-matrix or cell-cell interactions. The first chip, designed to increase direct cell-cell interactions, incorporates a nylon mesh with regular pores of 150 microns. The second chip minimizes interference from inert materials, thereby aiming to increase direct cell-matrix contact. It features an inert membrane with optimized macropores of 1 mm of diameter for collagen hydrogel deposition. Biological validation of both devices has been conducted through the implementation of cell migration and cell-to-cell interaction assays, as well as the development of epithelia, from isolated cells or spheroids. This endeavor contributes to the advancement of microfluidic technology, aimed at enhancing the precision and biological relevance of in vitro simulations in pursuit of more biomimetic models.
Collapse
Affiliation(s)
- Claudia Olaizola-Rodrigo
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- BEOnChip S.L., 50018 Zaragoza, Spain; (L.P.); (R.M.)
| | - Héctor Castro-Abril
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- Laboratorio de Biomiméticos, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| | - Ismael Perisé-Badía
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Lara Pancorbo
- BEOnChip S.L., 50018 Zaragoza, Spain; (L.P.); (R.M.)
| | - Ignacio Ochoa
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rosa Monge
- BEOnChip S.L., 50018 Zaragoza, Spain; (L.P.); (R.M.)
| | - Sara Oliván
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (C.O.-R.); (H.C.-A.); (I.P.-B.); (S.O.)
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| |
Collapse
|
39
|
Gokyer D, Akinboro S, Zhou LT, Kleinhans A, Laronda MM, Duncan FE, Riley JK, Goldman KN, Babayev E. The oocyte microenvironment is altered in adolescents compared to oocyte donors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.588118. [PMID: 38617323 PMCID: PMC11014529 DOI: 10.1101/2024.04.04.588118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Study question Are the molecular signatures of cumulus cells (CCs) and follicular fluid (FF) of adolescents undergoing fertility preservation differ from that of reproductively adult oocyte donors? Summary answer The microenvironment immediately surrounding the oocyte, including the CCs and FF, is altered in adolescents undergoing fertility preservation compared to oocyte donors. What is known already Adolescents experience a period of subfecundity following menarche. Recent evidence suggests that this may be at least partially due to increased oocyte aneuploidy. Reproductive juvenescence in mammals is associated with suboptimal oocyte quality. Study design size duration This was a prospective cohort study. Adolescents (10-19 years old, N=23) and oocyte donors (22-30 years old, N=31) undergoing ovarian stimulation and oocyte retrieval at the Northwestern Fertility and Reproductive Medicine Center between November 1, 2020 and May 1, 2023 were enrolled in this study. Participants/materials setting methods Patient demographics, ovarian stimulation, and oocyte retrieval outcomes were collected for all participants. The transcriptome of CCs associated with mature oocytes was compared between adolescents (10-19 years old, n=19), and oocyte donors (22-30 years old, n=19) using bulk RNA-sequencing. FF cytokine profiles (10-19 years old, n=18 vs. 25-30 years old, n=16) were compared using cytokine arrays. Main results and the role of chance RNA-seq analysis revealed 581 differentially expressed genes (DEGs) in cumulus cells of adolescents relative to oocyte donors, with 361 genes downregulated and 220 upregulated. Genes enriched in pathways involved in cell cycle and cell division (e.g., GO:1903047, p= 3.5 × 10-43; GO:0051983, p= 4.1 × 10-30; GO:0000281, p= 7.7 × 10-15; GO:0044839, p= 5.3 × 10-13) were significantly downregulated, while genes enriched in several pathways involved in cellular and vesicle organization (e.g., GO:0010256, p= 1.2 × 10-8; GO:0051129, p= 6.8 × 10-7; GO:0016050, p= 7.4 × 10-7; GO:0051640, p= 8.1 × 10-7) were upregulated in CCs of adolescents compared to oocyte donors. The levels of 9 cytokines were significantly increased in FF of adolescents compared to oocyte donors: IL-1 alpha (2-fold), IL-1 beta (1.7-fold), I-309 (2-fold), IL-15 (1.6-fold), TARC (1.9-fold), TPO (2.1-fold), IGFBP-4 (2-fold), IL-12-p40 (1.7-fold) and ENA-78 (1.4-fold). Interestingly, 7 of these cytokines have known pro-inflammatory roles. Importantly, neither the CC transcriptomes or FF cytokine profiles were different in adolescents with or without cancer. Large scale data Original high-throughput sequencing data will be deposited in Gene Expression Omnibus (GEO) before publication, and the GEO accession number will be provided here. Limitations reasons for caution This study aims to gain insights into the associated gamete quality by studying the immediate oocyte microenvironment. The direct study of oocytes is more challenging due to sample scarcity, as they are cryopreserved for future use, but will provide a more accurate assessment of oocyte reproductive potential. Wider implications of the findings Understanding the underpinnings of altered immediate oocyte microenvironment of adolescent patients may provide insights into the reproductive potential of the associated gametes in the younger end of the age spectrum. This has implications for the fertility preservation cycles for very young patients. Study funding/competing interests This project was supported by Friends of Prentice organization SP0061324 (M.M.L and E.B.), Gesualdo Family Foundation (Research Scholar: M.M.L.), and NIH/NICHD K12 HD050121 (E.B.). The authors have declared that no conflict of interest exists.
Collapse
Affiliation(s)
- Dilan Gokyer
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
| | - Sophia Akinboro
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
- Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, 60208
| | - Luhan T. Zhou
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
| | - Anna Kleinhans
- Department of Obstetrics and Gynecology, Northwestern Medicine Center for Fertility and Reproductive Medicine, Chicago, IL, 60611
| | - Monica M. Laronda
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, 60611
| | - Francesca E. Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
| | - Joan K. Riley
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
- Department of Obstetrics and Gynecology, Northwestern Medicine Center for Fertility and Reproductive Medicine, Chicago, IL, 60611
| | - Kara N. Goldman
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
- Department of Obstetrics and Gynecology, Northwestern Medicine Center for Fertility and Reproductive Medicine, Chicago, IL, 60611
| | - Elnur Babayev
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
- Department of Obstetrics and Gynecology, Northwestern Medicine Center for Fertility and Reproductive Medicine, Chicago, IL, 60611
| |
Collapse
|
40
|
Walker M, Moore H, Ataya A, Pham A, Corris PA, Laubenbacher R, Bryant AJ. A perfectly imperfect engine: Utilizing the digital twin paradigm in pulmonary hypertension. Pulm Circ 2024; 14:e12392. [PMID: 38933181 PMCID: PMC11199193 DOI: 10.1002/pul2.12392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/08/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
Pulmonary hypertension (PH) is a severe medical condition with a number of treatment options, the majority of which are introduced without consideration of the underlying mechanisms driving it within an individual and thus a lack of tailored approach to treatment. The one exception is a patient presenting with apparent pulmonary arterial hypertension and shown to have vaso-responsive disease, whose clinical course and prognosis is significantly improved by high dose calcium channel blockers. PH is however characterized by a relative abundance of available data from patient cohorts, ranging from molecular data characterizing gene and protein expression in different tissues to physiological data at the organ level and clinical information. Integrating available data with mechanistic information at the different scales into computational models suggests an approach to a more personalized treatment of the disease using model-based optimization of interventions for individual patients. That is, constructing digital twins of the disease, customized to a patient, promises to be a key technology for personalized medicine, with the aim of optimizing use of existing treatments and developing novel interventions, such as new drugs. This article presents a perspective on this approach in the context of a review of existing computational models for different aspects of the disease, and it lays out a roadmap for a path to realizing it.
Collapse
Affiliation(s)
- Melody Walker
- University of Florida College of MedicineGainesvilleFloridaUSA
| | - Helen Moore
- University of Florida College of MedicineGainesvilleFloridaUSA
| | - Ali Ataya
- University of Florida College of MedicineGainesvilleFloridaUSA
| | - Ann Pham
- University of Florida College of MedicineGainesvilleFloridaUSA
| | - Paul A. Corris
- The Faculty of Medical Sciences Newcastle UniversityNewcastle upon TyneUK
| | | | | |
Collapse
|
41
|
Su H, Peng C, Liu Y. Regulation of ferroptosis by PI3K/Akt signaling pathway: a promising therapeutic axis in cancer. Front Cell Dev Biol 2024; 12:1372330. [PMID: 38562143 PMCID: PMC10982379 DOI: 10.3389/fcell.2024.1372330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
The global challenge posed by cancer, marked by rising incidence and mortality rates, underscores the urgency for innovative therapeutic approaches. The PI3K/Akt signaling pathway, frequently amplified in various cancers, is central in regulating essential cellular processes. Its dysregulation, often stemming from genetic mutations, significantly contributes to cancer initiation, progression, and resistance to therapy. Concurrently, ferroptosis, a recently discovered form of regulated cell death characterized by iron-dependent processes and lipid reactive oxygen species buildup, holds implications for diseases, including cancer. Exploring the interplay between the dysregulated PI3K/Akt pathway and ferroptosis unveils potential insights into the molecular mechanisms driving or inhibiting ferroptotic processes in cancer cells. Evidence suggests that inhibiting the PI3K/Akt pathway may sensitize cancer cells to ferroptosis induction, offering a promising strategy to overcome drug resistance. This review aims to provide a comprehensive exploration of this interplay, shedding light on the potential for disrupting the PI3K/Akt pathway to enhance ferroptosis as an alternative route for inducing cell death and improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Hua Su
- Xingyi People’s Hospital, Xinyi, China
| | - Chao Peng
- Xingyi People’s Hospital, Xinyi, China
| | - Yang Liu
- The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
42
|
Bianchi E, Ruggeri M, Del Favero E, Pisano R, Artusio F, Ricci C, Vigani B, Ferraretto A, Boselli C, Icaro Cornaglia A, Rossi S, Sandri G. Chondroitin sulfate and caseinophosphopeptides doped polyurethane-based highly porous 3D scaffolds for tendon-to-bone regeneration. Int J Pharm 2024; 652:123822. [PMID: 38242257 DOI: 10.1016/j.ijpharm.2024.123822] [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: 11/14/2023] [Revised: 01/16/2024] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
Tendon disorders are common injuries, which can be greatly debilitating as they are often accompanied by great pain and inflammation. Moreover, several problems are also related to the laceration of the tendon-to-bone interface (TBI), a specific region subjected to great mechanical stresses. The techniques used nowadays for the treatment of tendon and TBI injuries often involve surgery. However, one critical aspect of this procedure involves the elevated risk of fail due to the tissues weakening and the postoperative alterations of the normal joint mechanics. Synthetic polymers, such as thermoplastic polyurethane, are of special interest in the tissue engineering field as they allow the production of scaffolds with tunable elastic and mechanical properties, that could guarantee an effective support during the new tissue formation. Based on these premises, the aim of this work was the design and the development of highly porous 3D scaffolds based on thermoplastic polyurethane, and doped with chondroitin sulfate and caseinophosphopeptides, able to mimic the structural, biomechanical, and biochemical functions of the TBI. The obtained scaffolds were characterized by a homogeneous microporous structure, and by a porosity optimal for cell nutrition and migration. They were also characterized by remarkable mechanical properties, reaching values comparable to the ones of the native tendons. The scaffolds promoted the tenocyte adhesion and proliferation when caseinophosphopetides and chondroitin sulfate are present in the 3D structure. In particular, caseinophosphopeptides' optimal concentration for cell proliferation resulted 2.4 mg/mL. Finally, the systems evaluation in vivo demonstrated the scaffolds' safety, since they did not cause any inflammatory effect nor foreign body response, representing interesting platforms for the regeneration of injured TBI.
Collapse
Affiliation(s)
- Eleonora Bianchi
- Department of Drug Sciences, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Marco Ruggeri
- Department of Drug Sciences, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Elena Del Favero
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milano, Italy
| | - Roberto Pisano
- Department of Applied Science and Technology (DISAT), Polytechnic of Torino, Torino, Italy
| | - Fiora Artusio
- Department of Applied Science and Technology (DISAT), Polytechnic of Torino, Torino, Italy
| | - Caterina Ricci
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milano, Italy
| | - Barbara Vigani
- Department of Drug Sciences, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Anita Ferraretto
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, via Mangiagalli 31, Milan, Italy; Laboratory of Experimental Biochemistry & Molecular Biology, IRCCS Istituto Galeazzi-Sant'Ambrogio, Via Cristina Belgioioso 173, 20157 Milan, Italy
| | - Cinzia Boselli
- Department of Drug Sciences, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Antonia Icaro Cornaglia
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, via Forlanini 2, 27100 Pavia, Italy
| | - Silvia Rossi
- Department of Drug Sciences, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Giuseppina Sandri
- Department of Drug Sciences, University of Pavia, via Taramelli 12, 27100 Pavia, Italy.
| |
Collapse
|
43
|
Crossley RM, Johnson S, Tsingos E, Bell Z, Berardi M, Botticelli M, Braat QJS, Metzcar J, Ruscone M, Yin Y, Shuttleworth R. Modeling the extracellular matrix in cell migration and morphogenesis: a guide for the curious biologist. Front Cell Dev Biol 2024; 12:1354132. [PMID: 38495620 PMCID: PMC10940354 DOI: 10.3389/fcell.2024.1354132] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/12/2024] [Indexed: 03/19/2024] Open
Abstract
The extracellular matrix (ECM) is a highly complex structure through which biochemical and mechanical signals are transmitted. In processes of cell migration, the ECM also acts as a scaffold, providing structural support to cells as well as points of potential attachment. Although the ECM is a well-studied structure, its role in many biological processes remains difficult to investigate comprehensively due to its complexity and structural variation within an organism. In tandem with experiments, mathematical models are helpful in refining and testing hypotheses, generating predictions, and exploring conditions outside the scope of experiments. Such models can be combined and calibrated with in vivo and in vitro data to identify critical cell-ECM interactions that drive developmental and homeostatic processes, or the progression of diseases. In this review, we focus on mathematical and computational models of the ECM in processes such as cell migration including cancer metastasis, and in tissue structure and morphogenesis. By highlighting the predictive power of these models, we aim to help bridge the gap between experimental and computational approaches to studying the ECM and to provide guidance on selecting an appropriate model framework to complement corresponding experimental studies.
Collapse
Affiliation(s)
- Rebecca M. Crossley
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Samuel Johnson
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Erika Tsingos
- Computational Developmental Biology Group, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, Netherlands
| | - Zoe Bell
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Massimiliano Berardi
- LaserLab, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Optics11 life, Amsterdam, Netherlands
| | | | - Quirine J. S. Braat
- Department of Applied Physics and Science Education, Eindhoven University of Technology, Eindhoven, Netherlands
| | - John Metzcar
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, United States
- Department of Informatics, Indiana University, Bloomington, IN, United States
| | | | - Yuan Yin
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
44
|
Macdonald JK, Mehta AS, Drake RR, Angel PM. Molecular analysis of the extracellular microenvironment: from form to function. FEBS Lett 2024; 598:602-620. [PMID: 38509768 PMCID: PMC11049795 DOI: 10.1002/1873-3468.14852] [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: 11/29/2023] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024]
Abstract
The extracellular matrix (ECM) proteome represents an important component of the tissue microenvironment that controls chemical flux and induces cell signaling through encoded structure. The analysis of the ECM represents an analytical challenge through high levels of post-translational modifications, protease-resistant structures, and crosslinked, insoluble proteins. This review provides a comprehensive overview of the analytical challenges involved in addressing the complexities of spatially profiling the extracellular matrix proteome. A synopsis of the process of synthesizing the ECM structure, detailing inherent chemical complexity, is included to present the scope of the analytical challenge. Current chromatographic and spatial techniques addressing these challenges are detailed. Capabilities for multimodal multiplexing with cellular populations are discussed with a perspective on developing a holistic view of disease processes that includes both the cellular and extracellular microenvironment.
Collapse
Affiliation(s)
- Jade K Macdonald
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC
| | - Anand S Mehta
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC
| | - Richard R Drake
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC
| | - Peggi M. Angel
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
45
|
Mohammadi A, Koruji M, Azami M, Shabani R, Mohandesnezhad S, Bashiri Z, Asgari H. Polycaprolactone/Testicular Extracellular Matrix/Graphene Oxide-Based Electrospun Tubular Scaffolds for Reproductive Medicine: Biomimetic Architecture of Seminiferous Tubules. Macromol Biosci 2024; 24:e2300342. [PMID: 37729950 DOI: 10.1002/mabi.202300342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/06/2023] [Indexed: 09/22/2023]
Abstract
Numerous scaffolds are developed in the field of testicular bioengineering. However, effectively replicating the spatial characteristics of native tissue, poses a challenge in maintaining the requisite cellular arrangement essential for spermatogenesis. In order to mimic the structural properties of seminiferous tubules, the objective is to fabricate a biocompatible tubular scaffold. Following the decellularization process of the testicular tissue, validation of cellular remnants' elimination from the specimens is conducted using 4',6-diamidino-2-phenylindole staining, hematoxylin and eosin staining, and DNA content analysis. The presence of extracellular matrix (ECM) components is confirmed through Alcian blue, Orcein, and Masson's trichrome staining techniques. The electrospinning technique is employed to synthesize the scaffolds using polycaprolactone (PCL), extracted ECM, and varying concentrations of graphene oxide (GO) (0.5%, 1%, and 2%). Subsequently, comprehensive evaluations are performed to assess the properties of the synthetic scaffolds. These evaluations encompass Fourier-transform infrared spectroscopy, scanning electron microscopy imaging, scaffold degradation testing, mechanical behavior analysis, methylthiazolyldiphenyl-tetrazolium bromide assay, and in vivo biocompatibility assessment. The PCL/decellularized extracellular matrix with 0.5% GO formulation exhibits superior fiber morphology and enhanced mechanical properties, and outperforms other groups in terms of in vitro biocompatibility. Consequently, these scaffolds present a viable option for implementation in "in vitro spermatogenesis" procedures, holding promise for future sperm production from spermatogonial cells.
Collapse
Affiliation(s)
- Amirhossein Mohammadi
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, 88770048, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1449614525, Iran
| | - Morteza Koruji
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, 88770048, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1449614525, Iran
| | - Mahmoud Azami
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
- Joint Reconstruction Research Center (JRRC), Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Ronak Shabani
- Reproductive Sciences and Technology Research Center, Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Sanam Mohandesnezhad
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Zahra Bashiri
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1449614525, Iran
- Endometrium and Endometriosis Research Center, Hamadan University of Medical Sciences, Hamadan, 6517789971, Iran
- Omid Fertility & Infertility Clinic, Hamedan, 6516796198, Iran
| | - Hamidreza Asgari
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, 88770048, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, 1449614525, Iran
| |
Collapse
|
46
|
Du W, Xia X, Hu F, Yu J. Extracellular matrix remodeling in the tumor immunity. Front Immunol 2024; 14:1340634. [PMID: 38332915 PMCID: PMC10850336 DOI: 10.3389/fimmu.2023.1340634] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 12/28/2023] [Indexed: 02/10/2024] Open
Abstract
The extracellular matrix (ECM) is a significant constituent of tumors, fulfilling various essential functions such as providing mechanical support, influencing the microenvironment, and serving as a reservoir for signaling molecules. The abundance and degree of cross-linking of ECM components are critical determinants of tissue stiffness. In the process of tumorigenesis, the interaction between ECM and immune cells within the tumor microenvironment (TME) frequently leads to ECM stiffness, thereby disrupting normal mechanotransduction and promoting malignant progression. Therefore, acquiring a thorough comprehension of the dysregulation of ECM within the TME would significantly aid in the identification of potential therapeutic targets for cancer treatment. In this regard, we have compiled a comprehensive summary encompassing the following aspects: (1) the principal components of ECM and their roles in malignant conditions; (2) the intricate interaction between ECM and immune cells within the TME; and (3) the pivotal regulators governing the onco-immune response in ECM.
Collapse
Affiliation(s)
- Wei Du
- Department of Targeting Therapy and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Xueming Xia
- Division of Head & Neck Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Fan Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jiayun Yu
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
47
|
Li J, Yue Z, Tang M, Wang W, Sun Y, Sun T, Chen C. Strategies to Reverse Hypoxic Tumor Microenvironment for Enhanced Sonodynamic Therapy. Adv Healthc Mater 2024; 13:e2302028. [PMID: 37672732 DOI: 10.1002/adhm.202302028] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/26/2023] [Indexed: 09/08/2023]
Abstract
Sonodynamic therapy (SDT) has emerged as a highly effective modality for the treatment of malignant tumors owing to its powerful penetration ability, noninvasiveness, site-confined irradiation, and excellent therapeutic efficacy. However, the traditional SDT, which relies on oxygen availability, often fails to generate a satisfactory level of reactive oxygen species because of the widespread issue of hypoxia in the tumor microenvironment of solid tumors. To address this challenge, various approaches are developed to alleviate hypoxia and improve the efficiency of SDT. These strategies aim to either increase oxygen supply or prevent hypoxia exacerbation, thereby enhancing the effectiveness of SDT. In view of this, the current review provides an overview of these strategies and their underlying principles, focusing on the circulation of oxygen from consumption to external supply. The detailed research examples conducted using these strategies in combination with SDT are also discussed. Additionally, this review highlights the future prospects and challenges of the hypoxia-alleviated SDT, along with the key considerations for future clinical applications. These considerations include the development of efficient oxygen delivery systems, the accurate methods for hypoxia detection, and the exploration of combination therapies to optimize SDT outcomes.
Collapse
Affiliation(s)
- Jialun Li
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Zhengya Yue
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Minglu Tang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Wenxin Wang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Yuan Sun
- Center of Pharmaceutical Engineering and Technology, Harbin University of Commerce, Harbin, 150076, P. R. China
| | - Tiedong Sun
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Chunxia Chen
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| |
Collapse
|
48
|
Ghazaly MM, Lee GE, Shaipulah NFM, Ibrahim YS, Mohammed A. Marine polychaete Namalycastis sp. extracts enhance proliferation and regeneration of mice 3T3 fibroblast and MCR-5 human fibroblast cells. Saudi J Biol Sci 2024; 31:103883. [PMID: 38125737 PMCID: PMC10730865 DOI: 10.1016/j.sjbs.2023.103883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/16/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
The Nereidid worm is a marine polychaete commonly found near the Nipa palm (Nypa fructicans) along the mangrove estuary. Recently, many usages have been documented for this polychaete family. Nevertheless, the true potentials of these marine worms, especially Namalycastis sp., from the medical perspective are still unknown. The current study investigated the cytotoxicity effect of Namalycastis sp. crude extracts on mice 3T3 fibroblast cells and human lung MRC-5 fibroblast cells. Thirteen concentrations (2, 4, 8, 16, 31, 63 µg/mL and 0.1, 0.3, 0.5, 1, 2, 4, 8 mg/mL) of the extracts were used as a treatment for 24 h, and cell viability was measured via the MTT assay. None of the 13 concentrations of Namalycastis sp. crude extracts showed cytotoxicity effects on the cell types investigated. However, based on the live images captured by the IncuCyte™ imaging system, the cells treated with Namalycastis sp. crude extracts showed an increased proliferation and growth rate in less than 10 h Furthermore, the extract concentration of 8 µg/mL induced the highest cell proliferation rate whereas 8 mg/mL led to the lowest cell proliferation rate following the treatment. Overall, Namalycastis sp. crude extracts were non-toxic on mice and human cells within the tested concentrations set. Still, it increased cell viability and proliferation compared with the control. This finding could pave the way for an alternative therapeutic strategy to treat debilitating disorders such as ageing, cardiovascular diseases, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Maziah M. Ghazaly
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
| | - Gaik Ee Lee
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
- Institute of Tropical Biodiversity and Sustainable Development, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
| | - Nur Fariza M. Shaipulah
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
| | - Yusof S. Ibrahim
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
| | - Arifullah Mohammed
- Department of Agriculture Science, Faculty of Agro-Based Industry, Universiti Malaysia Kelantan, Jeli 17600, Kelantan, Malaysia
| |
Collapse
|
49
|
Cifuentes SJ, Domenech M. Heparin-collagen I bilayers stimulate FAK/ERK½ signaling via α2β1 integrin to support the growth and anti-inflammatory potency of mesenchymal stromal cells. J Biomed Mater Res A 2024; 112:65-81. [PMID: 37723658 DOI: 10.1002/jbm.a.37614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/25/2023] [Accepted: 09/01/2023] [Indexed: 09/20/2023]
Abstract
Understanding mesenchymal stromal cells (MSCs) growth mechanisms in response to surface chemistries is essential to optimize culture methods for high-quality and robust cell yields in cell manufacturing applications. Heparin (HEP) and collagen 1 (COL) substrates have been reported to enhance cell adhesion, growth, viability, and secretory potential in MSCs. However, the biomolecular mechanisms underlying the benefits of combined HEP/COL substrates are unknown. This work used HEP/COL bilayered surfaces to investigate the role of integrin-HEP interactions in the advantages of MSC culture. The layer-by-layer approach (LbL) was used to create HEP/COL bilayers, which were made up of stacks of 8 and 9 layers that combined HEP and COL in an alternate arrangement. Surface spectroscopic investigations and laser scanning microscopy evaluations verified the biochemical fingerprint of each component and a total stacked bilayer thickness of roughly 150 nm. Cell growth and apoptosis in response to IC50 and IC75 levels of BTT-3033 and Cilengitide, α2β1 and αvβ3 integrin inhibitors respectively, were evaluated on HEP/COL coated surfaces using two bone marrow-derived MSC donors. While integrin activity did not affect cell growth rates, it significantly affected cell adhesion and apoptosis on HEP/COL surfaces. HEP-ending HEP/COL surfaces significantly increased FAK-ERK½ phosphorylation and endogenous cell COL deposition compared to COL, COL-ending HEP/COL and uncoated surfaces. BTT-3033 but not Cilengitide treatment markedly affected FAK-ERK½ activity levels on HEP-ending HEP/COL surfaces supporting a major role for α2β1 activity. BTT-3033 treatment on HEP-ending bilayers reduced MSC-mediated macrophage inhibitory activity and altered the cytokine profile of co-cultures. Overall, this study supports a novel role for HEP in regulating the survival and potency of MSCs via enhancing the α2β1-FAK-ERK½ signaling mechanism.
Collapse
Affiliation(s)
- Said J Cifuentes
- Bioengineering Graduate Program, University of Puerto Rico Mayaguez, Mayaguez, Puerto Rico, USA
| | - Maribella Domenech
- Bioengineering Graduate Program, University of Puerto Rico Mayaguez, Mayaguez, Puerto Rico, USA
- Department of Chemical Engineering, University of Puerto Rico Mayaguez, Mayaguez, Puerto Rico, USA
| |
Collapse
|
50
|
Czarnowska E, Ratajska A, Jankowska-Steifer E, Flaht-Zabost A, Niderla-Bielińska J. Extracellular matrix molecules associated with lymphatic vessels in health and disease. Histol Histopathol 2024; 39:13-34. [PMID: 37350542 DOI: 10.14670/hh-18-641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2023]
Abstract
Lymphatic vessels (LyVs), responsible for fluid, solute, and immune cell homeostasis in the body, are closely associated with the adjacent extracellular matrix (ECM) molecules whose structural and functional impact on LyVs is currently more appreciated, albeit not entirely elucidated. These molecules, serving as a platform for various connective tissue cell activities and affecting LyV biology should be considered also as an integral part of the lymphatic system. Any alterations and changes in ECM molecules over the course of disease impair the function and structure of the LyV network. Remodeling of LyV cells, which are components of lymphatic vessel walls, also triggers alterations in ECM molecules and interstitial tissue composition. Therefore, in this review we aimed to present the current knowledge on ECM in tissues and particularly on molecules surrounding lymphatics in normal conditions and in disease.
Collapse
Affiliation(s)
| | - Anna Ratajska
- Department of Pathology, Medical University of Warsaw, Warsaw, Poland.
| | - Ewa Jankowska-Steifer
- Department of Histology and Embryology, Medical University of Warsaw, Warsaw, Poland
| | | | | |
Collapse
|