1
|
Forjaz A, Kramer D, Shen Y, Bea H, Tsapatsis M, Ping J, Queiroga V, San KH, Joshi S, Grubel C, Beery ML, Kusmartseva I, Atkinson M, Kiemen AL, Wirtz D. Integration of nuclear morphology and 3D imaging to profile cellular neighborhoods. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646356. [PMID: 40236208 PMCID: PMC11996441 DOI: 10.1101/2025.03.31.646356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Nuclear morphology is an indicator of cellular function and disease states, as changes in nuclear size, shape, and texture often reflect underlying disease-related genetic, epigenetic, and microenvironmental alterations. For disease diagnosis, nuclear segmentation performed in 2D hematoxylin and eosin (H&E)-stained tissue sections has long represented the gold standard. However, recent advances in three-dimensional (3D) histology, which provide a more biologically accurate representation of the spatial heterogeneity of human microanatomy, has led to improved understandings of disease pathology. Yet challenges remain in the development of scalable and computationally efficient pipelines for extracting and interpreting nuclear features in 3D space. 2D histology neglects crucial spatial information, such as 3D connectivity, morphology, and rare events missed by sparser sampling. Here, through extension of the CODA platform, we integrate 3D imaging with nuclear segmentation to analyze nuclear morphological features in human tissue. Analysis of 3D tissue microenvironments uncovered critical changes in 3D morphometric heterogeneity. Additionally, it enables the spatial characterization of immune cell distribution in relation to tissue structures, such as variations in leukocyte density near pancreatic ducts and blood vessels of different sizes. This approach provides a more comprehensive understanding of tissue and nuclear structures, revealing spatial patterns and interactions that are critical for disease progression.
Collapse
|
2
|
Seledtsova GV, Seledtsov VI, Dorzhieva AB, Ivanova IP, Khabalova TS, Darinskas A, von Delwig AA. Tumour-Derived, Extracellular Microvesicles in the Treatment of Acute Renal Failure: An Experimental Study. Med Sci (Basel) 2025; 13:35. [PMID: 40265382 PMCID: PMC12015901 DOI: 10.3390/medsci13020035] [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: 01/25/2025] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/24/2025] Open
Abstract
Background/Objectives: This investigation compared the therapeutic efficacy of extracellular microvesicles (MVs) derived from murine L929 sarcoma cells and murine mesenchymal stem cells (MSCs). Methods: A mouse model of acute kidney injury (AKI) was used. Results: Both MVs from L929 cells (L929-MVs) and MSCs (MSC-MVs), unlike those obtained from murine peripheral blood mononuclear cells (PBMCs), enhanced survival rates in AKI mice and significantly improved kidney function. This was indicated by decreased levels of urine albumin and serum creatinine. Furthermore, treatment with L929-MVs and MSC-MVs elevated the proportions of CD4+CD25+FOXP3+ regulatory T cells while reducing the presence of pro-inflammatory CD4+CD44+ T cells in the spleens of AKI mice. Conclusions: the results highlight the potential of tumour-derived MVs to facilitate organ repair and exert cytoprotective immunomodulatory effects.
Collapse
Affiliation(s)
- Galina V. Seledtsova
- Institute for Fundamental and Clinical Immunology, 630099 Novosibirsk, Russia; (G.V.S.); (A.B.D.); (I.P.I.); (T.S.K.); (A.A.v.D.)
| | | | - Ayana B. Dorzhieva
- Institute for Fundamental and Clinical Immunology, 630099 Novosibirsk, Russia; (G.V.S.); (A.B.D.); (I.P.I.); (T.S.K.); (A.A.v.D.)
| | - Irina P. Ivanova
- Institute for Fundamental and Clinical Immunology, 630099 Novosibirsk, Russia; (G.V.S.); (A.B.D.); (I.P.I.); (T.S.K.); (A.A.v.D.)
| | - Tatiana S. Khabalova
- Institute for Fundamental and Clinical Immunology, 630099 Novosibirsk, Russia; (G.V.S.); (A.B.D.); (I.P.I.); (T.S.K.); (A.A.v.D.)
| | | | - Alexei A. von Delwig
- Institute for Fundamental and Clinical Immunology, 630099 Novosibirsk, Russia; (G.V.S.); (A.B.D.); (I.P.I.); (T.S.K.); (A.A.v.D.)
| |
Collapse
|
3
|
Dong Y, Qian S, Wang X, Zhang W, Lu W, Qu J, Cui M, Chen L, Zhao Y, Gao Y, Giomo M, Urciuolo A, Feng J, Zheng Y, Jiang B, Shen R, Zhu X, Elvassore N. In situ tailored confining microenvironment for lung cancer spheroids. Mater Today Bio 2025; 31:101602. [PMID: 40070872 PMCID: PMC11894329 DOI: 10.1016/j.mtbio.2025.101602] [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: 11/27/2024] [Revised: 02/15/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
The mechanical properties and physical confinement of the extracellular matrix (ECM) are crucial roles in regulating tumor growth and progression. Extensive efforts have been dedicated to replicating the physical characteristics of tumor tissue by developing two-dimensional (2D) and three-dimensional (3D) in vitro models. However, it remains a significant challenge to modulate the local microenvironment around the specific cells according to the culture progress. In this study, we develop a 3D culture platform for multicellular lung cancer spheroids using a gelatin-based hydrogel with adjustable density and stiffness. Then, by utilizing a two-photon mediated bioprinting technique, we construct 3D confining microstructures with micrometer accuracy to enclose the selected spheroids within the hydrogel matrix. Diverse transcriptional profilings of cells are observed in response to the increased ECM density and stiffness compared to the additional confining stress. In addition, changed confining stress can regulate the tumor cells with contrary impacts on the cell cycle-related pathways. Our model not only allows for modifications to the mechanical microenvironment of the overall matrix but also facilitates localized adjustments throughout the culture evolution. This approach serves as a valuable tool for investigating tumor progression and understanding cell-ECM interactions.
Collapse
Affiliation(s)
- Yixiao Dong
- Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, 201210, China
- Shanghai Academy of Sciences & Technology Institute of Model Animals Transformation, Shanghai, 201203, China
| | - Shuyi Qian
- Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Xuechun Wang
- Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Wang Zhang
- Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Weisheng Lu
- Shanghai Laboratory Animal Research Center, Shanghai, 201203, China
| | - Ju Qu
- Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Meihua Cui
- Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, 201210, China
| | - Linzhi Chen
- Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yingshuai Zhao
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yuehua Gao
- Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Monica Giomo
- Department of Industrial Engineering, University of Padova, Padova, 35131, Italy
| | - Anna Urciuolo
- Department of Industrial Engineering, University of Padova, Padova, 35131, Italy
- Department of Molecular Medicine, University of Padova, Padova, 35127, Italy
| | - Jian Feng
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, 200030, China
| | - Yijun Zheng
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Biao Jiang
- Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, 201210, China
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Ruling Shen
- Shanghai Laboratory Animal Research Center, Shanghai, 201203, China
| | - Xianmin Zhu
- Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, 201210, China
- Shanghai Academy of Sciences & Technology Institute of Model Animals Transformation, Shanghai, 201203, China
| | - Nicola Elvassore
- Shanghai Clinical Research and Trial Center, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, 201210, China
- Department of Industrial Engineering, University of Padova, Padova, 35131, Italy
| |
Collapse
|
4
|
Wang Z, Xu C, Wang Q, Wang Y. Repurposing of nervous system drugs for cancer treatment: recent advances, challenges, and future perspectives. Discov Oncol 2025; 16:396. [PMID: 40133751 PMCID: PMC11936871 DOI: 10.1007/s12672-025-02067-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
The nervous system plays a critical role in developmental biology and oncology, influencing processes from ontogeny to the complex dynamics of cancer progression. Interactions between the nervous system and cancer significantly affect oncogenesis, tumor growth, invasion, metastasis, treatment resistance, inflammation that promotes tumors, and the immune response. A comprehensive understanding of the signal transduction pathways involved in cancer biology is essential for devising effective anti-cancer strategies and overcoming resistance to existing therapies. Recent advances in cancer neuroscience promise to establish a new cornerstone of cancer therapy. Repurposing drugs originally developed for modulating nerve signal transduction represent a promising approach to target oncogenic signaling pathways in cancer treatment. This review endeavors to investigate the potential of repurposing neurological drugs, which target neurotransmitters and neural pathways, for oncological applications. In this context, it aims to bridge the interdisciplinary gap between neurology, psychiatry, internal medicine, and oncology. By leveraging already approved drugs, researchers can utilize existing extensive safety and efficacy data, thereby reducing both the time and financial resources necessary for the development of new cancer therapies. This strategy not only promises to enhance patient outcomes but also to expand the array of available treatments, thereby enriching the therapeutic landscape in oncology.
Collapse
Affiliation(s)
- Zixun Wang
- Nanshan School, Guangzhou Medical University, Jingxiu Road, Panyu District, Guangzhou, 511436, China
| | - Chen Xu
- Department of Gynecologic Oncology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai Key Laboratory of Embryo Original Disease, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Qi Wang
- Department of Gynecologic Oncology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai Key Laboratory of Embryo Original Disease, Shanghai Jiao Tong University, Shanghai, 200025, China.
| | - Yudong Wang
- Department of Gynecologic Oncology, the International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai Key Laboratory of Embryo Original Disease, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|
5
|
Quan T, Qin Z, He T, Fisher GJ. Integrin α11β1 as a Key Collagen Receptor in Human Skin Dermis: Insight into Fibroblast Function and Skin Dermal Aging. J Invest Dermatol 2025:S0022-202X(25)00364-1. [PMID: 40139564 DOI: 10.1016/j.jid.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025]
Abstract
Collagen-binding integrins play a crucial role in facilitating fibroblast-collagen interactions and regulating cellular functions. In this study, we identified that among 4 collagen-binding integrins, integrin α11 was the predominant type in human skin dermal fibroblasts and that loss of integrin α11 expression contributed to skin dermal aging. Integrin α11β1 was critical for regulating fibroblast-collagen interactions, including cell adhesion, spreading, morphology, mechanical tension, and the production of collagenous extracellular matrix. TGF-β was recognized as the primary regulator of integrin α11 expression. Notably, dermal fibroblasts in aged human skin demonstrated impaired TGF-β signaling, which coincided with a loss of integrin α11 expression, whereas the expression of other collagen-binding integrins remained unchanged. Similarly, in senescent dermal fibroblasts in vitro, impaired TGF-β signaling was associated with a significant reduction in integrin α11 expression, whereas other collagen-binding integrins were upregulated or unaffected. Furthermore, collapsed dermal fibroblasts, a key characteristic of dermal fibroblasts in aged human skin, specifically downregulated integrin α11, whereas other collagen-binding integrins were upregulated or remained unchanged. These findings suggest a negative feedback loop in which an impaired TGF-β-integrin α11β1 axis and fibroblast collapse promote dermal aging in human skin. This self-reinforcing cycle reflects the progressive and unidirectional nature of biological aging.
Collapse
Affiliation(s)
- Taihao Quan
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| | - Zhaoping Qin
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Tianyuan He
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Gary J Fisher
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
6
|
Tian L, Wang Y, Guan J, Zhang L, Fan J. The Prognostic Value and Immunomodulatory Role of Spsb2, a Novel Immune Checkpoint Molecule, in Hepatocellular Carcinoma. Genes (Basel) 2025; 16:346. [PMID: 40149497 PMCID: PMC11941779 DOI: 10.3390/genes16030346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Liver cancer, specifically hepatocellular carcinoma (LIHC), ranks as the second most common cause of cancer-related fatalities globally. Moreover, the occurrence rate of LIHC is steadily increasing. A recently identified gene, SPSB2, has been implicated in cell signaling, impacting the development and progression of non-small cell lung cancer. Nevertheless, studies on the role of SPSB2 in the pathogenesis of LIHC are lacking. METHODS Using the TCGA, GTEx, and GEO databases, we obtained differentially expressed genes that affect the prognosis of patients with LIHC. We utilized the Kruskal-Wallis test, along with univariate and multivariate COX regression analyses, to determine the correlation between SPSB2 and patient clinical indicators. Potential biological functions of SPSB2 in LIHC were explored by enrichment analysis, ssGSEA, and Spearman correlation analysis. Finally, LIHC cell lines Huh7 and SMMC-7721 were used to validate the biological function of SPSB2. RESULTS The results showed LIHC patients with higher SPSB2 expression had a poorer prognosis, and SPSB2 expression was significantly correlated with LIHC patients' Histologic grade, Pathologic T stage, Prothrombin time, Pathologic stage, BMI, weight, adjacent hepatic tissue inflammation, AFP level, and OS event (p < 0.05). SPSB2 shows notable enrichment in pathways linked to tumorigenesis and the immune system. Moreover, its expression is strongly connected to immune cells and immune checkpoints. Knockdown of SPSB2 expression in Huh7 cells and SMMC-7721 cells inhibits SPSB2's biological functions, including proliferation, invasion, metastasis, and other phenotypes. CONCLUSIONS SPSB2 plays a crucial role in the development of LIHC. It is related to the immune response and unfavorable outcomes. SPSB2 may function as a clinical biomarker for prognosis.
Collapse
Affiliation(s)
- Lv Tian
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Yiming Wang
- School of Nursing, Jilin University, Changchun 130021, China
| | - Jiexin Guan
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Lu Zhang
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Jun Fan
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 611731, China
| |
Collapse
|
7
|
Emerson MJ, Willacy O, Madsen CD, Reuten R, Brøchner CB, Lund TK, Dahl AB, Jensen THL, Erler JT, Mayorca-Guiliani AE. Machine learning identifies remodeling patterns in human lung extracellular matrix. Acta Biomater 2025; 195:94-103. [PMID: 39746529 DOI: 10.1016/j.actbio.2024.12.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/15/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Organ function depends on the three-dimensional integrity of the extracellular matrix (ECM). The structure resulting from the location and association of ECM components is a central regulator of cell behavior, but a dearth of matrix-specific analysis keeps it unresolved. Here, we deploy a high-resolution, 3D ECM mapping method and design a machine-learning powered pipeline to detect and characterize ECM architecture during health and disease. We deploy these tools in the human lung, an organ heavily dependent on ECM structure that can host diseases with different histopathologies. We analyzed segments from healthy, emphysema, usual interstitial pneumonia, sarcoidosis, and COVID-19 patients, and produced a remodeling signature per disease and a health/disease probability map from which we inferred the architecture of healthy and diseased ECM. Our methods demonstrate that exaggerated matrix deposition, or fibrosis, is not a single phenomenon, but a series of disease-specific alterations. STATEMENT OF SIGNIFICANCE: The extracellular matrix, or ECM, is the foremost biomaterial. It shapes and supports all tissues while regulating all cells. ECM structure is intricate, yet precise: each organ, at every stage, has a specific ECM structure. During disease, tissues suffer from structural changes that accelerate and perpetuate illness by dysregulating cells. Both healthy and diseased ECM structures are of great biomedical importance, but surprisingly, they have not been mapped in detail. Here, we present a method that combines tissue engineering with machine learning to reveal, map and analyze ECM structures, applied it to pulmonary diseases that kill millions every year. This method can bring objectivity and a higher degree of confidence into the diagnosis of pulmonary disease. In addition the amount of tissue needed for a firm diagnosis may be much smaller than required for manual microscopy evaluation.
Collapse
Affiliation(s)
- Monica J Emerson
- Section for Visual Computing, Department of Applied Mathematics and Computer Science, Technical University of Denmark. Kongens Lyngby, Denmark; Currently at Digital Science and Innovation, Novo Nordisk A/S, Måløv, Denmark
| | - Oliver Willacy
- Biotech Research and Innovation Center, Faculty of Health Sciences, University of Copenhagen. Copenhagen, Denmark; Department of Pathology, Rigshospitalet, University Hospital of Copenhagen. Copenhagen, Denmark
| | - Chris D Madsen
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University. Lund, Sweden; Currently at Symphogen A/S, Ballerup, Denmark
| | - Raphael Reuten
- Biotech Research and Innovation Center, Faculty of Health Sciences, University of Copenhagen. Copenhagen, Denmark; Currently at Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg. Freiburg, Germany
| | - Christian B Brøchner
- Department of Pathology, Rigshospitalet, University Hospital of Copenhagen. Copenhagen, Denmark
| | - Thomas K Lund
- Section for Lung Transplantation, Department of Cardiology, Rigshospitalet, University Hospital of Copenhagen. Copenhagen, Denmark
| | - Anders B Dahl
- Section for Visual Computing, Department of Applied Mathematics and Computer Science, Technical University of Denmark. Kongens Lyngby, Denmark
| | - Thomas H L Jensen
- Department of Pathology, Rigshospitalet, University Hospital of Copenhagen. Copenhagen, Denmark.
| | - Janine T Erler
- Biotech Research and Innovation Center, Faculty of Health Sciences, University of Copenhagen. Copenhagen, Denmark.
| | - Alejandro E Mayorca-Guiliani
- Biotech Research and Innovation Center, Faculty of Health Sciences, University of Copenhagen. Copenhagen, Denmark; Currently at Nordic Bioscience A/S. Herlev, Denmark.
| |
Collapse
|
8
|
Imtiaz I, Schloss J, Bugarcic A. Interplay Between Traditional and Scientific Knowledge: Phytoconstituents and Their Roles in Lung and Colorectal Cancer Signaling Pathways. Biomolecules 2025; 15:380. [PMID: 40149916 PMCID: PMC11940637 DOI: 10.3390/biom15030380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/27/2025] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
Natural plant products have been used for cancer treatment since ancient times and continue to play a vital role in modern anticancer drug development. However, only a small fraction of identified medicinal plants has been thoroughly investigated, particularly for their effects on cellular pathways in lung and colorectal cancers, two under-researched cancers with poor prognostic outcomes (lung cancers). This review focuses on the lung and colorectal cancer signaling pathways modulated by bioactive compounds from eleven traditional medicinal plants: Curcuma longa, Astragalus membranaceus, Glycyrrhiza glabra, Althaea officinalis, Echinacea purpurea, Sanguinaria canadensis, Codonopsis pilosula, Hydrastis canadensis, Lobelia inflata, Scutellaria baicalensis, and Zingiber officinale. These plants were selected based on their documented use in traditional medicine and modern clinical practice. Selection criteria involved cross-referencing herbs identified in a scoping review of traditional cancer treatments and findings from an international survey on herbal medicine currently used for lung and colorectal cancer management by our research group and the availability of existing literature on their anticancer properties. The review identifies several isolated phytoconstituents from these plants that exhibit anticancer properties by modulating key signaling pathways such as PI3K/Akt/mTOR, RAS/RAF/MAPK, Wnt/β-catenin, and TGF-β in vitro. Notable constituents include sanguinarine, berberine, hydrastine, lobeline, curcumin, gingerol, shogaol, caffeic acid, echinacoside, cichoric acid, glycyrrhizin, 18-β-glycyrrhetinic acid, astragaloside IV, lobetyolin, licochalcone A, baicalein, baicalin, wogonin, and glycyrol. Curcumin and baicalin show preclinical effectiveness but face bioavailability challenges, which may be overcome by combining them with piperine or using oral extracts to enhance gut microbiome conversion, integrating traditional knowledge with modern strategies for improved outcomes. Furthermore, herbal extracts from Echinacea, Glycyrrhiza, and Codonopsis, identified in traditional knowledge, are currently in clinical trials. Notably, curcumin and baicalin also modulate miRNA pathways, highlighting a promising intersection of modern science and traditional medicine. Thus, the development of anticancer therapeutics continues to benefit from the synergy of traditional knowledge, scientific innovation, and technological advancements.
Collapse
Affiliation(s)
| | | | - Andrea Bugarcic
- National Centre for Naturopathic Medicine, Faculty of Health, Southern Cross University, Military Road, Lismore, NSW 2480, Australia; (I.I.); (J.S.)
| |
Collapse
|
9
|
Peeney D, Kumar S, Singh TP, Liu Y, Jensen SM, Chowdhury A, Coates-Park S, Rich J, Gurung S, Fan Y, Meerzaman D, Stetler-Stevenson WG. Timp2 loss-of-function mutation and TIMP2 treatment in a murine model of NSCLC: Modulation of immunosuppression and oncogenic signaling. Transl Oncol 2025; 53:102309. [PMID: 39904284 PMCID: PMC11846589 DOI: 10.1016/j.tranon.2025.102309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/06/2024] [Accepted: 01/30/2025] [Indexed: 02/06/2025] Open
Abstract
Mounting evidence suggests that the tissue inhibitor of metalloproteinases-2 (TIMP2) can reduce tumor burden and metastasis. However, the demonstration of such anti-tumor activity and associated mechanisms using in vivo tumor models is lacking. The effects of a Timp2 functional mutation and administration of recombinant TIMP2 were examined in both orthotopic and heterotopic murine models of lung cancer using C57Bl/6 syngeneic Lewis Lung 2-luciferase 2 cells (LL2-Luc2) cells. Mice harboring a functional mutation of TIMP2 (mT2) display markedly increased primary lung tumor growth, increased mortality, enriched vasculature, and enhanced infiltration of pro-tumorigenic, immunosuppressive myeloid cells. Treatment with recombinant TIMP2 reduced primary tumor growth in both mutant and wild-type (wt) mice. Comparison of transcriptional profiles of lung tissues from tumor-free, wt versus mT2 mice reveals only minor changes. However, lung tumor-bearing mice of both genotypes demonstrate significant genotype-dependent changes in gene expression following treatment with TIMP. In tumor-bearing wt mice, TIMP2 treatment reduced the expression of upstream oncogenic mediators, whereas treatment of mT2 mice resulted in an immunomodulatory phenotype. A heterotopic subcutaneous model generating metastatic pulmonary tumors demonstrated that daily administration of recombinant TIMP2 significantly reduces the expression of heat shock proteins, suggesting a reduction of cell-stress responses. In summary, we describe how TIMP2 exerts novel, anti-tumor effects in a murine model of lung cancer and that rTIMP2 treatment supports a normalizing effect on the tumor microenvironment. Our findings show that TIMP2 treatment demonstrates significant potential as an adjuvant in the treatment of NSCLC.
Collapse
Affiliation(s)
- David Peeney
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA.
| | - Sarvesh Kumar
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA
| | - Tej Pratap Singh
- Laboratory of Molecular Immunology, National Institute for Allergy, and Infectious Disease (NIAID), Bethesda, MD 20892, USA
| | - Yueqin Liu
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA
| | - Sandra M Jensen
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA
| | - Ananda Chowdhury
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA
| | - Sasha Coates-Park
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA
| | - Joshua Rich
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA
| | - Sadeechya Gurung
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA
| | - Yu Fan
- Computational Genomics and Bioinformatics Group, Center for Biomedical Informatics & Information Technology, National Cancer Institute, Rockville, MD 20850, USA
| | - Daoud Meerzaman
- Computational Genomics and Bioinformatics Group, Center for Biomedical Informatics & Information Technology, National Cancer Institute, Rockville, MD 20850, USA
| | - William G Stetler-Stevenson
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA.
| |
Collapse
|
10
|
Xie Y, Liu F, Wu Y, Zhu Y, Jiang Y, Wu Q, Dong Z, Liu K. Inflammation in cancer: therapeutic opportunities from new insights. Mol Cancer 2025; 24:51. [PMID: 39994787 PMCID: PMC11849313 DOI: 10.1186/s12943-025-02243-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/20/2025] [Indexed: 02/26/2025] Open
Abstract
As one part of the innate immune response to external stimuli, chronic inflammation increases the risk of various cancers, and tumor-promoting inflammation is considered one of the enabling characteristics of cancer development. Recently, there has been growing evidence on the role of anti-inflammation therapy in cancer prevention and treatment. And researchers have already achieved several noteworthy outcomes. In the review, we explored the underlying mechanisms by which inflammation affects the occurrence and development of cancer. The pro- or anti-tumor effects of these inflammatory factors such as interleukin, interferon, chemokine, inflammasome, and extracellular matrix are discussed. Since FDA-approved anti-inflammation drugs like aspirin show obvious anti-tumor effects, these drugs have unique advantages due to their relatively fewer side effects with long-term use compared to chemotherapy drugs. The characteristics make them promising candidates for cancer chemoprevention. Overall, this review discusses the role of these inflammatory molecules in carcinogenesis of cancer and new inflammation molecules-directed therapeutic opportunities, ranging from cytokine inhibitors/agonists, inflammasome inhibitors, some inhibitors that have already been or are expected to be applied in clinical practice, as well as recent discoveries of the anti-tumor effect of non-steroidal anti-inflammatory drugs and steroidal anti-inflammatory drugs. The advantages and disadvantages of their application in cancer chemoprevention are also discussed.
Collapse
Affiliation(s)
- Yifei Xie
- Department of Pathology and Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450001, China
| | - Fangfang Liu
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450007, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450001, China
| | - Yunfei Wu
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yuer Zhu
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yanan Jiang
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450007, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450001, China
| | - Qiong Wu
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450007, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450001, China
| | - Zigang Dong
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China.
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450007, China.
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450001, China.
| | - Kangdong Liu
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China.
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450007, China.
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450001, China.
| |
Collapse
|
11
|
Tang L, Zhang W, Qi T, Jiang Z, Tang D. Exosomes play a crucial role in remodeling the tumor microenvironment and in the treatment of gastric cancer. Cell Commun Signal 2025; 23:82. [PMID: 39948541 PMCID: PMC11827163 DOI: 10.1186/s12964-024-02009-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/21/2024] [Indexed: 02/16/2025] Open
Abstract
Gastric cancer (GC) is a common and frequent malignant cancer of the digestive system with a poor prognosis. In addition to common therapies such as surgical resection and chemotherapy, novel biological interventions are quite valuable for research. Exosomes are extracellular vesicles (EVs) that originate from various cell types and contain proteins, RNA, DNA, and other components that transmit biological signals and mediate intercellular communication. Numerous studies have shown that exosomes shape the tumor microenvironment (TME) by affecting hypoxia, inflammation, immunity, metabolism, and interstitial changes in the tumor, playing a crucial role in the development and metastasis of GC. This article reviews the important role of exosomes in the TME of GC and explores their potential clinical applications in GC treatment.
Collapse
Affiliation(s)
- Lingyun Tang
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, China
| | - Wenjie Zhang
- School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Teng Qi
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
| | - Zhengting Jiang
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225000, China.
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital; The Yangzhou Clinical Medical College of Xuzhou Medical University; The Yangzhou School of Clinical Medicine of Dalian Medical University; The Yangzhou School of Clinical Medicine of Nanjing Medical University; Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou, 225000, China.
| |
Collapse
|
12
|
Tetrick MG, Emon MAB, Doha U, Marcellus M, Symanski J, Ramanathan V, Saif MTA, Murphy CJ. Decoupling chemical and mechanical signaling in colorectal cancer cell migration. Sci Rep 2025; 15:4952. [PMID: 39929899 PMCID: PMC11811049 DOI: 10.1038/s41598-025-89152-4] [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: 10/13/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Colorectal cancer metastasis is governed by a variety of chemical and mechanical signaling that are largely influenced by cancer-associated fibroblasts (CAFs) in the tumor microenvironment. Here, we deconvolute the chemical from mechanical signaling in the case of the colon cancer cell line HCT-116 and CAFs. We examined three chemoattractants (CXCL12, TGF-β, and activin A) which allegedly are secreted by CAFs and induce HCT-116 cell migration. None of the chemoattractants tested resulted in enhanced migration of HCT-116 in a 2D transwell assay, at low cell density. Similarly, CAF-conditioned media also did not lead to enhanced HCT-116 migration, while CAFs co-cultured in the transwell assay did lead to increased HCT-116 migration. This result suggests that either high cell densities are required for chemotaxis, and/or a reciprocal two-way signaling network between CAFs and HCT-116 is necessary to induce chemotaxis. Surprisingly, we find that HCT-116 cells exhibit enhanced migration along the axis of mechanical stress in a 3D collagen matrix, at very high cell densities. This migration is independent of whether the strain is induced mechanically or by CAFs. By comparing purely mechanical and purely chemical migration to a 3D co-culture of CAFs and HCT-116 containing both chemical and mechanical cues, it is concluded that HCT-116 migration is dominated by mechanical signaling, while chemical cues are less influential.
Collapse
Affiliation(s)
- Maxwell G Tetrick
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Md Abul Bashar Emon
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Umnia Doha
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Marsophia Marcellus
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Joseph Symanski
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Valli Ramanathan
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - M Taher A Saif
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Catherine J Murphy
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
13
|
Wang J, Tian N, Tian T, Xiao L, Zhou X, Liu G, Zhang Z, Zhao Y, Guo J, Lin Q, Jiang Y. Low toxicity ginsenoside Rg1-carbon nanodots as a potential therapeutic agent for human non-small cell lung cancer. Colloids Surf B Biointerfaces 2025; 246:114392. [PMID: 39579497 DOI: 10.1016/j.colsurfb.2024.114392] [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: 09/20/2024] [Revised: 11/16/2024] [Accepted: 11/19/2024] [Indexed: 11/25/2024]
Abstract
Here ginsenoside Rg1 was used to synthesise Rg1 carbon nanodots via a one-step hydrothermal method. The surface of the Rg1 carbon nanodots is rich in hydrophilic functional groups with good water solubility and biocompatibility. The Rg1 carbon nanodots exhibited a high inhibitory effect on the proliferation, migration, and proapoptotic ability of non-small cell lung cancer A549 cells. The changes in the levels of ROS, Ca2+, and MMP in A549 cells after the administration of Rg1 carbon nanodots were evaluated and further correlated with relevant proteins in the caspase apoptotic pathway. Proteomic screening revealed that the Rg1 carbon nanodots could regulate A549 cell apoptosis by activating the expression of MAPK pathway-related proteins. In the in vivo experiment, the therapeutic efficacy of the Rg1 carbon nanodots in inhibiting tumour growth was much higher than that of commonly used chemotherapy drugs, with negligible toxicity and side effects. Immunohistochemical staining showed that the expression of caspase- and MAPK pathway-related proteins in mouse tumour tissues was consistent with that at the cellular level. The results suggest that Rg1 carbon nanodots can promote tumour apoptosis and represent a potential therapeutic agent for human non-small-cell lung cancer.
Collapse
Affiliation(s)
- Jifeng Wang
- Jilin Ginseng Academy, Institute of Traditional Chinese Medicine, Hospital of Affiliated Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, PR China; Department of Chemistry, Laboratory of Advanced Materials, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, PR China
| | - Ning Tian
- Jilin Ginseng Academy, Institute of Traditional Chinese Medicine, Hospital of Affiliated Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Tenghui Tian
- Jilin Ginseng Academy, Institute of Traditional Chinese Medicine, Hospital of Affiliated Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Lizhi Xiao
- Jilin Ginseng Academy, Institute of Traditional Chinese Medicine, Hospital of Affiliated Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Xuechun Zhou
- Jilin Ginseng Academy, Institute of Traditional Chinese Medicine, Hospital of Affiliated Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Guancheng Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Zhe Zhang
- Jilin Ginseng Academy, Institute of Traditional Chinese Medicine, Hospital of Affiliated Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Yu Zhao
- Jilin Ginseng Academy, Institute of Traditional Chinese Medicine, Hospital of Affiliated Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, PR China.
| | - Jiajuan Guo
- Jilin Ginseng Academy, Institute of Traditional Chinese Medicine, Hospital of Affiliated Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, PR China.
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Yingnan Jiang
- Jilin Ginseng Academy, Institute of Traditional Chinese Medicine, Hospital of Affiliated Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, PR China; Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
14
|
Adler FR, Griffiths JI. Mathematical models of intercellular signaling in breast cancer. Semin Cancer Biol 2025; 109:91-100. [PMID: 39890041 PMCID: PMC11858920 DOI: 10.1016/j.semcancer.2025.01.005] [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: 02/28/2023] [Revised: 01/03/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND AND OBJECTIVES The development and regulation of healthy and cancerous breast tissue is guided by communication between cells. Diverse signals are exchanged between cancer cells and non-cancerous cells of the tumor microenvironment (TME), influencing all stages of tumor progression. Mathematical models are essential for understanding how this complex network determines cancer progression and the effectiveness of treatment. METHODOLOGY We reviewed the current dynamical mathematical models of intercellular signaling in breast cancer, examining models with cancer cells only, fibroblasts, endothelial cells, macrophages and the immune system as whole. We categorized the goals and complexity of these models, to highlight how they can explain many features of cancer emergence and progression. RESULTS We found that dynamical models of intercellular signaling can elucidate tissue-level dysregulation in cancer by explaining: i) maintenance of non-heritable intratumor phenotypic heterogeneity, ii) transitions between tumor dormancy and accelerated invasive growth, iii) stromal support of tumor vascularization and growth factor enrichment and iv) suppression of immune infiltration and cancer surveillance. These models also provide a framework to propose novel TME-targeting treatment strategies. However, most models were focused on a highly selected and small set of signaling interactions between a few cell types, and their translational applicability were severely limited by the availability of tumor-specific data for personalized model calibration. CONCLUSIONS AND IMPLICATIONS Mathematical models of breast cancer have many challenges and opportunities to incorporate signaling. The four key challenges are: 1) finding ways to treat signaling networks as a context-dependent language that incorporates non-linear and non-additive responses, 2) identifying the key cell phenotypes that signals control and understanding the feedbacks between signals and phenotype that determine the progression of cancer, (3) estimating parameters of specific patient tumors early in treatment, 4) linking models with novel data collection methods that have single cell and spatial resolution. As our approaches advance, it is our hope that dynamical mathematical models of inter-cellular signaling can play a central role in identifying and testing new treatment strategies as well as forecasting impacts of disease treatment.
Collapse
Affiliation(s)
- Frederick R Adler
- Department of Mathematics, University of Utah, 155 South 1400 East, Salt Lake City, UT 84112, USA; School of Biological Sciences, 257 South 1400 East, University of Utah, Salt Lake City, UT, 84112 USA..
| | - Jason I Griffiths
- Department of Mathematics, University of Utah, 155 South 1400 East, Salt Lake City, UT 84112, USA; Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| |
Collapse
|
15
|
Huang Q, Hu B, Zhang P, Yuan Y, Yue S, Chen X, Liang J, Tang Z, Zhang B. Neuroscience of cancer: unraveling the complex interplay between the nervous system, the tumor and the tumor immune microenvironment. Mol Cancer 2025; 24:24. [PMID: 39825376 PMCID: PMC11740516 DOI: 10.1186/s12943-024-02219-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 12/26/2024] [Indexed: 01/20/2025] Open
Abstract
The study of the multifaceted interactions between neuroscience and cancer is an emerging field with significant implications for understanding tumor biology and the innovation in therapeutic approaches. Increasing evidence suggests that neurological functions are connected with tumorigenesis. In particular, the peripheral and central nervous systems, synapse, neurotransmitters, and neurotrophins affect tumor progression and metastasis through various regulatory approaches and the tumor immune microenvironment. In this review, we summarized the neurological functions that affect tumorigenesis and metastasis, which are controlled by the central and peripheral nervous systems. We also explored the roles of neurotransmitters and neurotrophins in cancer progression. Moreover, we examined the interplay between the nervous system and the tumor immune microenvironment. We have also identified drugs that target the nervous system for cancer treatment. In this review we present the work supporting that therapeutic agent targeting the nervous system could have significant potential to improve cancer therapy.
Collapse
Affiliation(s)
- Qibo Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, NHC Key Laboratory of Organ Transplantation, Wuhan, China
| | - Bai Hu
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ping Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ye Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Shiwei Yue
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, NHC Key Laboratory of Organ Transplantation, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China.
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, NHC Key Laboratory of Organ Transplantation, Wuhan, China.
| | - Junnan Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China.
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, NHC Key Laboratory of Organ Transplantation, Wuhan, China.
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China.
- Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Wuhan, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, NHC Key Laboratory of Organ Transplantation, Wuhan, China.
| |
Collapse
|
16
|
Zhao F, Jiang X, Li Y, Huang T, Xiahou Z, Nie W, Li Q. Characterizing tumor biology and immune microenvironment in high-grade serous ovarian cancer via single-cell RNA sequencing: insights for targeted and personalized immunotherapy strategies. Front Immunol 2025; 15:1500153. [PMID: 39896800 PMCID: PMC11782144 DOI: 10.3389/fimmu.2024.1500153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 12/19/2024] [Indexed: 02/04/2025] Open
Abstract
Background High-grade serous ovarian cancer (HGSOC), the predominant subtype of epithelial ovarian cancer, is frequently diagnosed at an advanced stage due to its nonspecific early symptoms. Despite standard treatments, including cytoreductive surgery and platinum-based chemotherapy, significant improvements in survival have been limited. Understanding the molecular mechanisms, immune landscape, and drug sensitivity of HGSOC is crucial for developing more effective and personalized therapies. This study integrates insights from cancer immunology, molecular profiling, and drug sensitivity analysis to identify novel therapeutic targets and improve treatment outcomes. Utilizing single-cell RNA sequencing (scRNA-seq), the study systematically examines tumor heterogeneity and immune microenvironment, focusing on biomarkers influencing drug response and immune activity, aiming to enhance patient outcomes and quality of life. Methods scRNA-seq data was obtained from the GEO database in this study. Differential gene expression was analyzed using gene ontology and gene set enrichment methods. InferCNV identified malignant epithelial cells, while Monocle, Cytotrace, and Slingshot software inferred subtype differentiation trajectories. The CellChat software package predicted cellular communication between malignant cell subtypes and other cells, while pySCENIC analysis was utilized to identify transcription factor regulatory networks within malignant cell subtypes. Finally, the analysis results were validated through functional experiments, and a prognostic model was developed to assess prognosis, immune infiltration, and drug sensitivity across various risk groups. Results This study investigated the cellular heterogeneity of HGSOC using scRNA-seq, focusing on tumor cell subtypes and their interactions within the tumor microenvironment. We confirmed the key role of the C2 IGF2+ tumor cell subtype in HGSOC, which was significantly associated with poor prognosis and high levels of chromosomal copy number variations. This subtype was located at the terminal differentiation of the tumor, displaying a higher degree of malignancy and close association with stage IIIC tissue types. The C2 subtype was also associated with various metabolic pathways, such as glycolysis and riboflavin metabolism, as well as programmed cell death processes. The study highlighted the complex interactions between the C2 subtype and fibroblasts through the MK signaling pathway, which may be closely related to tumor-associated fibroblasts and tumor progression. Elevated expression of PRRX1 was significantly connected to the C2 subtype and may impact disease progression by modulating gene transcription. A prognostic model based on the C2 subtype demonstrated its association with adverse prognosis outcomes, emphasizing the importance of immune infiltration and drug sensitivity analysis in clinical intervention strategies. Conclusion This study integrates molecular oncology, immunotherapy, and drug sensitivity analysis to reveal the mechanisms driving HGSOC progression and treatment resistance. The C2 IGF2+ tumor subtype, linked to poor prognosis, offers a promising target for future therapies. Emphasizing immune infiltration and drug sensitivity, the research highlights personalized strategies to improve survival and quality of life for HGSOC patients.
Collapse
Affiliation(s)
- Fu Zhao
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaojing Jiang
- Affiliated Hospital of Shandong Academy of Traditional Chinese Medicine, Jinan, China
| | - Yumeng Li
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tianjiao Huang
- The First School of Clinical Medicine, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Zhikai Xiahou
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Wenyang Nie
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qian Li
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
17
|
Sergazy S, Seydahmetova R, Gulyayev A, Shulgau Z, Aljofan M. The Role of Exosomes in Cancer Progression and Therapy. BIOLOGY 2025; 14:27. [PMID: 39857258 PMCID: PMC11763171 DOI: 10.3390/biology14010027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/03/2024] [Accepted: 12/16/2024] [Indexed: 01/27/2025]
Abstract
Exosomes are small extracellular vesicles and are crucial in intercellular communication. Interestingly, tumor-derived exosomes carry oncogenic molecules, such as proteins and microRNAs, which can reprogram recipient cells, promote angiogenesis, and stimulate cancer pre-metastatic niche, supporting cancer growth and metastasis. On the other hand, their biocompatibility, stability, and ability to cross biological barriers make them attractive candidates for drug delivery. Recent advances have shown the potential for exosomes to be used in early disease detection and in targeted drug therapy by delivering therapeutic agents specifically to tumor sites. Despite the promising applications, a number of challenges remain, including exosome isolation and characterization, as well as their inherent heterogeneity. Thus, the current review aims to describe the roles of exosomes in health and disease, and discuss the challenges that hinder their development into becoming useful medical tools.
Collapse
Affiliation(s)
- Shynggys Sergazy
- LLP VICTUS PHARM, Astana 010000, Kazakhstan; (S.S.); (R.S.); (A.G.); (Z.S.)
- National Laboratory Astana, Center for Life Sciences, Nazarbayev University, Astana 010000, Kazakhstan
| | - Roza Seydahmetova
- LLP VICTUS PHARM, Astana 010000, Kazakhstan; (S.S.); (R.S.); (A.G.); (Z.S.)
| | - Alexandr Gulyayev
- LLP VICTUS PHARM, Astana 010000, Kazakhstan; (S.S.); (R.S.); (A.G.); (Z.S.)
- National Laboratory Astana, Center for Life Sciences, Nazarbayev University, Astana 010000, Kazakhstan
| | - Zarina Shulgau
- LLP VICTUS PHARM, Astana 010000, Kazakhstan; (S.S.); (R.S.); (A.G.); (Z.S.)
- National Laboratory Astana, Center for Life Sciences, Nazarbayev University, Astana 010000, Kazakhstan
| | - Mohamad Aljofan
- National Laboratory Astana, Center for Life Sciences, Nazarbayev University, Astana 010000, Kazakhstan
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan
| |
Collapse
|
18
|
Galluzzi L, Spada S. Epigenetic regulation of cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2025; 390:xiii-xvii. [PMID: 39864898 DOI: 10.1016/s1937-6448(25)00015-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Affiliation(s)
- Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, United States.
| | - Sheila Spada
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy.
| |
Collapse
|
19
|
Del Toro K, Licon-Munoz Y, Crabtree W, Oper T, Robbins C, Hines WC. Breast pericytes: a newly identified driver of tumor cell proliferation. Front Oncol 2024; 14:1455484. [PMID: 39741968 PMCID: PMC11685225 DOI: 10.3389/fonc.2024.1455484] [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: 06/26/2024] [Accepted: 11/27/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction Effective treatment of breast cancer remains a formidable challenge, partly due to our limited understanding of the complex microenvironmental factors that contribute to disease pathology. Among these factors are tissue-resident perivascular cells, which play crucial roles in shaping vascular basement membranes, maintaining vessel integrity, and communicating with adjacent endothelial cells. Despite their essential functions, perivascular cells have been relatively overlooked. Identifying them by immunostaining has been challenging due to their low abundance, inherent heterogeneity, and shared marker expression with other cell types. These challenges have hindered efforts to purify pericytes and generate primary cell models for studying their biology. Methods Using a recently developed FACS method, we successfully identified and purified each cell type from breast tissues, allowing us to deep-sequence their transcriptomes and generate primary cell models of each cell type-including pericytes. Here, we used these data to analyze cell-type-specific gene expression in tumors, which revealed a strong association between pericyte-specific genes and breast cancer patient mortality. To explore this association, we defined the heterogeneity of breast pericytes using single-cell RNA sequencing and identified a broad marker for visualizing perivascular cells in breast tumors. Results Remarkably, we discovered perivascular cells dissociated from vessels and emerged as a dominant mesenchymal cell type in a subset of breast tumors that contrasted with their normal perivascular location. Moreover, when we purified pericytes from the breast and cultured them alongside breast tumor cells, we discovered that they induced rapid tumor cell growth significantly greater than isogenic fibroblast controls. Discussion These findings identify perivascular cells as a key microenvironmental factor in breast cancer, highlighting the critical need for further research to explore their biology and identify specific stimulatory mechanisms that could be targeted therapeutically.
Collapse
Affiliation(s)
| | | | | | | | | | - William C. Hines
- Department of Biochemistry and Molecular Biology, University of New Mexico School of
Medicine, 1 University of New Mexico MSC08 4670, Albuquerque, NM, United States
| |
Collapse
|
20
|
Hashimoto A, Hashimoto S. Plasticity and Tumor Microenvironment in Pancreatic Cancer: Genetic, Metabolic, and Immune Perspectives. Cancers (Basel) 2024; 16:4094. [PMID: 39682280 DOI: 10.3390/cancers16234094] [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: 11/09/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer has long been believed to be a genetic disease caused by the accumulation of mutations in key genes involved in cellular processes. However, recent advances in sequencing technology have demonstrated that cells with cancer driver mutations are also present in normal tissues in response to aging, environmental damage, and chronic inflammation, suggesting that not only intrinsic factors within cancer cells, but also environmental alterations are important key factors in cancer development and progression. Pancreatic cancer tissue is mostly comprised of stromal cells and immune cells. The desmoplasmic microenvironment characteristic of pancreatic cancer is hypoxic and hypotrophic. Pancreatic cancer cells may adapt to this environment by rewiring their metabolism through epigenomic changes, enhancing intrinsic plasticity, creating an acidic and immunosuppressive tumor microenvironment, and inducing noncancerous cells to become tumor-promoting. In addition, pancreatic cancer has often metastasized to local and distant sites by the time of diagnosis, suggesting that a similar mechanism is operating from the precancerous stage. Here, we review key recent findings on how pancreatic cancers acquire plasticity, undergo metabolic reprogramming, and promote immunosuppressive microenvironment formation during their evolution. Furthermore, we present the following two signaling pathways that we have identified: one based on the small G-protein ARF6 driven by KRAS/TP53 mutations, and the other based on the RNA-binding protein Arid5a mediated by inflammatory cytokines, which promote both metabolic reprogramming and immune evasion in pancreatic cancer. Finally, the striking diversity among pancreatic cancers in the relative importance of mutational burden and the tumor microenvironment, their clinical relevance, and the potential for novel therapeutic strategies will be discussed.
Collapse
Affiliation(s)
- Ari Hashimoto
- Department of Molecular Biology, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Shigeru Hashimoto
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0818, Japan
| |
Collapse
|
21
|
Wang X, Ma X, Chen S, Fan M, Jin C, Chen Y, Wang S, Wang Z, Meng F, Zhang C, Yang L. Harnessing m1A modification: a new frontier in cancer immunotherapy. Front Immunol 2024; 15:1517604. [PMID: 39687616 PMCID: PMC11647001 DOI: 10.3389/fimmu.2024.1517604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
N1-methyladenosine (m1A) modification is an epigenetic change that occurs on RNA molecules, regulated by a suite of enzymes including methyltransferases (writers), demethylases (erasers), and m1A-recognizing proteins (readers). This modification significantly impacts the function of RNA and various biological processes by affecting the structure, stability, translation, metabolism, and gene expression of RNA. Thereby, m1A modification is closely associated with the occurrence and progression of cancer. This review aims to explore the role of m1A modification in tumor immunity. m1A affects tumor immune responses by directly regulating immune cells and indirectly modulating tumor microenvironment. Besides, we also discuss the implications of m1A-mediated metabolic reprogramming and its nexus with immune checkpoint inhibitors, unveiling promising avenues for immunotherapeutic intervention. Additionally, the m1AScore, established based on the expression patterns of m1A modification, can be used to predict tumor prognosis and guide personalized therapy. Our review underscores the significance of m1A modification as a burgeoning frontier in cancer biology and immuno-oncology, with the potential to revolutionize cancer treatment strategies.
Collapse
Affiliation(s)
- Xinru Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xiaoqing Ma
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Siyu Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Minyan Fan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Chenying Jin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yushi Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Shaodong Wang
- Affiliated Nanjing Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Zhiying Wang
- Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Fei Meng
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Chengwan Zhang
- Department of Central Laboratory, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, Jiangsu, China
| | - Lin Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
22
|
Huang G, Ternes L, Lanciault C, MacPherson-Hawthorne K, Chang YH, Sears RC, Muschler JL. Suppression of dystroglycan function accompanies pancreatic acinar-to-ductal metaplasia and favours dysplasia development. J Pathol 2024; 264:411-422. [PMID: 39435649 PMCID: PMC11560643 DOI: 10.1002/path.6356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 09/04/2024] [Indexed: 10/23/2024]
Abstract
The basement membrane (BM) is among the predominant microenvironmental factors of normal epithelia and of precancerous epithelial lesions. Evidence suggests that the BM functions not only as a barrier to tumour invasion but also as an active tumour-suppressing signalling substrate during premalignancy. However, the molecular foundations of such mechanisms have not been elucidated. Here we explore potential tumour-suppressing functions of the BM during precancer evolution, focusing on the expression and function of the extracellular matrix receptor dystroglycan in the pancreas and pancreatic disease. We show that the dystroglycan protein is highly expressed in the acinar compartment of the normal pancreas but lower in the ductal compartment. Moreover, there is a strong suppression of dystroglycan protein expression with acinar-to-ductal metaplasia in chronic pancreatitis and in all stages of pancreatic precancer and cancer evolution, from acinar-to-ductal metaplasia to dysplasia to adenocarcinoma. The conditional knockout of dystroglycan in the murine pancreas produced little evidence of developmental or functional deficiency. However, conditional deletion of dystroglycan expression in the context of oncogenic Kras expression led to a clear acceleration of pancreatic disease evolution, including accelerated dysplasia development and decreased survival. These data establish dystroglycan as a suppressor of pancreatic dysplasia development and one that is muted in chronic pancreatitis and at the earliest stages of oncogene-induced transformation. We conclude that dystroglycan is an important mediator of the tumour-suppressing functions of the BM during precancer evolution and that reduced dystroglycan function increases cancer risk, highlighting the dynamics of cell-BM interactions as important determinants of early cancer progression. © 2024 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Ge Huang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Luke Ternes
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | | | | | - Young Hwan Chang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Rosalie C. Sears
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
| | - John L. Muschler
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
23
|
Samanta S, Sarkar S, Kayal K. Cancer model and its possible control-A Z-type control approach. MethodsX 2024; 13:102895. [PMID: 39687596 PMCID: PMC11647952 DOI: 10.1016/j.mex.2024.102895] [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: 05/16/2024] [Revised: 07/28/2024] [Accepted: 08/06/2024] [Indexed: 12/18/2024] Open
Abstract
This paper investigates the dynamics of a three-dimensional nonlinear cancer model involving interactions among cancer cells, normal cells, and immune cells. By performing a linear stability analysis of the equilibria and investigating the Hopf bifurcation in relation to the immune cell growth rate, we reveal the possibility of chaotic behavior when radiation is absent. However, with the appropriate implementation of radiotherapy, the cancer model demonstrates stable solutions, transitioning from chaotic oscillations through period-halving bifurcation. Additionally, we propose and examine an indirect Z-control mechanism within the cancer model. Our findings indicate that using the indirect Z-controller on the immune population successfully manages chaos and adjusts the cancer cell density to a desired level. Through extensive investigation, we demonstrate the robustness of the Z-controller in managing oscillations and provide insights into determining the minimum number of immune cells needed to achieve a predetermined cancer cell density. This study underscores the importance of control mechanisms in mitigating cancer progression and highlights the potential of Z-control for therapeutic intervention strategies.
Collapse
Affiliation(s)
- Sudip Samanta
- Department of Mathematics, Bankura University, Bankura 722155, India
| | - Sandip Sarkar
- Sri Aurobindo Vidyamandir, Chandannagar, Hooghly, West Bengal 712136, India
| | - Kaushik Kayal
- Agricultural and Ecological Research Unit, Indian Statistical Institute, 203, B. T. Road, Kolkata 700108, India
| |
Collapse
|
24
|
Da Silva André G, Labouesse C. Mechanobiology of 3D cell confinement and extracellular crowding. Biophys Rev 2024; 16:833-849. [PMID: 39830117 PMCID: PMC11735831 DOI: 10.1007/s12551-024-01244-z] [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: 07/27/2024] [Accepted: 09/30/2024] [Indexed: 01/22/2025] Open
Abstract
Cells and tissues are often under some level of confinement, imposed by the microenvironment and neighboring cells, meaning that there are limitations to cell size, volume changes, and fluid exchanges. 3D cell culture, increasingly used for both single cells and organoids, inherently impose levels of confinement absent in 2D systems. It is thus key to understand how different levels of confinement influences cell survival, cell function, and cell fate. It is well known that the mechanical properties of the microenvironment, such as stiffness and stress relaxation, are important in activating mechanosensitive pathways, and these are responsive to confinement conditions. In this review, we look at how low, intermediate, and high levels of confinement modulate the activation of known mechanobiology pathways, in single cells, organoids, and tumor spheroids, with a specific focus on 3D confinement in microwells, elastic, or viscoelastic scaffolds. In addition, a confining microenvironment can drastically limit cellular communication in both healthy and diseased tissues, due to extracellular crowding. We discuss potential implications of extracellular crowding on molecular transport, extracellular matrix deposition, and fluid transport. Understanding how cells sense and respond to various levels of confinement should inform the design of 3D engineered matrices that recapitulate the physical properties of tissues.
Collapse
Affiliation(s)
- Gabriela Da Silva André
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Céline Labouesse
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland
| |
Collapse
|
25
|
Lenart NA, Rao SS. Cell-cell interactions mediating primary and metastatic breast cancer dormancy. Cancer Metastasis Rev 2024; 44:6. [PMID: 39585533 DOI: 10.1007/s10555-024-10223-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 10/18/2024] [Indexed: 11/26/2024]
Abstract
Breast cancer remains one of the leading causes of death in women around the world. A majority of deaths from breast cancer occur due to cancer cells colonizing distant organ sites. When colonizing these distant organ sites, breast cancer cells have been known to enter into a state of dormancy for extended periods of time. However, the mechanisms that promote dormancy as well as dormant-to-proliferative switch are not fully understood. The tumor microenvironment plays a key role in mediating cancer cell phenotype including regulation of the dormant state. In this review, we highlight cell-cell interactions in the tumor microenvironment mediating breast cancer dormancy at the primary and metastatic sites. Specifically, we describe how immune cells from the lymphoid lineage, tumor-associated myeloid lineage cells, and stromal cells of non-hematopoietic origin as well as tissue resident stromal cells impact dormancy vs. proliferation in breast cancer cells as well as the associated mechanisms. In addition, we highlight the importance of developing model systems and the associated considerations that will be critical in unraveling the mechanisms that promote primary and metastatic breast cancer dormancy mediated via cell-cell interactions.
Collapse
Affiliation(s)
- Nicholas A Lenart
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, 35487-0203, USA
| | - Shreyas S Rao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, 35487-0203, USA.
| |
Collapse
|
26
|
Del Toro K, Sayaman R, Thi K, Licon-Munoz Y, Hines WC. Transcriptomic analysis of the 12 major human breast cell types reveals mechanisms of cell and tissue function. PLoS Biol 2024; 22:e3002820. [PMID: 39499736 PMCID: PMC11537416 DOI: 10.1371/journal.pbio.3002820] [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: 02/28/2024] [Accepted: 08/29/2024] [Indexed: 11/07/2024] Open
Abstract
A fundamental question in biology, central to our understanding of cancer and other pathologies, is determining how different cell types coordinate to form and maintain tissues. Recognizing the distinct features and capabilities of the cells that compose these tissues is critical. Unfortunately, the complexity of tissues often hinders our ability to distinguish between neighboring cell types and, in turn, scrutinize their transcriptomes and generate reliable and tractable cell models for studying their inherently different biologies. We have recently introduced a novel method that permits the identification and purification of the 12 cell types that compose the human breast-nearly all of which could be reliably propagated in the laboratory. Here, we explore the nature of these cell types. We sequence mRNAs from each purified population and investigate transcriptional patterns that reveal their distinguishing features. We describe the differentially expressed genes and enriched biological pathways that capture the essence of each cell type, and we highlight transcripts that display intriguing expression patterns. These data, analytic tools, and transcriptional analyses form a rich resource whose exploration provides remarkable insights into the inner workings of the cell types composing the breast, thus furthering our understanding of the rules governing normal cell and tissue function.
Collapse
Affiliation(s)
- Katelyn Del Toro
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Rosalyn Sayaman
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Kate Thi
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Yamhilette Licon-Munoz
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - William Curtis Hines
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, University of New Mexico, Albuquerque, New Mexico, United States of America
| |
Collapse
|
27
|
Wang Z, Wang F, Yang Y, Fan W, Wen L, Zhang D. Development of a nomogram-based model incorporating radiomic features from follow-up longitudinal lung CT images to distinguish invasive adenocarcinoma from benign lesions: a retrospective study. BMC Pulm Med 2024; 24:534. [PMID: 39455958 PMCID: PMC11515265 DOI: 10.1186/s12890-024-03360-8] [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: 07/05/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024] Open
Abstract
PURPOSE To develop and validate a radiomic model for differentiating pulmonary invasive adenocarcinomas from benign lesions based on follow-up longitudinal CT images. METHODS This is a retrospective study including 336 patients (161 with invasive adenocarcinomas and 175 with benign lesions) who underwent baseline (T0) and follow-up (T1) CT scans from January 2016 to June 2022. The patients were randomized in a 7:3 ratio into training and test sets. Radiomic features were extracted from lesion volumes of interest on longitudinal CT images at T0 and T1. Differences in radiomic features between T1 and T0 were defined as delta-radiomic features. Logistic regression was used to build models based on clinicoradiological (CR), T0, T1, and delta radiomic features and compute signatures. Finally, a nomogram based on the CR, T0, T1 and delta signatures was constructed. Model performance was evaluated for calibration, discrimination, and clinical utility. RESULTS The T1 radiomic model was superior to the other independent models. In the training set, it had an area under the curve (AUC) of 0.858), superior to the CR model (AUC 0.694), the T0 radiomic model (AUC 0.825), and the delta radiomic model (AUC 0.734). In the test set, it had an AUC of 0.817, again outperforming the CR model (AUC 0.578), the T0 radiomic model (AUC 0.789), and the delta radiomic model (AUC 0.647). The nomogram incorporating the CR, T0, T1 and delta signatures showed the best predictive performance in both the training (AUC: 0.906) and test sets (AUC: 0.856), and it exhibited excellent fit with calibration curves. Decision curve analysis provided additional validation of the clinical utility of the nomogram. CONCLUSION A nomogram utilizing radiomic features extracted from longitudinal CT images can enhance the discriminative capability between pulmonary invasive adenocarcinomas and benign lesions.
Collapse
Affiliation(s)
- Zhengming Wang
- Department of Radiology, XinQiao Hospital of Army Medical University, Chongqing, 400037, China
| | - Fei Wang
- Department of Radiology, XinQiao Hospital of Army Medical University, Chongqing, 400037, China
- Department of Medical imaging, Luzhou People's Hospital, Luzhou, 646000, China
| | - Yan Yang
- Department of Radiology, XinQiao Hospital of Army Medical University, Chongqing, 400037, China
| | - Weijie Fan
- Department of Radiology, XinQiao Hospital of Army Medical University, Chongqing, 400037, China
| | - Li Wen
- Department of Radiology, XinQiao Hospital of Army Medical University, Chongqing, 400037, China
| | - Dong Zhang
- Department of Radiology, XinQiao Hospital of Army Medical University, Chongqing, 400037, China.
| |
Collapse
|
28
|
Zhang T, Chen J, Yang H, Sun X, Ou Y, Wang Q, Edderkaoui M, Zheng S, Ren F, Tong Y, Hu R, Liu J, Gao Y, Pandol SJ, Han YP, Zheng X. Stromal softness confines pancreatic cancer growth through lysosomal-cathepsin mediated YAP1 degradation. Cell Mol Life Sci 2024; 81:442. [PMID: 39460766 PMCID: PMC11512982 DOI: 10.1007/s00018-024-05466-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 09/03/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024]
Abstract
The progression and malignancy of many tumors are associated with increased tissue stiffness. Conversely, the oncogenically transformed cells can be confined in soft stroma. Yet, the underlying mechanisms by which soft matrix confines tumorigenesis and metastasis remain elusive. Here, we show that pancreatic cancer cells are suppressed in the soft extracellular matrix, which is associated with YAP1 degradation through autophagic-lysosomal pathway rather than Hippo signal mediated proteasome pathway. In the soft stroma, PTEN is upregulated and activated, which consequently promotes lysosomal biogenesis, leading to the activation of cysteine-cathepsins for YAP1 degradation. In vitro, purified cathepsin L can directly digest YAP1 under acidic conditions. Lysosomal stress, either caused by chloroquine or overexpression of cystatin A/B, results in YAP1 accumulation and malignant transformation. Likewise, liver fibrosis induced stiffness can promote malignant potential in mice. Clinical data show that down-regulation of lysosomal biogenesis is associated with pancreatic fibrosis and stiffness, YAP1 accumulation, and poor prognosis in PDAC patients. Together, our findings suggest that soft stroma triggers lysosomal flux-mediated YAP1 degradation and induces cancer cell dormancy.
Collapse
Affiliation(s)
- Tianci Zhang
- Department of Endocrinology and Metabolism, Research Center for Islet Transplantation, West China Hospital, Sichuan University, Chengdu, China
- The Center for Growth, Metabolism and Aging, College of Life Sciences, The State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Jingjing Chen
- The Center for Growth, Metabolism and Aging, College of Life Sciences, The State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Huan Yang
- The Center for Growth, Metabolism and Aging, College of Life Sciences, The State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Xiaoyan Sun
- Department of Endocrinology and Metabolism, Research Center for Islet Transplantation, West China Hospital, Sichuan University, Chengdu, China
| | - Yiran Ou
- Department of Endocrinology and Metabolism, Research Center for Islet Transplantation, West China Hospital, Sichuan University, Chengdu, China
| | - Qiang Wang
- Cedars-Sinai Medical Center, Los Angeles, USA
| | | | - Sujun Zheng
- Beijing Youan Hospital, the Capital Medical University, Beijing, China
| | - Feng Ren
- Beijing Youan Hospital, the Capital Medical University, Beijing, China
| | - Ying Tong
- The Center for Growth, Metabolism and Aging, College of Life Sciences, The State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Richard Hu
- Olive View-UCLA Medical Center, Los Angeles, CA, USA
| | - Jiaye Liu
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yun Gao
- Department of Endocrinology and Metabolism, Research Center for Islet Transplantation, West China Hospital, Sichuan University, Chengdu, China
| | | | - Yuan-Ping Han
- The Center for Growth, Metabolism and Aging, College of Life Sciences, The State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Xiaofeng Zheng
- Department of Endocrinology and Metabolism, Research Center for Islet Transplantation, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
29
|
Esteller M, Dawson MA, Kadoch C, Rassool FV, Jones PA, Baylin SB. The Epigenetic Hallmarks of Cancer. Cancer Discov 2024; 14:1783-1809. [PMID: 39363741 DOI: 10.1158/2159-8290.cd-24-0296] [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: 02/29/2024] [Revised: 05/08/2024] [Accepted: 06/24/2024] [Indexed: 10/05/2024]
Abstract
Cancer is a complex disease in which several molecular and cellular pathways converge to foster the tumoral phenotype. Notably, in the latest iteration of the cancer hallmarks, "nonmutational epigenetic reprogramming" was newly added. However, epigenetics, much like genetics, is a broad scientific area that deserves further attention due to its multiple roles in cancer initiation, progression, and adaptive nature. Herein, we present a detailed examination of the epigenetic hallmarks affected in human cancer, elucidating the pathways and genes involved, and dissecting the disrupted landscapes for DNA methylation, histone modifications, and chromatin architecture that define the disease. Significance: Cancer is a disease characterized by constant evolution, spanning from its initial premalignant stages to the advanced invasive and disseminated stages. It is a pathology that is able to adapt and survive amidst hostile cellular microenvironments and diverse treatments implemented by medical professionals. The more fixed setup of the genetic structure cannot fully provide transformed cells with the tools to survive but the rapid and plastic nature of epigenetic changes is ready for the task. This review summarizes the epigenetic hallmarks that define the ecological success of cancer cells in our bodies.
Collapse
Affiliation(s)
- Manel Esteller
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
- Centro de Investigacion Biomedica en Red Cancer (CIBERONC), Madrid, Spain
- Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain
| | - Mark A Dawson
- Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, Australia
| | - Cigall Kadoch
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Feyruz V Rassool
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Peter A Jones
- Department of Epigenetics, Van Andel Institute, Grand Rapids, Michigan
| | - Stephen B Baylin
- Department of Epigenetics, Van Andel Institute, Grand Rapids, Michigan
- Department of Oncology, The Johns Hopkins School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| |
Collapse
|
30
|
Thi K, Del Toro K, Licon-Munoz Y, Sayaman RW, Hines WC. Comprehensive identification, isolation, and culture of human breast cell types. J Biol Chem 2024; 300:107637. [PMID: 39122004 PMCID: PMC11459906 DOI: 10.1016/j.jbc.2024.107637] [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: 02/08/2024] [Revised: 07/03/2024] [Accepted: 07/16/2024] [Indexed: 08/12/2024] Open
Abstract
Tissues are formed and shaped by cells of many different types and are orchestrated through countless interactions. Deciphering a tissue's biological complexity thus requires studying it at cell-level resolution, where molecular and biochemical features of different cell types can be explored and thoroughly dissected. Unfortunately, the lack of comprehensive methods to identify, isolate, and culture each cell type from many tissues has impeded progress. Here, we present a method for the breadth of cell types composing the human breast. Our goal has long been to understand the essence of each of these different breast cell types, to reveal the underlying biology explaining their intrinsic features, the consequences of interactions, and their contributions to the tissue. This biological exploration has required cell purification, deep-RNA sequencing, and a thorough dissection of the genes and pathways defining each cell type. While the molecular analysis is presented in an adjoining article, we present here an exhaustive cellular dissection of the human breast and explore its cellular composition and histological organization. Moreover, we introduce a novel FACS antibody panel and rigorous gating strategy capable of isolating each of the 12 major breast cell types to purity. Finally, we describe the creation of primary cell models from nearly every breast cell type-some the first of their kind-and submit these as critical tools for studying the dynamic cellular interactions within breast tissues and tumors. Together, this body of work delivers a unique perspective of the breast, revealing insights into its cellular, molecular, and biochemical composition.
Collapse
Affiliation(s)
- Kate Thi
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Katelyn Del Toro
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Yamhilette Licon-Munoz
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Rosalyn W Sayaman
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - William C Hines
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA.
| |
Collapse
|
31
|
Han Y, Chen B, Bian J, Kang R, Shang X. Cancerous time estimation for interpreting the evolution of lung adenocarcinoma. Brief Bioinform 2024; 25:bbae520. [PMID: 39413800 PMCID: PMC11483137 DOI: 10.1093/bib/bbae520] [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: 02/25/2024] [Revised: 08/19/2024] [Accepted: 10/02/2024] [Indexed: 10/18/2024] Open
Abstract
The evolution of lung adenocarcinoma is accompanied by a multitude of gene mutations and dysfunctions, rendering its phenotypic state and evolutionary direction highly complex. To interpret the evolution of lung adenocarcinoma, various methods have been developed to elucidate the molecular pathogenesis and functional evolution processes. However, most of these methods are constrained by the absence of cancerous temporal information, and the challenges of heterogeneous characteristics. To handle these problems, in this study, a patient quasi-potential landscape method was proposed to estimate the cancerous time of phenotypic states' emergence during the evolutionary process. Subsequently, a total of 39 different oncogenetic paths were identified based on cancerous time and mutations, reflecting the molecular pathogenesis of the evolutionary process of lung adenocarcinoma. To interpret the evolution patterns of lung adenocarcinoma, three oncogenetic graphs were obtained as the common evolutionary patterns by merging the oncogenetic paths. Moreover, patients were evenly re-divided into early, middle, and late evolutionary stages according to cancerous time, and a feasible framework was developed to construct the functional evolution network of lung adenocarcinoma. A total of six significant functional evolution processes were identified from the functional evolution network based on the pathway enrichment analysis, which plays critical roles in understanding the development of lung adenocarcinoma.
Collapse
Affiliation(s)
- Yourui Han
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710012, China
| | - Bolin Chen
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710012, China
- Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University, Ministry of Industry and Information Technology, Xi’an 710012, China
| | - Jun Bian
- Department of General Surgery, Xi’an Children’s Hospital, Xi’an Jiaotong University Affiliated Children’s Hospital, Xi’an 710003, China
| | - Ruiming Kang
- Rewise (Hangzhou) Information Technology Co., LTD, Hangzhou 310000, China
| | - Xuequn Shang
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710012, China
- Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University, Ministry of Industry and Information Technology, Xi’an 710012, China
| |
Collapse
|
32
|
Zhang F, Guo J, Yu S, Zheng Y, Duan M, Zhao L, Wang Y, Yang Z, Jiang X. Cellular senescence and metabolic reprogramming: Unraveling the intricate crosstalk in the immunosuppressive tumor microenvironment. Cancer Commun (Lond) 2024; 44:929-966. [PMID: 38997794 PMCID: PMC11492308 DOI: 10.1002/cac2.12591] [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: 11/26/2023] [Revised: 06/23/2024] [Accepted: 07/07/2024] [Indexed: 07/14/2024] Open
Abstract
The intrinsic oncogenic mechanisms and properties of the tumor microenvironment (TME) have been extensively investigated. Primary features of the TME include metabolic reprogramming, hypoxia, chronic inflammation, and tumor immunosuppression. Previous studies suggest that senescence-associated secretory phenotypes that mediate intercellular information exchange play a role in the dynamic evolution of the TME. Specifically, hypoxic adaptation, metabolic dysregulation, and phenotypic shifts in immune cells regulated by cellular senescence synergistically contribute to the development of an immunosuppressive microenvironment and chronic inflammation, thereby promoting the progression of tumor events. This review provides a comprehensive summary of the processes by which cellular senescence regulates the dynamic evolution of the tumor-adapted TME, with focus on the complex mechanisms underlying the relationship between senescence and changes in the biological functions of tumor cells. The available findings suggest that components of the TME collectively contribute to the progression of tumor events. The potential applications and challenges of targeted cellular senescence-based and combination therapies in clinical settings are further discussed within the context of advancing cellular senescence-related research.
Collapse
Affiliation(s)
- Fusheng Zhang
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
- Department of Hepatobiliary and Pancreatic SurgeryPeking University First HospitalBeijingP. R. China
| | - Junchen Guo
- Department of RadiologyThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Shengmiao Yu
- Outpatient DepartmentThe Fourth Affiliated HospitalChina Medical UniversityShenyangLiaoningP. R. China
| | - Youwei Zheng
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Meiqi Duan
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Liang Zhao
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Yihan Wang
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Zhi Yang
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Xiaofeng Jiang
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| |
Collapse
|
33
|
Parihar K, Ko SHB, Bradley RP, Taylor P, Ramakrishnan N, Baumgart T, Guo W, Weaver VM, Janmey PA, Radhakrishnan R. Asymmetric crowders and membrane morphology at the nexus of intracellular trafficking and oncology. MECHANOBIOLOGY IN MEDICINE 2024; 2:100071. [PMID: 38899029 PMCID: PMC11185830 DOI: 10.1016/j.mbm.2024.100071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
A definitive understanding of the interplay between protein binding/migration and membrane curvature evolution is emerging but needs further study. The mechanisms defining such phenomena are critical to intracellular transport and trafficking of proteins. Among trafficking modalities, exosomes have drawn attention in cancer research as these nano-sized naturally occurring vehicles are implicated in intercellular communication in the tumor microenvironment, suppressing anti-tumor immunity and preparing the metastatic niche for progression. A significant question in the field is how the release and composition of tumor exosomes are regulated. In this perspective article, we explore how physical factors such as geometry and tissue mechanics regulate cell cortical tension to influence exosome production by co-opting the biophysics as well as the signaling dynamics of intracellular trafficking pathways and how these exosomes contribute to the suppression of anti-tumor immunity and promote metastasis. We describe a multiscale modeling approach whose impact goes beyond the fundamental investigation of specific cellular processes toward actual clinical translation. Exosomal mechanisms are critical to developing and approving liquid biopsy technologies, poised to transform future non-invasive, longitudinal profiling of evolving tumors and resistance to cancer therapies to bring us one step closer to the promise of personalized medicine.
Collapse
Affiliation(s)
- Kshitiz Parihar
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Seung-Hyun B. Ko
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan P. Bradley
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Phillip Taylor
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - N. Ramakrishnan
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Tobias Baumgart
- Department of Chemistry, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei Guo
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Valerie M. Weaver
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
| | - Paul A. Janmey
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ravi Radhakrishnan
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
34
|
Suh YJ, Li AT, Pandey M, Nordmann CS, Huang YL, Wu M. Decoding physical principles of cell migration under controlled environment using microfluidics. BIOPHYSICS REVIEWS 2024; 5:031302. [PMID: 39091432 PMCID: PMC11290890 DOI: 10.1063/5.0199161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 06/26/2024] [Indexed: 08/04/2024]
Abstract
Living cells can perform incredible tasks that man-made micro/nano-sized robots have not yet been able to accomplish. One example is that white blood cells can sense and move to the site of pathogen attack within minutes. The robustness and precision of cellular functions have been perfected through billions of years of evolution. In this context, we ask the question whether cells follow a set of physical principles to sense, adapt, and migrate. Microfluidics has emerged as an enabling technology for recreating well-defined cellular environment for cell migration studies, and its ability to follow single cell dynamics allows for the results to be amenable for theoretical modeling. In this review, we focus on the development of microfluidic platforms for recreating cellular biophysical (e.g., mechanical stress) and biochemical (e.g., nutrients and cytokines) environments for cell migration studies in 3D. We summarize the basic principles that cells (including bacteria, algal, and mammalian cells) use to respond to chemical gradients learned from microfluidic systems. We also discuss about novel biological insights gained from studies of cell migration under biophysical cues and the need for further quantitative studies of cell function under well-controlled biophysical environments in the future.
Collapse
Affiliation(s)
- Young Joon Suh
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Alan T. Li
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Mrinal Pandey
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Cassidy S. Nordmann
- Department of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Yu Ling Huang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Mingming Wu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
35
|
Younesi FS, Hinz B. The Myofibroblast Fate of Therapeutic Mesenchymal Stromal Cells: Regeneration, Repair, or Despair? Int J Mol Sci 2024; 25:8712. [PMID: 39201399 PMCID: PMC11354465 DOI: 10.3390/ijms25168712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) can be isolated from various tissues of healthy or patient donors to be retransplanted in cell therapies. Because the number of MSCs obtained from biopsies is typically too low for direct clinical application, MSC expansion in cell culture is required. However, ex vivo amplification often reduces the desired MSC regenerative potential and enhances undesired traits, such as activation into fibrogenic myofibroblasts. Transiently activated myofibroblasts restore tissue integrity after organ injury by producing and contracting extracellular matrix into scar tissue. In contrast, persistent myofibroblasts cause excessive scarring-called fibrosis-that destroys organ function. In this review, we focus on the relevance and molecular mechanisms of myofibroblast activation upon contact with stiff cell culture plastic or recipient scar tissue, such as hypertrophic scars of large skin burns. We discuss cell mechanoperception mechanisms such as integrins and stretch-activated channels, mechanotransduction through the contractile actin cytoskeleton, and conversion of mechanical signals into transcriptional programs via mechanosensitive co-transcription factors, such as YAP, TAZ, and MRTF. We further elaborate how prolonged mechanical stress can create persistent myofibroblast memory by direct mechanotransduction to the nucleus that can evoke lasting epigenetic modifications at the DNA level, such as histone methylation and acetylation. We conclude by projecting how cell culture mechanics can be modulated to generate MSCs, which epigenetically protected against myofibroblast activation and transport desired regeneration potential to the recipient tissue environment in clinical therapies.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
36
|
Wang X, Yang Y, Zhao S, Wu D, Li L, Zhao Z. Chitosan-based biomaterial delivery strategies for hepatocellular carcinoma. Front Pharmacol 2024; 15:1446030. [PMID: 39161903 PMCID: PMC11330802 DOI: 10.3389/fphar.2024.1446030] [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: 06/08/2024] [Accepted: 07/23/2024] [Indexed: 08/21/2024] Open
Abstract
Background Hepatocellular carcinoma accounts for 80% of primary liver cancers, is the most common primary liver malignancy. Hepatocellular carcinoma is the third leading cause of tumor-related deaths worldwide, with a 5-year survival rate of approximately 18%. Chemotherapy, although commonly used for hepatocellular carcinoma treatment, is limited by systemic toxicity and drug resistance. Improving targeted delivery of chemotherapy drugs to tumor cells without causing systemic side effects is a current research focus. Chitosan, a biopolymer derived from chitin, possesses good biocompatibility and biodegradability, making it suitable for drug delivery. Enhanced chitosan formulations retain the anti-tumor properties while improving stability. Chitosan-based biomaterials promote hepatocellular carcinoma apoptosis, exhibit antioxidant and anti-inflammatory effects, inhibit tumor angiogenesis, and improve extracellular matrix remodeling for enhanced anti-tumor therapy. Methods We summarized published experimental papers by querying them. Results and Conclusions This review discusses the physicochemical properties of chitosan, its application in hepatocellular carcinoma treatment, and the challenges faced by chitosan-based biomaterials.
Collapse
Affiliation(s)
- Xianling Wang
- Department of Gastroenterology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yan Yang
- Department of Gastroenterology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Shuang Zhao
- Endoscopy Center, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Di Wu
- First Digestive Endoscopy Department, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Le Li
- Department of Gastroenterology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhifeng Zhao
- Department of Gastroenterology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
37
|
Zhang Y, Li J, Feng L, Cheng Y, Shi L, Yang Q, Wang Z, Yi X, Zhong G, Sun X, Cheng Z, Guo M. STAC3 as a poor prognostic biomarker in renal clear cell carcinoma: relationship with immune infiltration. Am J Cancer Res 2024; 14:3294-3316. [PMID: 39113874 PMCID: PMC11301302 DOI: 10.62347/eaqw3113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 06/17/2024] [Indexed: 08/10/2024] Open
Abstract
Calcium ions (Ca2+) are crucial in tumorigenesis and progression, with their elevated levels indicating a negative prognosis in Kidney Renal Clear Cell Carcinoma (KIRC). The influence of genes regulating calcium ions on the survival outcomes of KIRC patients and their interaction with the tumor's immune microenvironment is yet to be fully understood. This study analyzed gene expression data from KIRC tumor and adjacent non-tumor tissues using the TCGA-KIRC dataset to pinpoint genes that are differentially expressed in KIRC. Intersection of these genes with those regulating calcium ions highlighted specific calcium ion-regulating genes that exhibit differential expression in KIRC. Subsequently, prognostic risk models were developed using univariate Cox and LASSO-Cox regression analyses to verify their diagnostic precision. Additionally, the study investigated the correlation between tumor immunity and KIRC patient outcomes, assessing the contribution of STAC3 genes to tumor immunity. Further exploration entailed SSGASE, single-cell analysis, pseudotime analysis and both in vivo and in vitro experiments to evaluate STAC3's role in tumor immunity and progression. Notably, STAC3 was significantly overexpressed in tumor specimens and positively correlated with the degree of malignancy of KIRC, affecting patients' prognosis. Elevated STAC3 expression correlated with enhanced immune infiltration in KIRC tumors. Furthermore, silencing STAC3 curtailed KIRC cell proliferation, migration, invasion, and stemness properties. Experimental models in mice confirmed that STAC3 knockdown led to a reduction in tumor growth. Elevated STAC3 expression is intricately linked with immune infiltration in KIRC tumors, as well as with the aggressive biological behaviors of tumor cells, including their proliferation, migration, and invasion. Targeting STAC3 presents a promising strategy to augment the efficacy of current therapeutic approaches and to better the survival outcomes of patients with KIRC.
Collapse
Affiliation(s)
- Yingnan Zhang
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Jingtao Li
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Luwen Feng
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Yue Cheng
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Linlin Shi
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Qian Yang
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Zhaoyang Wang
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Xuan Yi
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Guocai Zhong
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Xueying Sun
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Zhifeng Cheng
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Min Guo
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| |
Collapse
|
38
|
Zhang Y, Wang C, Li JJ. Revisiting the role of mesenchymal stromal cells in cancer initiation, metastasis and immunosuppression. Exp Hematol Oncol 2024; 13:64. [PMID: 38951845 PMCID: PMC11218091 DOI: 10.1186/s40164-024-00532-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024] Open
Abstract
Immune checkpoint blockade (ICB) necessitates a thorough understanding of intricate cellular interactions within the tumor microenvironment (TME). Mesenchymal stromal cells (MSCs) play a pivotal role in cancer generation, progression, and immunosuppressive tumor microenvironment. Within the TME, MSCs encompass both resident and circulating counterparts that dynamically communicate and actively participate in TME immunosurveillance and response to ICB. This review aims to reevaluate various facets of MSCs, including their potential self-transformation to function as cancer-initiating cells and contributions to the creation of a conducive environment for tumor proliferation and metastasis. Additionally, we explore the immune regulatory functions of tumor-associated MSCs (TA-MSCs) and MSC-derived extracellular vesicles (MSC-EVs) with analysis of potential connections between circulating and tissue-resident MSCs. A comprehensive understanding of the dynamics of MSC-immune cell communication and the heterogeneous cargo of tumor-educated versus naïve MSCs may unveil a new MSC-mediated immunosuppressive pathway that can be targeted to enhance cancer control by ICB.
Collapse
Affiliation(s)
- Yanyan Zhang
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Radiation Oncology, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Charles Wang
- Department of Radiation Oncology, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Jian Jian Li
- Department of Radiation Oncology, School of Medicine, University of California Davis, Sacramento, CA, USA.
- NCI-Designated Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA.
| |
Collapse
|
39
|
Adler FR. A modelling framework for cancer ecology and evolution. J R Soc Interface 2024; 21:20240099. [PMID: 39013418 PMCID: PMC11251767 DOI: 10.1098/rsif.2024.0099] [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: 02/08/2024] [Accepted: 05/10/2024] [Indexed: 07/18/2024] Open
Abstract
Cancer incidence increases rapidly with age, typically as a polynomial. The somatic mutation theory explains this increase through the waiting time for enough mutations to build up to generate cells with the full set of traits needed to grow without control. However, lines of evidence ranging from tumour reversion and dormancy to the prevalence of presumed cancer mutations in non-cancerous tissues argue that this is not the whole story, and that cancer is also an ecological process, and that mutations only lead to cancer when the systems of control within and across cells have broken down. Aging thus has two effects: the build-up of mutations and the breakdown of control. This paper presents a mathematical modelling framework to unify these theories with novel approaches to model the mutation and diversification of cell lineages and of the breakdown of the layers of control both within and between cells. These models correctly predict the polynomial increase of cancer with age, show how germline defects in control accelerate cancer initiation, and compute how the positive feedback between cell replication, ecology and layers of control leads to a doubly exponential growth of cell populations.
Collapse
Affiliation(s)
- Frederick R. Adler
- Department of Mathematics, School of Biological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
40
|
Pan Y, Cheng J, Zhu Y, Zhang J, Fan W, Chen X. Immunological nanomaterials to combat cancer metastasis. Chem Soc Rev 2024; 53:6399-6444. [PMID: 38745455 DOI: 10.1039/d2cs00968d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Metastasis causes greater than 90% of cancer-associated deaths, presenting huge challenges for detection and efficient treatment of cancer due to its high heterogeneity and widespread dissemination to various organs. Therefore, it is imperative to combat cancer metastasis, which is the key to achieving complete cancer eradication. Immunotherapy as a systemic approach has shown promising potential to combat metastasis. However, current clinical immunotherapies are not effective for all patients or all types of cancer metastases owing to insufficient immune responses. In recent years, immunological nanomaterials with intrinsic immunogenicity or immunomodulatory agents with efficient loading have been shown to enhance immune responses to eliminate metastasis. In this review, we would like to summarize various types of immunological nanomaterials against metastasis. Moreover, this review will summarize a series of immunological nanomaterial-mediated immunotherapy strategies to combat metastasis, including immunogenic cell death, regulation of chemokines and cytokines, improving the immunosuppressive tumour microenvironment, activation of the STING pathway, enhancing cytotoxic natural killer cell activity, enhancing antigen presentation of dendritic cells, and enhancing chimeric antigen receptor T cell therapy. Furthermore, the synergistic anti-metastasis strategies based on the combinational use of immunotherapy and other therapeutic modalities will also be introduced. In addition, the nanomaterial-mediated imaging techniques (e.g., optical imaging, magnetic resonance imaging, computed tomography, photoacoustic imaging, surface-enhanced Raman scattering, radionuclide imaging, etc.) for detecting metastasis and monitoring anti-metastasis efficacy are also summarized. Finally, the current challenges and future prospects of immunological nanomaterial-based anti-metastasis are also elucidated with the intention to accelerate its clinical translation.
Collapse
Affiliation(s)
- Yuanbo Pan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Junjie Cheng
- Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Yang Zhu
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, Fujian, China.
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 211198, China.
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
| |
Collapse
|
41
|
Vela-Alcántara AM, Santiago-García J, Barragán-Palacios M, León-Chacón A, Domínguez-Pantoja M, Barceinas-Dávila I, Juárez-Aguilar E, Tamariz E. Differential modulation of cell morphology, migration, and Neuropilin-1 expression in cancer and non-cancer cell lines by substrate stiffness. Front Cell Dev Biol 2024; 12:1352233. [PMID: 38903533 PMCID: PMC11188430 DOI: 10.3389/fcell.2024.1352233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/10/2024] [Indexed: 06/22/2024] Open
Abstract
Physical changes in the tumor microenvironment, such as increased stiffness, regulate cancer hallmarks and play an essential role in gene expression, cell morphology, migration, and malignancy. However, the response of cancer cells to stiffness is not homogeneous and varies depending on the cell type and its mechanosensitivity. In this study, we investigated the differential responses of cervical (HeLa) and prostate (PC-3) cancer cell lines, as well as non-tumoral cell lines (HEK293 and HPrEC), to stiffness using polyacrylamide hydrogels mimicking normal and tumoral tissues. We analyzed cell morphology, migration, and the expression of neuropilin 1 (NRP1), a receptor involved in angiogenesis, cell migration, and extracellular matrix remodeling, known to be associated with cancer progression and poor prognosis. Our findings reveal that NRP1 expression increases on substrates mimicking the high stiffness characteristic of tumoral tissue in the non-tumoral cell lines HPrEC and HEK293. Conversely, in tumoral PC-3 cells, stiffness resembling normal prostate tissue induces an earlier and more sustained expression of NRP1. Furthermore, we observed that stiffness influences cell spreading, pseudopodia formation, and the mode of cell protrusion during migration. Soft substrates predominantly trigger bleb cell protrusion, while pseudopodia protrusions increase on substrates mimicking normal and tumor-like stiffnesses in HPrEC cells compared to PC-3 cells. Stiffer substrates also enhance the percentage of migratory cells, as well as their velocity and total displacement, in both non-tumoral and tumoral prostate cells. However, they only improve the persistence of migration in tumoral PC-3 cells. Moreover, we found that NRP1 co-localizes with actin, and its suppression impairs tumoral PC-3 spreading while decreasing pseudopodia protrusion mode. Our results suggest that the modulation of NRP1 expression by the stiffness can be a feedback loop to promote malignancy in non-tumoral and cancer cells, contingent upon the mechanosensitivity of the cells.
Collapse
Affiliation(s)
- Ana Monserrat Vela-Alcántara
- Programa de Doctorado en Ciencias de la Salud, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
- Laboratorio de Cultivo Celular, Departamento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
| | - Juan Santiago-García
- Laboratorio de Biología Molecular, Instituto de Investigaciones Biológicas, Universidad Veracruzana, Xalapa, Mexico
| | - Madeleine Barragán-Palacios
- Laboratorio de Cultivo Celular, Departamento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
- Programa de Maestría en Ciencias de la Salud, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
| | - Aylin León-Chacón
- Laboratorio de Cultivo Celular, Departamento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
| | | | - Irene Barceinas-Dávila
- Laboratorio de Cultivo Celular, Departamento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
- Programa de Maestría en Ciencias de la Salud, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
| | - Enrique Juárez-Aguilar
- Laboratorio de Cultivo Celular, Departamento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
| | - Elisa Tamariz
- Laboratorio de Cultivo Celular, Departamento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
| |
Collapse
|
42
|
Xin T, Gallini S, Wei H, Gonzalez DG, Matte-Martone C, Machida H, Fujiwara H, Pasolli HA, Suozzi KC, Gonzalez LE, Regot S, Greco V. Oncogenic Kras induces spatiotemporally specific tissue deformation through converting pulsatile into sustained ERK activation. Nat Cell Biol 2024; 26:859-867. [PMID: 38689013 PMCID: PMC11519783 DOI: 10.1038/s41556-024-01413-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/28/2024] [Indexed: 05/02/2024]
Abstract
Tissue regeneration and maintenance rely on coordinated stem cell behaviours. This orchestration can be impaired by oncogenic mutations leading to cancer. However, it is largely unclear how oncogenes perturb stem cells' orchestration to disrupt tissue. Here we used intravital imaging to investigate the mechanisms by which oncogenic Kras mutation causes tissue disruption in the hair follicle. Through longitudinally tracking hair follicles in live mice, we found that KrasG12D, a mutation that can lead to squamous cell carcinoma, induces epithelial tissue deformation in a spatiotemporally specific manner, linked with abnormal cell division and migration. Using a reporter mouse capture real-time ERK signal dynamics at the single-cell level, we discovered that KrasG12D, but not a closely related mutation HrasG12V, converts ERK signal in stem cells from pulsatile to sustained. Finally, we demonstrated that interrupting sustained ERK signal reverts KrasG12D-induced tissue deformation through modulating specific features of cell migration and division.
Collapse
Affiliation(s)
- Tianchi Xin
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
| | - Sara Gallini
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Haoyang Wei
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - David G Gonzalez
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | | | - Hiroki Machida
- Laboratory for Tissue Microenvironment, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hironobu Fujiwara
- Laboratory for Tissue Microenvironment, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Graduate School of Medicine, Osaka University, Suita, Japan
| | - H Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller University, New York, NY, USA
| | - Kathleen C Suozzi
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Lauren E Gonzalez
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Sergi Regot
- Department of Molecular Biology and Genetics, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Valentina Greco
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA.
- Department of Cell Biology, Yale Stem Cell Center, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
43
|
Reynolds LE, Maallin S, Haston S, Martinez-Barbera JP, Hodivala-Dilke KM, Pedrosa AR. Effects of senescence on the tumour microenvironment and response to therapy. FEBS J 2024; 291:2306-2319. [PMID: 37873605 DOI: 10.1111/febs.16984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/04/2023] [Accepted: 10/19/2023] [Indexed: 10/25/2023]
Abstract
Cellular senescence is a state of durable cell arrest that has been identified both in vitro and in vivo. It is associated with profound changes in gene expression and a specific secretory profile that includes pro-inflammatory cytokines, growth factors and matrix-remodelling enzymes, referred to as the senescence-associated secretory phenotype (SASP). In cancer, senescence can have anti- or pro-tumour effects. On one hand, it can inhibit tumour progression in a cell autonomous manner. On the other hand, senescence can also promote tumour initiation, progression, metastatic dissemination and resistance to therapy in a paracrine manner. Therefore, despite efforts to target senescence as a potential strategy to inhibit tumour growth, senescent cancer and microenvironmental cells can eventually lead to uncontrolled proliferation and aggressive tumour phenotypes. This can happen either through overcoming senescence growth arrest or through SASP-mediated effects in adjacent tumour cells. This review will discuss how senescence affects the tumour microenvironment, including extracellular matrix remodelling, the immune system and the vascular compartment, to promote tumourigenesis, metastasis and resistance to DNA-damaging therapies. It will also discuss current approaches used in the field to target senescence: senolytics, improving the immune clearance of senescent cells and targeting the SASP.
Collapse
Affiliation(s)
- Louise E Reynolds
- Adhesion and Angiogenesis Lab, Centre for Tumour Microenvironment, Barts Cancer Institute, John Vane Science Centre, Queen Mary University London, UK
| | - Seynab Maallin
- Adhesion and Angiogenesis Lab, Centre for Tumour Microenvironment, Barts Cancer Institute, John Vane Science Centre, Queen Mary University London, UK
| | - Scott Haston
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London, UK
| | - Juan Pedro Martinez-Barbera
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London, UK
| | - Kairbaan M Hodivala-Dilke
- Adhesion and Angiogenesis Lab, Centre for Tumour Microenvironment, Barts Cancer Institute, John Vane Science Centre, Queen Mary University London, UK
| | - Ana-Rita Pedrosa
- Adhesion and Angiogenesis Lab, Centre for Tumour Microenvironment, Barts Cancer Institute, John Vane Science Centre, Queen Mary University London, UK
| |
Collapse
|
44
|
Pio-Lopez L, Levin M. Aging as a loss of morphostatic information: A developmental bioelectricity perspective. Ageing Res Rev 2024; 97:102310. [PMID: 38636560 DOI: 10.1016/j.arr.2024.102310] [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: 11/05/2023] [Revised: 02/21/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024]
Abstract
Maintaining order at the tissue level is crucial throughout the lifespan, as failure can lead to cancer and an accumulation of molecular and cellular disorders. Perhaps, the most consistent and pervasive result of these failures is aging, which is characterized by the progressive loss of function and decline in the ability to maintain anatomical homeostasis and reproduce. This leads to organ malfunction, diseases, and ultimately death. The traditional understanding of aging is that it is caused by the accumulation of molecular and cellular damage. In this article, we propose a complementary view of aging from the perspective of endogenous bioelectricity which has not yet been integrated into aging research. We propose a view of aging as a morphostasis defect, a loss of biophysical prepattern information, encoding anatomical setpoints used for dynamic tissue and organ homeostasis. We hypothesize that this is specifically driven by abrogation of the endogenous bioelectric signaling that normally harnesses individual cell behaviors toward the creation and upkeep of complex multicellular structures in vivo. Herein, we first describe bioelectricity as the physiological software of life, and then identify and discuss the links between bioelectricity and life extension strategies and age-related diseases. We develop a bridge between aging and regeneration via bioelectric signaling that suggests a research program for healthful longevity via morphoceuticals. Finally, we discuss the broader implications of the homologies between development, aging, cancer and regeneration and how morphoceuticals can be developed for aging.
Collapse
Affiliation(s)
- Léo Pio-Lopez
- Allen Discovery Center, Tufts University, Medford, MA 02155, USA
| | - Michael Levin
- Allen Discovery Center, Tufts University, Medford, MA 02155, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA.
| |
Collapse
|
45
|
Shi J, Zhu L, Tang BY, Yang WQ, Xi SY, Zhang CL, Li PF, Wang YJ, Guo KH, Huang JR, Huang CR, Yu ZX, Yu BK, Zhang CF, Zhang YM. Regulatory effect of Yinchenhao decoction on bile acid metabolism to improve the inflammatory microenvironment of hepatocellular carcinoma in mice. J Nat Med 2024; 78:633-643. [PMID: 38704807 DOI: 10.1007/s11418-024-01812-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 04/03/2024] [Indexed: 05/07/2024]
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor with extremely high mortality. The tumor microenvironment is the "soil" of its occurrence and development, and the inflammatory microenvironment is an important part of the "soil". Bile acid is closely related to the occurrence of HCC. Bile acid metabolism disorder is not only directly involved in the occurrence and development of HCC but also affects the inflammatory microenvironment of HCC. Yinchenhao decoction, a traditional Chinese medicine formula, can regulate bile acid metabolism and may affect the inflammatory microenvironment of HCC. To determine the effect of Yinchenhao decoction on bile acid metabolism in mice with HCC and to explore the possible mechanism by which Yinchenhao decoction improves the inflammatory microenvironment of HCC by regulating bile acid metabolism, we established mice model of orthotopic transplantation of hepatocellular carcinoma. These mice were treated with three doses of Yinchenhao decoction, then liver samples were collected and tested. Yinchenhao decoction can regulate the disorder of bile acid metabolism in liver cancer mice. Besides, it can improve inflammatory reactions, reduce hepatocyte degeneration and necrosis, and even reduce liver weight and the liver index. Taurochenodeoxycholic acid, hyodeoxycholic acid, and taurohyodeoxycholic acid are important molecules in the regulation of the liver inflammatory microenvironment, laying a foundation for the regulation of the liver tumor inflammatory microenvironment based on bile acids. Yinchenhao decoction may improve the inflammatory microenvironment of mice with HCC by ameliorating hepatic bile acid metabolism.
Collapse
Affiliation(s)
- Jian Shi
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Lin Zhu
- Department of Traditional Chinese Medicine, Binhai County People's Hospital, Yancheng, China
| | - Bang-Yi Tang
- Department of Science and Education, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, China
| | - Wan-Qing Yang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Sheng-Yan Xi
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Chen-Long Zhang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
- Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Peng-Fei Li
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
- Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Yu-Jie Wang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
- Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Kai-Hang Guo
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
- Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, Xiamen, China
| | - Jing-Ru Huang
- Central Laboratory, School of Medicine, Xiamen University, Xiamen, China
| | - Chen-Rui Huang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Zhou-Xin Yu
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Bao-Kang Yu
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Chun-Fang Zhang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China.
- Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, Xiamen, China.
| | - Yu-Mei Zhang
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, China.
- Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, Xiamen, China.
| |
Collapse
|
46
|
Sánchez-Ramírez D, Mendoza-Rodríguez MG, Alemán OR, Candanedo-González FA, Rodríguez-Sosa M, Montesinos-Montesinos JJ, Salcedo M, Brito-Toledo I, Vaca-Paniagua F, Terrazas LI. Impact of STAT-signaling pathway on cancer-associated fibroblasts in colorectal cancer and its role in immunosuppression. World J Gastrointest Oncol 2024; 16:1705-1724. [PMID: 38764833 PMCID: PMC11099434 DOI: 10.4251/wjgo.v16.i5.1705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/28/2024] [Accepted: 04/01/2024] [Indexed: 05/09/2024] Open
Abstract
Colorectal cancer (CRC) remains one of the most commonly diagnosed and deadliest types of cancer worldwide. CRC displays a desmoplastic reaction (DR) that has been inversely associated with poor prognosis; less DR is associated with a better prognosis. This reaction generates excessive connective tissue, in which cancer-associated fibroblasts (CAFs) are critical cells that form a part of the tumor microenvironment. CAFs are directly involved in tumorigenesis through different mechanisms. However, their role in immunosuppression in CRC is not well understood, and the precise role of signal transducers and activators of transcription (STATs) in mediating CAF activity in CRC remains unclear. Among the myriad chemical and biological factors that affect CAFs, different cytokines mediate their function by activating STAT signaling pathways. Thus, the harmful effects of CAFs in favoring tumor growth and invasion may be modulated using STAT inhibitors. Here, we analyze the impact of different STATs on CAF activity and their immunoregulatory role.
Collapse
Affiliation(s)
- Damián Sánchez-Ramírez
- Unidad de Investigacion en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autonoma de Mexico, Tlalnepantla 54090, Estado de Mexico, Mexico
| | - Mónica G Mendoza-Rodríguez
- Unidad de Investigacion en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autonoma de Mexico, Tlalnepantla 54090, Estado de Mexico, Mexico
| | - Omar R Alemán
- Department of Biology, Facultad de Quimica, Universidad Nacional Autonoma de Mexico, Ciudad Universitaria, Mexico City 04510, Mexico
| | - Fernando A Candanedo-González
- Department of Pathology, National Medical Center Century XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Miriam Rodríguez-Sosa
- Unidad de Investigacion en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autonoma de Mexico, Tlalnepantla 54090, Estado de Mexico, Mexico
| | - Juan José Montesinos-Montesinos
- Laboratorio de Células Troncales Mesenquimales, Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncología Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Mauricio Salcedo
- Unidad de Investigacion en Biomedicina y Oncologia Genomica, Instituto Mexciano del Seguro Social, Mexico City 07300, Mexico
| | - Ismael Brito-Toledo
- Servicio de Colon y Recto, Hospital de Oncología Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Felipe Vaca-Paniagua
- Unidad de Investigacion en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autonoma de Mexico, Tlalnepantla 54090, Estado de Mexico, Mexico
- Laboratorio Nacional en Salud, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de Mexico, Mexico
| | - Luis I Terrazas
- Unidad de Investigacion en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autonoma de Mexico, Tlalnepantla 54090, Estado de Mexico, Mexico
- Laboratorio Nacional en Salud, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de Mexico, Mexico
| |
Collapse
|
47
|
Bamodu OA, Chung CC, Pisanic TR, Wu ATH. The intricate interplay between cancer stem cells and cell-of-origin of cancer: implications for therapeutic strategies. Front Oncol 2024; 14:1404628. [PMID: 38800385 PMCID: PMC11116576 DOI: 10.3389/fonc.2024.1404628] [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: 03/21/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
Background Cancer stem cells (CSCs) have emerged as pivotal players in tumorigenesis, disease progression, and resistance to therapies. Objective This comprehensive review delves into the intricate relationship between CSCs and the cell-of-origin in diverse cancer types. Design Comprehensive review of thematically-relevant literature. Methods We explore the underlying molecular mechanisms that drive the conversion of normal cells into CSCs and the impact of the cell-of-origin on CSC properties, tumor initiation, and therapeutic responses. Moreover, we discuss potential therapeutic interventions targeting CSCs based on their distinct cell-of-origin characteristics. Results Accruing evidence suggest that the cell-of-origin, the cell type from which the tumor originates, plays a crucial role in determining the properties of CSCs and their contribution to tumor heterogeneity. Conclusion By providing critical insights into the complex interplay between CSCs and their cellular origins, this article aims to enhance our understanding of cancer biology and pave the way for more effective and personalized cancer treatments.
Collapse
Affiliation(s)
- Oluwaseun Adebayo Bamodu
- Directorate of Postgraduate Studies, School of Clinical Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
- Ocean Road Cancer Institute, Dar es Salaam, Tanzania
| | - Chen-Chih Chung
- Department of Neurology, Taipei Medical University - Shuang Ho Hospital, New Taipei City, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University - Shuang Ho Hospital, New Taipei City, Taiwan
| | - Thomas R. Pisanic
- Johns Hopkins Institute for NanoBioTechnology, Baltimore, MD, United States
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Oncology - Cancer Genetics and Epigenetics, Johns Hopkins University, Baltimore, MD, United States
| | - Alexander T. H. Wu
- The Program for Translational Medicine, Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
48
|
Sun H, Chang Z, Li H, Tang Y, Liu Y, Qiao L, Feng G, Huang R, Han D, Yin DT. Multi-omics analysis-based macrophage differentiation-associated papillary thyroid cancer patient classifier. Transl Oncol 2024; 43:101889. [PMID: 38382228 PMCID: PMC10900934 DOI: 10.1016/j.tranon.2024.101889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/02/2024] [Accepted: 01/21/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND The reclassification of Papillary Thyroid Carcinoma (PTC) is an area of research that warrants attention. The connection between thyroid cancer, inflammation, and immune responses necessitates considering the mechanisms of differential prognosis of thyroid tumors from an immunological perspective. Given the high adaptability of macrophages to environmental stimuli, focusing on the differentiation characteristics of macrophages might offer a novel approach to address the issues related to PTC subtyping. METHODS Single-cell RNA sequencing data of medullary cells infiltrated by papillary thyroid carcinoma obtained from public databases was subjected to dimensionality reduction clustering analysis. The RunUMAP and FindAllMarkers functions were utilized to identify the gene expression matrix of different clusters. Cell differentiation trajectory analysis was conducted using the Monocle R package. A complex regulatory network for the classification of Immune status and Macrophage differentiation-associated Papillary Thyroid Cancer Classification (IMPTCC) was constructed through quantitative multi-omics analysis. Immunohistochemistry (IHC) staining was utilized for pathological histology validation. RESULTS Through the integration of single-cell RNA and bulk sequencing data combined with multi-omics analysis, we identified crucial transcription factors, immune cells/immune functions, and signaling pathways. Based on this, regulatory networks for three IMPTCC clusters were established. CONCLUSION Based on the co-expression network analysis results, we identified three subtypes of IMPTCC: Immune-Suppressive Macrophage differentiation-associated Papillary Thyroid Carcinoma Classification (ISMPTCC), Immune-Neutral Macrophage differentiation-associated Papillary Thyroid Carcinoma Classification (INMPTCC), and Immune-Activated Macrophage differentiation-associated Papillary Thyroid Carcinoma Classification (IAMPTCC). Each subtype exhibits distinct metabolic, immune, and regulatory characteristics corresponding to different states of macrophage differentiation.
Collapse
Affiliation(s)
- Hanlin Sun
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Zhengyan Chang
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Hongqiang Li
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Yifeng Tang
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Yihao Liu
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Lixue Qiao
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Guicheng Feng
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, PR China
| | - Runzhi Huang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, PR China.
| | - Dongyan Han
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, PR China.
| | - De-Tao Yin
- Department of Thyroid Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, PR China; Engineering Research Center of Multidisciplinary Diagnosis and Treatment of Thyroid Cancer of Henan Province, Zhengzhou 450052, Henan, PR China; Key Medicine Laboratory of Thyroid Cancer of Henan Province, Zhengzhou 450052, Henan, PR China.
| |
Collapse
|
49
|
Jia Y, Chen X, Guo H, Zhang B, Liu B. Comprehensive characterization of β-alanine metabolism-related genes in HCC identified a novel prognostic signature related to clinical outcomes. Aging (Albany NY) 2024; 16:7073-7100. [PMID: 38637116 PMCID: PMC11087131 DOI: 10.18632/aging.205744] [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: 07/21/2023] [Accepted: 02/02/2024] [Indexed: 04/20/2024]
Abstract
Hepatocellular carcinoma (HCC) stands out as the most prevalent type of liver cancer and a significant contributor to cancer-related fatalities globally. Metabolic reprogramming, particularly in glucose, lipid, and amino acid metabolism, plays a crucial role in HCC progression. However, the functions of β-alanine metabolism-related genes (βAMRGs) in HCC remain understudied. Therefore, a comprehensive evaluation of βAMRGs is required, specifically in HCC. Initially, we explored the pan-cancer landscape of βAMRGs, integrating expression profiles, prognostic values, mutations, and methylation levels. Subsequently, scRNA sequencing results indicated that hepatocytes had the highest scores of β-alanine metabolism. In the process of hepatocyte carcinogenesis, metabolic pathways were further activated. Using βAMRGs scores and expression profiles, we classified HCC patients into three subtypes and examined their prognosis and immune microenvironments. Cluster 3, characterized by the highest βAMRGs scores, displayed the best prognosis, reinforcing β-alanine's significant contribution to HCC pathophysiology. Notably, immune microenvironment, metabolism, and cell death modes significantly varied among the β-alanine subtypes. We developed and validated a novel prognostic panel based on βAMRGs and constructed a nomogram incorporating risk degree and clinicopathological characteristics. Among the model genes, EHHADH has been identified as a protective protein in HCC. Its expression was notably downregulated in tumors and exhibited a close correlation with factors such as tumor staging, grading, and prognosis. Immunohistochemical experiments, conducted using HCC tissue microarrays, substantiated the validation of its expression levels. In conclusion, this study uncovers β-alanine's significant role in HCC for the first time, suggesting new research targets and directions for diagnosis and treatment.
Collapse
Affiliation(s)
- Yi Jia
- Department of General Surgery, Xinhua Hospital of Dalian University, Dalian, Liaoning, China
| | - Xu Chen
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Hui Guo
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Biao Zhang
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Bin Liu
- Department of General Surgery, Xinhua Hospital of Dalian University, Dalian, Liaoning, China
| |
Collapse
|
50
|
Parihar K, Ko SH, Bradley R, Taylor P, Ramakrishnan N, Baumgart T, Guo W, Weaver VM, Janmey PA, Radhakrishnan R. Free energy calculations for membrane morphological transformations and insights to physical biology and oncology. Methods Enzymol 2024; 701:359-386. [PMID: 39025576 PMCID: PMC11258396 DOI: 10.1016/bs.mie.2024.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
In this chapter, we aim to bridge basic molecular and cellular principles surrounding membrane curvature generation with rewiring of cellular signals in cancer through multiscale models. We describe a general framework that integrates signaling with other cellular functions like trafficking, cell-cell and cell-matrix adhesion, and motility. The guiding question in our approach is: how does a physical change in cell membrane configuration caused by external stimuli (including those by the extracellular microenvironment) alter trafficking, signaling and subsequent cell fate? We answer this question by constructing a modeling framework based on stochastic spatial continuum models of cell membrane deformations. We apply this framework to explore the link between trafficking, signaling in the tumor microenvironment, and cell fate. At each stage, we aim to connect the results of our predictions with cellular experiments.
Collapse
Affiliation(s)
- Kshitiz Parihar
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Seung-Hyun Ko
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Ryan Bradley
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Phillip Taylor
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - N Ramakrishnan
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Tobias Baumgart
- Department of Chemistry, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, United States
| | - Wei Guo
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, United States
| | - Valerie M Weaver
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, United States
| | - Paul A Janmey
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ravi Radhakrishnan
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|