1
|
Oh JM, Park Y, Lee J, Shen K. Microfabricated Organ-Specific Models of Tumor Microenvironments. Annu Rev Biomed Eng 2025; 27:307-333. [PMID: 40310890 DOI: 10.1146/annurev-bioeng-110222-103522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Despite the advances in detection, diagnosis, and treatments, cancer remains a lethal disease, claiming the lives of more than 600,000 people in the United States alone in 2024. To accelerate the development of new therapeutic strategies with improved responses, significant efforts have been made to develop microfabricated in vitro models of tumor microenvironments (TMEs) that address the limitations of animal-based cancer models. These models incorporate several advanced tissue engineering techniques to better reflect the organ- and patient-specific TMEs. Additionally, microfabricated models integrated with next-generation single-cell omics technologies provide unprecedented insights into patient's cellular and molecular heterogeneity and complexity. This review provides an overview of the recent understanding of cancer development and outlines the key TME elements that can be captured in microfabricated models to enhance their physiological relevance. We highlight the recent advances in microfabricated cancer models that reflect the unique characteristics of their organs of origin or sites of dissemination.
Collapse
Affiliation(s)
- Jeong Min Oh
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA;
| | - Yongkuk Park
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA;
| | - Jungwoo Lee
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA;
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
| | - Keyue Shen
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA;
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
2
|
Dawalibi A, Bakir M, Mohammad KS. The genetic architecture of bone metastases: unveiling the role of epigenetic and genetic modifications in drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:19. [PMID: 40342734 PMCID: PMC12059479 DOI: 10.20517/cdr.2025.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/26/2025] [Accepted: 04/17/2025] [Indexed: 05/11/2025]
Abstract
Bone metastases represent frequent and severe complications in various cancers, notably impacting prognosis and quality of life. This review article delves into the genetic and epigenetic mechanisms underpinning drug resistance in bone metastases, a key challenge in effective cancer treatment. The development of drug resistance in cancer can manifest as either intrinsic or acquired, with genetic heterogeneity playing a pivotal role. Intrinsic resistance is often due to pre-existing mutations, while acquired resistance evolves through genetic and epigenetic alterations during treatment. These alterations include mutations in driver genes like TP53 and RB1, epigenetic modifications such as DNA methylation and histone changes, and pathway alterations, notably involving RANK-RANKL signaling and the PI3K/AKT/mTOR cascade. Recent studies underline the significance of the tumor microenvironment in fostering drug resistance, with components such as cancer-associated fibroblasts and hypoxia playing crucial roles. The interactions between metastatic cancer cells and the bone microenvironment facilitate survival and the proliferation of drug-resistant clones. This review highlights the necessity of understanding these complex interactions to develop targeted therapies that can overcome resistance and improve treatment outcomes. Current therapeutic strategies and future directions are discussed, emphasizing the integration of genomic profiling and targeted interventions in managing bone metastases. The evolving landscape of genetic research, including the application of next-generation sequencing and CRISPR technology, offers promising avenues for novel and more effective therapeutic strategies. This comprehensive exploration aims to provide insights into the molecular intricacies of drug resistance in bone metastases, paving the way for improved clinical management and patient care.
Collapse
Affiliation(s)
- Ahmad Dawalibi
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Mohamad Bakir
- Department of Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Khalid S. Mohammad
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
3
|
Bessot A, Gunter J, McGovern J, Bock N. Bone marrow adipocytes in cancer: Mechanisms, models, and therapeutic implications. Biomaterials 2025; 322:123341. [PMID: 40315628 DOI: 10.1016/j.biomaterials.2025.123341] [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: 04/02/2025] [Accepted: 04/12/2025] [Indexed: 05/04/2025]
Abstract
Adipose tissue is the primary site of energy storage in the body and a key regulator of metabolism. However, different adipose depots exhibit distinct molecular and phenotypic characteristics that have yet to be fully unraveled. While initially considered inert, bone marrow adipocytes (BMAs) have been recognized as key regulators of bone homeostasis, and more recently bone pathologies, although many unknowns remain. In this review, we summarize the current knowledge on BMAs, focusing on their distinct characteristics, functional significance in bone physiology and metabolism, as well as their emerging role in cancer pathogenesis. We present and discuss the current methodologies for investigating BMA-cancer interactions, encompassing both in vitro 3D culture systems and in vivo models, and their limitations in accurately replicating the phenotypes and biological processes of the human species. We highlight the imperative for advancing towards humanized models to better mimic the complexities of human physiology and disease progression. Finally, therapeutic strategies targeting metabolism or BMA-secreted factors, such as anti-cholesterol drugs, hold considerable promise in cancer treatment. We present the synergistic avenue of combining conventional cancer therapies with agents targeting adipocyte signaling to amplify treatment efficacy. Developing preclinical models that more faithfully replicate human pathological and physiological processes will lead to more accurate mechanistic understanding of the role of BMAs in bone metastasis and lead to more relevant preclinical drug screening.
Collapse
Affiliation(s)
- Agathe Bessot
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia; Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia; Max Planck Queensland Centre for the Materials Science of Extracellular Matrices, Brisbane, QLD, 4000, Australia
| | - Jennifer Gunter
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia; Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia; Centre for Genomics and Personalised Health, QUT, Brisbane, QLD, 4102, Australia
| | - Jacqui McGovern
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia; Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia; Max Planck Queensland Centre for the Materials Science of Extracellular Matrices, Brisbane, QLD, 4000, Australia; Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering Technologies (CTET), QUT, Brisbane, QLD, 4000, Australia
| | - Nathalie Bock
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia; Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia; Max Planck Queensland Centre for the Materials Science of Extracellular Matrices, Brisbane, QLD, 4000, Australia; Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia; Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling, and Manufacturing (M3D Innovation), Queensland University of Technology, Brisbane, QLD, 4000, Australia.
| |
Collapse
|
4
|
Saadh MJ, Bishoyi AK, Ballal S, Singh A, Kareem RA, Devi A, Sharma GC, Naidu KS, Sead FF. MicroRNAs as behind-the-scenes molecules in breast cancer metastasis and their therapeutic role through novel microRNA-based delivery strategies. Gene 2025; 944:149272. [PMID: 39894085 DOI: 10.1016/j.gene.2025.149272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
Breast cancer is the primary cause of cancer-related death and the most frequent malignancy among women in Western countries. Although there have been advancements in combination treatments and targeted therapies for the metastatic diseases management, metastatic breast cancer is still the second most common cause of cancer-related deaths among U.S. women. The routes of metastasis encompass invasion, intravasation, circulation, extravasation, infiltration into a remote location to establish a metastatic niche, and the formation of micro-metastases in a new environment. Each of these processes is regulated by changes in gene expression. MicroRNAs (miRNAs) are widely expressed by a variety of organisms and have a key role in cell activities including suppressing or promoting cancer through regulating various pathways. Target gene expression is post-transcriptionally regulated by miRNAs, which contribute to the development, spread, and metastasis of breast cancer. In this study, we comprehensively discussed the role of miRNAs as predictors of breast cancer metastasis, their correlation with the spread of the disease to certain organs, and their potential application as targets for breast cancer treatment. We also provided molecular mechanisms of miRNAs in the progression of breast cancer, as well as current challenges in miRNA-based therapeutic approaches. Furthermore, as one of the primary issues with the treatment of solid malignancies is the efficient delivery of miRNAs, we examined a number of cutting-edge carriers for miRNA-based therapies and CRISPR/Cas9 as a targeted therapy for breast cancer.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | - Ashok Kumar Bishoyi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot 360003, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India
| | | | - Anita Devi
- Department of Chemistry Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Fadhil Faez Sead
- Department of Dentistry, College of Dentistry, The Islamic University, Najaf, Iraq; Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
| |
Collapse
|
5
|
Whitman MA, Mantri M, Spanos E, Estroff LA, De Vlaminck I, Fischbach C. Bone mineral density affects tumor growth by shaping microenvironmental heterogeneity. Biomaterials 2025; 315:122916. [PMID: 39490060 DOI: 10.1016/j.biomaterials.2024.122916] [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: 07/19/2024] [Revised: 09/09/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Breast cancer bone metastasis is a major cause of mortality in patients with advanced breast cancer. Although decreased mineral density is a known risk factor for bone metastasis, the underlying mechanisms remain poorly understood because studying the isolated effect of bone mineral density on tumor heterogeneity is challenging with conventional approaches. Moreover, mineralized biomaterials are commonly utilized for clinical bone defect repair, but how mineralized biomaterials affect the foreign body response and wound healing is unclear. Here, we investigate how bone mineral affects tumor growth and microenvironmental complexity in vivo by combining single-cell RNA-sequencing with mineral-containing or mineral-free decellularized bone matrices. We discover that the absence of bone mineral significantly influences fibroblast and immune cell heterogeneity, promoting phenotypes that increase tumor growth and alter the response to injury or disease. Importantly, we observe that the stromal response to bone mineral content depends on the murine tumor model used. While lack of bone mineral induces tumor-promoting microenvironments in both immunocompromised and immunocompetent animals, these changes are mediated by altered fibroblast phenotype in immunocompromised mice and macrophage polarization in immunocompetent mice. Collectively, our findings suggest that bone mineral density affects tumor growth by impacting microenvironmental complexity in an organism-dependent manner.
Collapse
Affiliation(s)
- Matthew A Whitman
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA
| | - Madhav Mantri
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA
| | - Emmanuel Spanos
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA
| | - Lara A Estroff
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, 14850, USA; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, 14850, USA
| | - Iwijn De Vlaminck
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA.
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, 14850, USA.
| |
Collapse
|
6
|
Zhu L, Xie L, Wang Z, Li KL, Cai W. Mass spectrometry-based metabolomics reveal the effects and potential mechanism of isochlorogenic acid A in MC3T3-E1 cells. Front Mol Biosci 2025; 12:1518873. [PMID: 40201241 PMCID: PMC11975594 DOI: 10.3389/fmolb.2025.1518873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/27/2025] [Indexed: 04/10/2025] Open
Abstract
Introduction The bioactive compound 3,5-DiCQA, derived from Duhaldea nervosa, has been traditionally utilized in folk remedies for bone fractures and osteoporosis. However, its therapeutic mechanisms remain unclear. Methods We employed UHPLC-Q Exactive Orbitrap MS-based cell metabolomics to investigate the molecular mechanisms of 3,5-DiCQA in MC3T3-E1 cells. Cell proliferation was assessed via MTT assay, differentiation by alkaline phosphatase (ALP) activity, and mineralization through alizarin red staining and cetylpyridinium chloride quantification. Metabolomic profiling compared drug-treated and control groups. Results Results from MTT assays demonstrated that 3,5-DiCQA significantly promoted cell proliferation at 100 μM. Alkaline phosphatase (ALP) assays and alizarin red staining revealed enhanced osteoblast differentiation and mineralization, respectively. Calcification deposition was significantly increased in the calcified stained cells by cetylpyridinium chloride quantization, indicating that 3,5-DiCQA can promote the mineralization of MC3T3-E1 cells. Metabolomic analysis identified key metabolic changes, including the downregulation of phytosphingosine and upregulation of sphinganine and citric acid. Discussion These findings suggest that 3,5-DiCQA promotes osteoblast proliferation, differentiation and mineralization through pathways such as sphingolipid metabolism, arginine and proline metabolism, mucin type O-glycan biosynthesis and the citrate cycle (TCA cycle). This study provides insights into the therapeutic potential of 3,5-DiCQA for osteoporosis and highlights the utility of metabolomics in elucidating traditional Chinese medicine (TCM).
Collapse
Affiliation(s)
- Lian Zhu
- School of Pharmaceutical Sciences, Sino-Pakistan Center on Traditional Chinese Medicine, Hunan University of Medicine, Huaihua, China
| | - Liu Xie
- Department of Pathology and Research Office of the School of Basic Medicine, Hunan University of Medicine, Huaihua, China
| | - Ziming Wang
- School of Pharmaceutical Sciences, Sino-Pakistan Center on Traditional Chinese Medicine, Hunan University of Medicine, Huaihua, China
| | - Kai-Lin Li
- School of Pharmaceutical Sciences, Sino-Pakistan Center on Traditional Chinese Medicine, Hunan University of Medicine, Huaihua, China
| | - Wei Cai
- School of Pharmaceutical Sciences, Sino-Pakistan Center on Traditional Chinese Medicine, Hunan University of Medicine, Huaihua, China
| |
Collapse
|
7
|
Srikrajang S, Kabir L, Sagadevan S, Wijaya K, Oh WC. Representative modeling of biocompatible MXene nanocomposites for next-generation biomedical technologies and healthcare. J Mater Chem B 2025; 13:2912-2951. [PMID: 39886804 DOI: 10.1039/d4tb02478h] [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: 02/01/2025]
Abstract
MXenes are a class of 2D transition metal carbides and nitrides (Mn+1XnT) that have attracted significant interest owing to their remarkable potential in various fields. The unique combination of their excellent electromagnetic, optical, mechanical, and physical properties have extended their applications to the biological realm as well. In particular, their ultra-thin layered structure holds specific promise for diverse biomedical applications. This comprehensive review explores the synthesis methods of MXene composites, alongside the biological and medical design strategies that have been employed for their surface engineering. This review delves into the interplay between these strategies and the resulting properties, biological activities, and unique effects at the nano-bio-interface. Furthermore, the latest advancements in MXene-based biomaterials and medicine are systematically summarized. Further discussion on MXene composites designed for various applications, including biosensors, antimicrobial agents, bioimaging, tissue engineering, and regenerative medicine, are also provided. Finally, with a focus on translating research results into real-world applications, this review addresses the current challenges and exciting future prospects of MXene composite-based biomaterials.
Collapse
Affiliation(s)
- Siwaluk Srikrajang
- Department of Physical Therapy, Faculty of Medicine, Prince of Songkla University, Songkla, Thailand
| | - Latiful Kabir
- Department of Advanced Materials Science & Engineering, Hanseo University, Seosan, Chungnam 31962, Republic of Korea.
| | - Suresh Sagadevan
- Nanotechnology & Catalysis Research Centre, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Karna Wijaya
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Won-Chun Oh
- Department of Advanced Materials Science & Engineering, Hanseo University, Seosan, Chungnam 31962, Republic of Korea.
| |
Collapse
|
8
|
San Martin R, Das P, Xue T, Brown MR, Dos Reis Marques R, Essington M, Gonzalez A, McCord RP. Amorphous calcium phosphate-coated surfaces as a model for bone microenvironment in prostate cancer. Heliyon 2025; 11:e41929. [PMID: 39931470 PMCID: PMC11808503 DOI: 10.1016/j.heliyon.2025.e41929] [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: 10/01/2024] [Revised: 01/09/2025] [Accepted: 01/12/2025] [Indexed: 02/13/2025] Open
Abstract
Background Bone metastasis remains one of the biggest challenges in the treatment of prostate cancer, and other solid tumors such as breast, lung, and colon. Modeling a complex microenvironment in-vitro such as the bone niche, requires interrogation of cell-cell interactions, specific extracellular matrix proteins, and a high calcium environment. Methods Here, we present a fast and cost-effective system in which commercially available, non-adhesive cell culture vessels are coated with amorphous calcium phosphate (ACP) as a surrogate for bone matrix. We also present modified protocols for subculturing cells and collecting nucleic acids and protein in high-calcium samples. Results We find that prostate epithelial cell lines show increased adhesion and proliferation when cultured in these amorphous calcium surfaces, accompanied by independence from androgen starvation. We observe gene expression changes on ACP surfaces in early adenocarcinoma cell lines which match alterations relevant to prostate cancer progression. Conclusions Incorporating biologically relevant in-vitro systems that address the microenvironment milieu of the metastatic site is essential for accurately modeling cancer progression. In the case of bone metastasis, calcium availability, uptake, and downstream signaling are of paramount importance for the survival of the cancer cell and should be considered in the development of pre-clinical models.
Collapse
Affiliation(s)
- Rebeca San Martin
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, 309 Ken and Blaire Mossman Bldg. 1311 Cumberland Ave, Knoxville, TN, 37996, USA
| | - Prijoyit Das
- UT-ORNL Graduate School of Genome Science and Technology, University of Tennessee, 309 Ken and Blaire Mossman Bldg. 1311 Cumberland Ave, Knoxville, TN, 37996, USA
| | - Tianchun Xue
- UT-ORNL Graduate School of Genome Science and Technology, University of Tennessee, 309 Ken and Blaire Mossman Bldg. 1311 Cumberland Ave, Knoxville, TN, 37996, USA
| | - Morgan Rose Brown
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, 309 Ken and Blaire Mossman Bldg. 1311 Cumberland Ave, Knoxville, TN, 37996, USA
| | - Renata Dos Reis Marques
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, 309 Ken and Blaire Mossman Bldg. 1311 Cumberland Ave, Knoxville, TN, 37996, USA
| | - Michael Essington
- Department of Biosystems Engineering and Soil Science. University of Tennessee, Institute of Agriculture, 2621 Morgan Circle, Knoxville, TN, 37996, USA
| | - Adrian Gonzalez
- Water Quality Core Facility. Department of Civil and Environmental Engineering, Tickle College of Engineering. University of Tennessee, 325 John D. Tickle Engineering Building 851 Neyland Drive, Knoxville, TN, 37996, USA
| | - Rachel Patton McCord
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, 309 Ken and Blaire Mossman Bldg. 1311 Cumberland Ave, Knoxville, TN, 37996, USA
| |
Collapse
|
9
|
Vijayakumar S, González-Sánchez ZI, Amanullah M, Sonamuthu J, Rajkumar M, Divya M, Durán-Lara EF, Li M. Shark chondroitin sulfate gold nanoparticles: A biocompatible apoptotic agent for osteosarcoma. Int J Biol Macromol 2025; 290:138793. [PMID: 39689798 DOI: 10.1016/j.ijbiomac.2024.138793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/19/2024] [Accepted: 12/13/2024] [Indexed: 12/19/2024]
Abstract
Osteosarcoma is a highly aggressive tumor that originates in the bone and often infiltrates nearby bone cells. It is the most prevalent type of primary bone cancer among the various bone malignancies. Traditional cancer treatment methods such as surgery, chemotherapy, immunotherapy, and radiotherapy have had restricted success. However, the integration of nanotechnology into cancer research has led to notable progress. One promising area is the use of marine-derived polysaccharide-based nano formulations for treating various human diseases, including cancer. This study presents a straightforward method for synthesizing biocompatible gold nanoparticles (AuNPs), utilizing sodium borohydride as a reducing agent and a cost-effective, water-soluble chondroitin sulfate (CS) derived from shark cartilage as a stabilizing agent. The synthesized CS-Au NPs appeared purple and were mainly spherical, with 40.768 nm of average size. Cytotoxicity assays (MTT) indicated that CS-Au NPs significantly reduced the viability of human osteosarcoma cells (MG63) at 100 μg/mL, while it showed no cytotoxic effects on mouse embryonic fibroblast cells (NIH3T3) at the same concentration. The observed toxicity of the CS-Au NPs was linked to a rise in the production of reactive oxygen species (ROS) within damaged mitochondria. ROS generation and changes in mitochondrial membrane potential were detected in MG63 cells treated with CS-Au NPs. Furthermore, apoptotic analysis through ethidium bromide dual staining and flow cytometry demonstrated that CS-Au NPs at higher concentrations significantly increased the amount of apoptotic cells, as demonstrated by acridine orange/ethidium bromide staining. Flow cytometry also confirmed that CS-Au NPs activated the apoptotic pathway in MG63 cells.
Collapse
Affiliation(s)
- Sekar Vijayakumar
- College of Material Science and Engineering, Huaqiao University, Engineering Research Center of Environment-Friendly Functional Materials, Ministry of Education, Xiamen 361021, PR China.
| | - Zaira I González-Sánchez
- Nanobiology Laboratory, Department of Natural and Exact Sciences, Pontificia Universidad Católica Madre y Maestra, PUCMM, Autopista Duarte Km 1 ½, Santiago de los Caballeros, Dominican Republic; Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Mohammed Amanullah
- Department of clinical Biochemistry, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Jegatheeswaran Sonamuthu
- Advanced Laboratory of Bio-nanomaterials, BioMe Live Analytical Centre, Kannappa Tower, College Road, Karaikudi 630 003, Tamilnadu, India
| | - Mangaiyarkarasi Rajkumar
- Advanced Laboratory of Bio-nanomaterials, BioMe Live Analytical Centre, Kannappa Tower, College Road, Karaikudi 630 003, Tamilnadu, India
| | - Mani Divya
- Advanced Laboratory of Bio-nanomaterials, BioMe Live Analytical Centre, Kannappa Tower, College Road, Karaikudi 630 003, Tamilnadu, India
| | - Esteban F Durán-Lara
- Bio&NanoMaterialsLab Drug Delivery and Controlled Release, Universidad de Talca, Talca 3460000, Maule, Chile; Departamento de Microbiología, Facultad de Ciencias de la Salud, Universidad de Talca, Talca 3460000, Maule, Chile
| | - Mingchun Li
- College of Material Science and Engineering, Huaqiao University, Engineering Research Center of Environment-Friendly Functional Materials, Ministry of Education, Xiamen 361021, PR China.
| |
Collapse
|
10
|
Bessot A, Röhl J, Emmerich M, Klotz A, Ravichandran A, Meinert C, Waugh D, McGovern J, Gunter J, Bock N. ECM-mimicking hydrogel models of human adipose tissue identify deregulated lipid metabolism in the prostate cancer-adipocyte crosstalk under antiandrogen therapy. Mater Today Bio 2025; 30:101424. [PMID: 39866784 PMCID: PMC11764633 DOI: 10.1016/j.mtbio.2024.101424] [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: 09/09/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/28/2025] Open
Abstract
Antiandrogen therapies are effectively used to treat advanced prostate cancer, but eventually cancer adaptation drives unresolved metastatic castration-resistant prostate cancer (mCRPC). Adipose tissue influences metabolic reprogramming in cancer and was proposed as a contributor to therapy resistance. Using extracellular matrix (ECM)-mimicking hydrogel coculture models of human adipocytes and prostate cancer cells, we show that adipocytes from subcutaneous or bone marrow fat have dissimilar responses under the antiandrogen Enzalutamide. We demonstrate that androgen receptor (AR)-dependent cancer cells (LNCaP) are more influenced by human adipocytes than AR-independent cells (C4-2B), with altered lipid metabolism and adipokine secretion. This response changes under Enzalutamide, with increased AR expression and adipogenic and lipogenic genes in cancer cells and decreased lipid content and gene dysregulation associated with insulin resistance in adipocytes. This is in line with the metabolic syndrome that men with mCRPC under Enzalutamide experience. The all-human, all-3D, models presented here provide a significant advance to dissect the role of fat in therapy response for mCRPC.
Collapse
Affiliation(s)
- Agathe Bessot
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- Max Planck Queensland Centre, Brisbane, QLD, 4000, Australia
- Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia
| | - Joan Röhl
- Faculty of Health Sciences and Medicine, Bond University, Robina, QLD, 4226, Australia
| | - Maria Emmerich
- School of Computation, Information and Technology, Technical University of Munich (TUM), Munich, Germany
| | - Anton Klotz
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Akhilandeshwari Ravichandran
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, QUT, Brisbane, QLD 4000, Australia
- Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering Technologies (CTET), QUT, Brisbane, QLD 4000, Australia
| | | | - David Waugh
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5005, Australia
| | - Jacqui McGovern
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- Max Planck Queensland Centre, Brisbane, QLD, 4000, Australia
- Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering Technologies (CTET), QUT, Brisbane, QLD 4000, Australia
| | - Jenni Gunter
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia
| | - Nathalie Bock
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- Max Planck Queensland Centre, Brisbane, QLD, 4000, Australia
- Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia
- Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling, and Manufacturing (M3D Innovation), Queensland University of Technology, Brisbane, QLD 4000, Australia
| |
Collapse
|
11
|
Haefeli PC, Schelling G, Baumgärtner R, Chang DH, Link BC. Combined interdisciplinary treatment of metastatic bone lesions using 3D robot-assisted image-guided navigation : Embolization, biopsy, ablation, and surgery in one operative session. OPERATIVE ORTHOPADIE UND TRAUMATOLOGIE 2025; 37:34-46. [PMID: 39730876 DOI: 10.1007/s00064-024-00881-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 09/29/2024] [Accepted: 10/10/2024] [Indexed: 12/29/2024]
Abstract
OBJECTIVE To maximize local tumor control, stabilize affected bones, and preserve or replace joints with minimal interventional burden, thereby enhancing quality of life for empowered living. INDICATIONS Suitable for patients with bone metastases, particularly those with severe pain and/or fractures and appropriate life expectancy. CONTRAINDICATIONS In primary bone tumors, refer to the sarcoma surgery team for evaluation of wide resection. For patients with poor general condition and/or limited life expectancy (< 6 weeks), consider best supportive care. SURGICAL TECHNIQUE Radiological interventions involve angiography and embolization for hypervascularized metastases, followed by precise biopsy and local tumor control through radiofrequency ablation or cryoablation using navigated imaging. The surgical treatment aims to create a durable, minimally invasive construct for stability, considering various options from percutaneous screws with cement augmentation to joint replacement. Intraoperative imaging and 3D scans guide the procedure, ensuring accurate placement of implants and confirming optimal results. POSTOPERATIVE MANAGEMENT Postoperative care involves immediate mobilization with pain-adapted full weightbearing and daily physiotherapy. The goal is to regain preoperative mobility. Follow-up with regular clinical and radiographic assessments and CT in the case of tumor progression and complications. RESULTS Since introducing the combined surgical and interventional therapy in October 2021, 16 patients have undergone successful procedures. Complications included material failure, component loosening, and surgical site infection. Five patients (31%) died during observation, while surviving patients surpassed their estimated survival, emphasizing the advantages of minimally invasive treatment with durable constructs.
Collapse
Affiliation(s)
- Pascal C Haefeli
- Department for Orthopaedic and Trauma Surgery, Lucerne Cantonal Hospital LUKS, Spitalstrasse, Lucerne, Switzerland.
| | - Georg Schelling
- Department for Orthopaedic and Trauma Surgery, Lucerne Cantonal Hospital LUKS, Spitalstrasse, Lucerne, Switzerland
| | - Ralf Baumgärtner
- Department for Orthopaedic and Trauma Surgery, Lucerne Cantonal Hospital LUKS, Spitalstrasse, Lucerne, Switzerland
| | - De-Hua Chang
- Department for Interventional Radiology, Lucerne Cantonal Hospital LUKS, Lucerne, Switzerland
| | - Björn-Christian Link
- Department for Orthopaedic and Trauma Surgery, Lucerne Cantonal Hospital LUKS, Spitalstrasse, Lucerne, Switzerland
| |
Collapse
|
12
|
Liu X, Ma R, Wei F, Wang M, Jiang Y, Zheng P, Cao Z. Tumor-derived exosomal lncRNA-MIR193BHG promotes bone metastasis of breast cancer by targeting the miR-489-3p/DNMT3A signaling axis in osteoclasts. J Transl Med 2025; 23:142. [PMID: 39891171 PMCID: PMC11786480 DOI: 10.1186/s12967-025-06156-4] [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: 11/03/2024] [Accepted: 01/19/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Breast cancer exhibits high incidence and mortality among women, with distant metastasis, especially bone metastasis, being the leading cause of death. Despite advances in adjuvant therapies, bone metastasis remains a challenge for patient survival and quality of life. Exosomes, small vesicles capable of mediating intercellular communication, play a crucial role in tumor metastasis. RESULTS This study investigated the role of tumor-derived exosomal long noncoding RNA (lncRNA)-MIR193BHG in breast cancer bone metastasis. LncRNA-MIR193BHG was delivered to osteoclasts via exosomes and promoted osteoclast formation and activity by targeting the miR-489-3p/DNA methyltransferase 3A (DNMT3A) signaling axis, thereby accelerating breast cancer-induced osteolysis. Knockdown experiments demonstrated that reducing the levels of exosomal lncRNA-MIR193BHG significantly inhibited osteoclast differentiation and bone resorption, which was confirmed both in vitro and in vivo. Additionally, mechanistic studies revealed that lncRNA-MIR193BHG acted as a competitive endogenous RNA (ceRNA) interacting with miR-489-3p, regulating DNMT3A expression and subsequently affecting osteoclast differentiation. CONCLUSIONS These findings suggest that lncRNA-MIR193BHG plays a critical regulatory role in breast cancer bone metastasis, and the lncRNA-MIR193BHG/miR-489-3p/DNMT3A signaling axis could be a potential target for the treatment of breast cancer bone metastasis. Future studies should further explore the broader applicability of this mechanism and its clinical feasibility.
Collapse
Affiliation(s)
- Xiaoya Liu
- Department of General Surgery, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
- Department of General Surgery, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Rui Ma
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Feng Wei
- Department of General Surgery, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Maihuan Wang
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Yiwei Jiang
- Department of General Surgery, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
- Department of General Surgery, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Peng Zheng
- Department of General Surgery, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China.
| | - Zhen Cao
- Department of General Surgery, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China.
- Department of General Surgery, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
13
|
Yan J, Wei D, Zhao Z, Sun K, Sun Y. Osteosarcoma-targeting Pt IV prodrug amphiphile for enhanced chemo-immunotherapy via Ca 2+ trapping. Acta Biomater 2025; 193:474-483. [PMID: 39719178 DOI: 10.1016/j.actbio.2024.12.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/15/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
Platinum (PtII)-based anticancer agents exhibit a lack of selectivity in the treatment of osteosarcoma, resulting in significant toxicity. Furthermore, immune surveillance withinthe tumor microenvironment impedes the uptake of platinum drugs by osteosarcoma cells. To overcome these challenges, an oxaliplatin-based PtIV prodrug amphiphile (Lipo-OXA-ALN) was designed and synthesized by incorporatingan osteosarcoma-targeting alendronate (ALN) alongside a lipid tail. The lipid nanoparticles (ALN-OXA), which self-assemble from Lipo-OXA-ALN, enhanced intracellular platinum uptake due to their superior Ca2+ trapping ability and significantly inhibit osteosarcoma cell activity. Moreover, ALN-OXA exhibited potent targeting capabilities, effectively suppressing osteosarcoma growth while preventing bone destruction. Importantly, ALN-OXA induces a series of immune responses characterized by the activation of immune cells, maturation of dendritic cells, and secretion of related cytokines, followed by the activation and infiltration of T lymphocytes and a significant increase in the ratio of cytotoxic T cells. Additionally, the ratio of M1/M2 macrophages increased markedly after ALN-OXA treatment, suggesting potential reprogramming of the tumor microenvironment by ALN-OXA. Overall, the improved therapeutic efficacy against osteosarcoma demonstrates that the PtIV prodrug amphiphile represents a promising strategy for combining targeted chemotherapy with strategies aimed at reversing immune suppression. STATEMENT OF SIGNIFICANCE: Platinum (PtII)-based chemotherapy for osteosarcoma faces challenges due to poor tumor selectivity, leading to suboptimal efficacy and increased toxicity. Additionally, the osteosarcoma microenvironment impedes effective drug delivery. To overcome these limitations, we developed an oxaliplatin-based PtIV prodrug nanoparticle (ALN-OXA) for targeted chemo-immunotherapy. ALN-OXA showed significant in vivo efficacy, effectively preventing bone damage and enhancing the immune microenvironment to improve treatment outcomes. This innovative approach not only targets the tumor more efficiently but also boosts immune response, offering a promising strategy for tumor blockade, tumor starvation, and other therapeutic applications in osteosarcoma treatment.
Collapse
Affiliation(s)
- Jianqin Yan
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Dengshuai Wei
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Zijian Zhao
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Kaichuang Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
14
|
Feng Y, Mo Y, Zhang Y, Teng Y, Xi D, Zhou J, Zeng G, Zong S. Polyphyllin VI: A promising treatment for prostate cancer bone metastasis. Int Immunopharmacol 2025; 144:113684. [PMID: 39602960 DOI: 10.1016/j.intimp.2024.113684] [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: 01/30/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Prostate cancer, as one of the most prevalent malignant tumors in men, seriously affects the prognosis and survival of patients due to its extremely high rate of bone metastasis. This study investigated the effect of Polyphyllin VI (PPVI) on metastatic bone disease for the first time in prostate cancer, focusing on its impact on osteoclast and tumor cell. In vitro studies utilized TRAP staining, ghost pen cyclic peptide staining, and bone resorption assays to evaluate the differentiation and function of receptor activator of nuclear factor-κB ligand (RANKL) induced and RM-1 conditional medium (CM) induced osteoclasts. The colony formation assay, wound healing assay, and Transwell assay were employed to analyze tumor cell proliferation, migration, and invasion in vitro. Flow cytometry was used to detect the cycling and apoptosis of tumor cells in vitro. Western Blot and PCR assays were conducted to assess the expression of genes. In vivo, micro-CT, hematoxylin-eosin staining, and immunohistochemical staining evaluated the impact of PPVI on bone destruction and tumor growth in a mouse model of tumor tibial metastasis. The study results indicated that PPVI effectively inhibited osteoclast differentiation, suppresses tumor cell proliferation, migration, and invasion in vitro, and induces apoptosis and G2/M phase arrest. In vivo, PPVI not only inhibits the growth of metastatic tumors but also mitigates the resulting bone destruction. These results suggest that PPVI holds significant potential as an alternative treatment for prostate cancer with bone metastasis, providing insights into its molecular mechanisms and therapeutic efficacy.
Collapse
Affiliation(s)
- Yanbin Feng
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China; Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Jinan, Shandong, China
| | - Yaomin Mo
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Yang Zhang
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Jinan, Shandong, China
| | - Yilin Teng
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Deshuang Xi
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Junhong Zhou
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Gaofeng Zeng
- Department of Nutrition and Food Hygiene, College of Public Hygiene of Guangxi Medical University, Nanning, Guangxi, China.
| | - Shaohui Zong
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
15
|
Deng M, Huang PZ, Huang ZY, Chen TT, Luo X, Liao CY, Xu WH, Zhao J, Wu QJ, Zheng J. SOX2 control activation of dormant prostate cancer cells in bone metastases by promoting CCNE2 gene expression. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2024; 12:375-388. [PMID: 39839747 PMCID: PMC11744349 DOI: 10.62347/ascy2532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025]
Abstract
BACKGROUND Cancer stem cells (CSCs) have a powerful tumor initiation ability, which can promote the early dissemination of single disseminated tumor cells (DTCs), leading to tumor progression. SOX2, a pluripotent inducible transcription factor, is key to maintaining self-renewal and pluripotency of prostate cancer stem cells. However, there is a lack of comprehensive understanding of how SOX2 regulates DTCs dormancy and proliferation in the bone marrow microenvironment. METHODS AND RESULTS By constructing a mouse bone metastasis model to simulate the progression of prostate cancer with bone metastasis, the bone tissue immunofluorescence showed that SOX2 expression increased with the progression of prostate cancer in the bone marrow microenvironment. We validated this phenomenon with publicly available single-cell and transcriptome datasets and found that SOX2 is involved in multiple phenotypes associated with prostate cancer dormancy, proliferation, and invasion. Further, CCNE2, a potential target downstream of SOX2, was identified through multiple transcription factor databases and protein interaction networks. CONCLUSION The expression of SOX2 affects multiple phenotypes related to dormancy, proliferation and invasion of prostate cancer, and may indirectly activate the dormant prostate cancer cells through the downstream target gene CCNE2, thus affecting the progression and bone metastasis of prostate cancer.
Collapse
Affiliation(s)
- Min Deng
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
| | - Pei-Zheng Huang
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
- School of Medicine, Chongqing UniversityChongqing, China
| | - Ze-Yu Huang
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
| | - Ting-Ting Chen
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
| | - Xing Luo
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
| | - Chao-Yu Liao
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
| | - Wen-Hao Xu
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
| | - Jiang Zhao
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
| | - Qing-Jian Wu
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
| | - Ji Zheng
- Department of Urology, The Second Affiliated Hospital, Army Military Medical UniversityChongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Third Military Medical UniversityChongqing, China
| |
Collapse
|
16
|
Kaur J, Adhikari M, Sabol HM, Anloague A, Khan S, Kurihara N, Diaz-delCastillo M, Andreasen CM, Barnes CL, Stambough JB, Palmieri M, Reyes-Castro O, Zarrer J, Taipaleenmäki H, Ambrogini E, Almeida M, O’Brien CA, Nookaw I, Delgado-Calle J. Single-Cell Transcriptomic Analysis Identifies Senescent Osteocytes That Trigger Bone Destruction in Breast Cancer Metastasis. Cancer Res 2024; 84:3936-3952. [PMID: 39312185 PMCID: PMC11611663 DOI: 10.1158/0008-5472.can-24-0857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/22/2024] [Accepted: 09/19/2024] [Indexed: 09/25/2024]
Abstract
Breast cancer bone metastases increase fracture risk and are a major cause of morbidity and mortality among women. Upon colonization by tumor cells, the bone microenvironment undergoes profound reprogramming to support cancer progression, which disrupts the balance between osteoclasts and osteoblasts and leads to bone lesions. A deeper understanding of the processes mediating this reprogramming could help develop interventions for treating patients with bone metastases. Here, we demonstrated that osteocytes (Ot) in established breast cancer bone metastasis develop premature senescence and a distinctive senescence-associated secretory phenotype (SASP) that favors bone destruction. Single-cell RNA sequencing identified Ots from mice with breast cancer bone metastasis enriched in senescence, SASP markers, and pro-osteoclastogenic genes. Multiplex in situ hybridization and artificial intelligence-assisted analysis depicted Ots with senescence-associated satellite distension, telomere dysfunction, and p16Ink4a expression in mice and patients with breast cancer bone metastasis. Breast cancer cells promoted Ot senescence and enhanced their osteoclastogenic potential in in vitro and ex vivo organ cultures. Clearance of senescent cells with senolytics suppressed bone resorption and preserved bone mass in mice with breast cancer bone metastasis. These results demonstrate that Ots undergo pathological reprogramming by breast cancer cells and identify Ot senescence as an initiating event triggering lytic bone disease in breast cancer metastases. Significance: Breast cancer cells remodel the bone microenvironment by promoting premature cellular senescence and SASP in osteocytes, which can be targeted with senolytics to alleviate bone loss induced by metastatic breast cancer. See related commentary by Frieling and Lynch, p. 3917.
Collapse
Affiliation(s)
- Japneet Kaur
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Manish Adhikari
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Hayley M. Sabol
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Aric Anloague
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Sharmin Khan
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Noriyoshi Kurihara
- Division of Hematology and Oncology, Department of Medicine, Indiana University, Indianapolis, IN, US
| | | | - Christina Møller Andreasen
- Molecular Bone Histology lab, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Clinical Pathology, Odense University Hospital, Odense University Hospital, Odense, Denmark
| | - C. Lowry Barnes
- Department of Orthopedic Surgery; University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Jeffrey B. Stambough
- Department of Orthopedic Surgery; University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Michela Palmieri
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US
| | - Olivia Reyes-Castro
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US
| | - Jennifer Zarrer
- Institute of Musculoskeletal Medicine, Musculoskeletal University Center Munich, University Hospital, LMU Munich, Germany
| | - Hanna Taipaleenmäki
- Institute of Musculoskeletal Medicine, Musculoskeletal University Center Munich, University Hospital, LMU Munich, Germany
| | - Elena Ambrogini
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US
| | - Maria Almeida
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US
| | - Charles A. O’Brien
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Intawat Nookaw
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, US
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Jesus Delgado-Calle
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US
- Department of Orthopedic Surgery; University of Arkansas for Medical Sciences, Little Rock, AR, US
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, US
| |
Collapse
|
17
|
Fan D, Li J, Li L, An M, Yang H, Zhou G, Gao S, Bottini M, Zhang J, Ge K. Phosphate Ion-Responsive and Calcium Peroxide-Based Nanomedicine for Bone-Targeted Treatment of Breast Cancer Bone Metastasis. Adv Healthc Mater 2024; 13:e2402216. [PMID: 39109966 DOI: 10.1002/adhm.202402216] [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: 06/24/2024] [Indexed: 12/18/2024]
Abstract
The treatment of breast cancer bone metastasis is an unresolved clinical challenge, mostly because currently therapeutic approaches cannot simultaneously block the tumor growth and repair the osteolytic bone injuries at the metastatic site. Herein, the study develops a novel nanomedicine to treat breast cancer bone metastasis. The nanomedicine is based on phosphate ion-responsive and calcium peroxide-based nanoparticles carrying the bone-targeting agent zoledronic acid on the surface and loaded with the photosensitizer indocyanine green. Following intravenous administration to a mouse model of breast cancer bone metastasis, the nanoparticles efficiently accumulate at the bone metastasis site, react with free phosphate ions, and form hydroxyapatite nanoaggregates and O2, while releasing the photosensitizer. Hydroxyapatite nanoaggregates elicit the remineralization of the collagenous bone matrix and trigger tumor cell apoptosis. Upon irradiating tumor-bearing legs with an 808 nm laser source, the O2 and free photosensitizer produced 1O2 by the reaction of the nanoparticles with phosphate ions, further boosting the anti-tumor effect. Tumor killing hampers the vicious cycle at the site of bone metastasis, translating to osteolysis blockade and further encouraging the remineralization of bone matrix. This work sheds light on the development of a novel, safe, and efficient approach for the treatment of breast cancer bone metastasis.
Collapse
Affiliation(s)
- Dehui Fan
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Material Science, Hebei University, Baoding, 071002, China
| | - Jing Li
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Material Science, Hebei University, Baoding, 071002, China
| | - Luwei Li
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Material Science, Hebei University, Baoding, 071002, China
| | - Ming An
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Material Science, Hebei University, Baoding, 071002, China
- Orthopedics Department, Bao Ding NO.1 Central Hospital, Baoding, 071000, China
| | - Hua Yang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Guoqiang Zhou
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Material Science, Hebei University, Baoding, 071002, China
- Hebei University, Baoding, 071002, China
| | - Shutao Gao
- College of Science, Hebei Agricultural University, Baoding, 071002, China
| | - Massimo Bottini
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
- Sanford Burnham Prebys, La Jolla, CA, 92037, USA
| | - Jinchao Zhang
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Material Science, Hebei University, Baoding, 071002, China
| | - Kun Ge
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Material Science, Hebei University, Baoding, 071002, China
| |
Collapse
|
18
|
Bai H, Li Z, Weng Y, Cui F, Chen W. Integrated analysis of single-cell RNA-seq and bulk RNA-seq revealed key genes for bone metastasis and chemoresistance in prostate cancer. Genes Genomics 2024; 46:1445-1460. [PMID: 39395905 DOI: 10.1007/s13258-024-01575-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 09/24/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND Prostate cancer (PCa) is a serious malignancy. The main causes of PCa aggravation and death are unexplained resistance to chemotherapy and bone metastases. OBJECTIVE This study aimed to investigate the molecular mechanisms associated with the dynamic processes of progression, bone metastasis, and chemoresistance in PCa. METHODS Through comprehensive analysis of single-cell RNA sequencing (scRNA-seq) data, Gene Expression Omnibus (GEO) tumor progression and metastasis-related genes were identified. These genes were subjected to lasso regression modeling using the Cancer Genome Atlas (TCGA) database. Tartrate-resistant acid phosphatase (TRAP) staining and real-time quantitative PCR (RT-qPCR) were used to evaluate osteoclast differentiation. CellMiner was used to confirm the effect of LDHA on chemoresistance. Finally, the relationship between LDHA and chemoresistance was verified using doxorubicin-resistant PCa cell lines. RESULTS 7928 genes were identified as genes related to tumor progression and metastasis. Of these, 7 genes were found to be associated with PCa prognosis. The scRNA-seq and TCGA data showed that the expression of LDHA was higher in tumors and associated with poor prognosis of PCa. In addition, upregulation of LDHA in PCa cells induces osteoclast differentiation. Additionally, high LDHA expression was associated with resistance to Epirubicin, Elliptinium acetate, and doxorubicin. Cellular experiments demonstrated that LDHA knockdown inhibited doxorubicin resistance in PCa cells. CONCLUSIONS LDHA may play a potential contributory role in PCa initiation and development, bone metastasis, and chemoresistance. LDHA is a key target for the treatment of PCa.
Collapse
Affiliation(s)
- Hongai Bai
- Clinical Trial Department, Wenzhou Central Hospital, Wenzhou, People's Republic of China
| | - Zhenyue Li
- Pharmacy Department, Wenzhou Central Hospital, Wenzhou, People's Republic of China
| | - Yueyue Weng
- Pharmacy Department, Wenzhou Central Hospital, Wenzhou, People's Republic of China
| | - Facai Cui
- Department of Clinical Laboratory, Henan provincial people's hospital, The people's hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Wenpu Chen
- Urology Surgery, Jinshan Branch of Shanghai Sixth People's Hospital, Shanghai, People's Republic of China.
| |
Collapse
|
19
|
Sevcikova A, Martiniakova M, Omelka R, Stevurkova V, Ciernikova S. The Link Between the Gut Microbiome and Bone Metastasis. Int J Mol Sci 2024; 25:12086. [PMID: 39596154 PMCID: PMC11593804 DOI: 10.3390/ijms252212086] [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: 10/30/2024] [Revised: 11/08/2024] [Accepted: 11/09/2024] [Indexed: 11/28/2024] Open
Abstract
The gut microbiome is essential for regulating host metabolism, defending against pathogens, and shaping the host's immune system. Mounting evidence highlights that disruption in gut microbial communities significantly impacts cancer development and treatment. Moreover, tumor-associated microbiota, along with its metabolites and toxins, may contribute to cancer progression by promoting epithelial-to-mesenchymal transition, angiogenesis, and metastatic spread to distant organs. Bones, in particular, are common sites for metastasis due to a rich supply of growth and neovascularization factors and extensive blood flow, especially affecting patients with thyroid, prostate, breast, lung, and kidney cancers, where bone metastases severely reduce the quality of life. While the involvement of the gut microbiome in bone metastasis formation is still being explored, proposed mechanisms suggest that intestinal dysbiosis may alter the bone microenvironment via the gut-immune-bone axis, fostering a premetastatic niche and immunosuppressive milieu suitable for cancer cell colonization. Disruption in the delicate balance of bone modeling and remodeling may further create a favorable environment for metastatic growth. This review focuses on the link between beneficial or dysbiotic microbiome composition and bone homeostasis, as well as the role of the microbiome in bone metastasis development. It also provides an overview of clinical trials evaluating the impact of gut microbial community structure on bone parameters across various conditions or health-related issues. Dietary interventions and microbiota modulation via probiotics, prebiotics, and fecal microbiota transplantation help support bone health and might offer promising strategies for addressing bone-related complications in cancer.
Collapse
Affiliation(s)
- Aneta Sevcikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (A.S.); (V.S.)
| | - Monika Martiniakova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia;
| | - Radoslav Omelka
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia;
| | - Viola Stevurkova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (A.S.); (V.S.)
| | - Sona Ciernikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (A.S.); (V.S.)
| |
Collapse
|
20
|
Wei X, Liang M, Deng M, Zheng J, Luo F, Ma Q. A switch from lysosomal degradation to secretory autophagy initiates osteogenic bone metastasis in prostate cancer. J Extracell Vesicles 2024; 13:e70002. [PMID: 39497621 PMCID: PMC11535520 DOI: 10.1002/jev2.70002] [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: 12/29/2023] [Revised: 07/27/2024] [Accepted: 09/29/2024] [Indexed: 11/08/2024] Open
Abstract
The identification of both autophagy-related material degradation and unconventional secretion has paved the way for significant breakthroughs linking autophagy to a plethora of physiological processes and disease conditions. However, the mechanisms that coordinate these two pathways remain elusive. Here, we demonstrate that a switch from the lysosomal degradation to a secretory autophagy pathway is governed by protein tyrosine phosphatase 1B (PTP1B, encoded by PTPN1). Dephosphorylation at two tyrosine residues of syntaxin17 (STX17) by PTP1B reduces autophagosome-lysosome fusion while switching the cells to a secretory autophagy pathway. Both PTP1B overexpression and tumour-derived extracellular vesicles (EVs) can activate the secretory autophagy pathway in osteoblasts. Moreover, we demonstrate that osteoblastic LC3+ EVs, generated via the secretory autophagy pathway, are the primary contributor to tumour-associated bone remodelling in prostate cancer. Depletion of tumour-derived EVs secretion or genetic ablation of osteoblastic PTP1B rescues aberrant bone remodelling and lesions, highlighting the relevance between LC3+ EVs and the formation of bone metastatic niche. Our results reveal the significance of tumour-regulated PTP1B in the fate decision of autophagosomes, and propose a role ofLC3+ EVs in shaping the bone metastatic niche.
Collapse
Affiliation(s)
- Xiaoyu Wei
- Department of Orthopedics, Southwest HospitalThird Military Medical UniversityChongqingChina
| | - Mengmeng Liang
- Department of Orthopedics, Southwest HospitalThird Military Medical UniversityChongqingChina
- Institute of Environment and Operational MedicineAcademy of Military Medicine Sciences, Academy of Military SciencesTianjinChina
| | - Min Deng
- Department of UrologyXinqiao Hospital, Third Military Medical UniversityChongqingChina
| | - Ji Zheng
- Department of UrologyXinqiao Hospital, Third Military Medical UniversityChongqingChina
| | - Fei Luo
- Department of Orthopedics, Southwest HospitalThird Military Medical UniversityChongqingChina
| | - Qinyu Ma
- Department of Orthopedics, Southwest HospitalThird Military Medical UniversityChongqingChina
- Institute of CancerXinqiao Hospital, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of ImmunotherapyChongqingChina
- Shigatse Branch, Xinqiao HospitalThird Military Medical UniversityShigatseChina
| |
Collapse
|
21
|
Kawaai K, Oishi Y, Kuroda Y, Tamura R, Toda M, Matsuo K. Chordoma cells possess bone-dissolving activity at the bone invasion front. Cell Oncol (Dordr) 2024; 47:1663-1677. [PMID: 38652222 PMCID: PMC11466907 DOI: 10.1007/s13402-024-00946-6] [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] [Accepted: 04/01/2024] [Indexed: 04/25/2024] Open
Abstract
PURPOSE Chordomas are malignant tumors that destroy bones, compress surrounding nerve tissues and exhibit phenotypes that recapitulate notochordal differentiation in the axial skeleton. Chordomas recur frequently, as they resist radio-chemotherapy and are difficult to completely resect, leading to repeated bone destruction and local expansion via unknown mechanisms. Here, using chordoma specimens and JHC7 chordoma cells, we asked whether chordoma cells possess bone-dissolving activity. METHODS CT imaging and histological analysis were performed to evaluate the structure and mineral density of chordoma-invaded bone and osteolytic marker expression. JHC7 cells were subjected to immunocytochemistry, imaging of cell fusion, calcium dynamics and acidic vacuoles, and bone lysis assays. RESULTS In patients, we found that the skull base invaded by chordoma was highly porous, showed low mineral density and contained brachyury-positive chordoma cells and conventional osteoclasts both expressing the osteolytic markers tartrate-resistant acid phosphatase (TRAP) and collagenases. JHC7 cells expressed TRAP and cathepsin K, became multinucleated via cell-cell fusion, showed spontaneous calcium oscillation, and were partly responsive to the osteoclastogenic cytokine RANKL. JHC7 cells exhibited large acidic vacuoles, and nonregulatory bone degradation without forming actin rings. Finally, bone-derived factors, calcium ions, TGF-β1, and IGF-1 enhanced JHC7 cell proliferation. CONCLUSION In chordoma, we propose that in addition to conventional bone resorption by osteoclasts, chordoma cells possess bone-dissolving activity at the tumor-bone boundary. Furthermore, bone destruction and tumor expansion may occur in a positive feedback loop.
Collapse
Affiliation(s)
- Katsuhiro Kawaai
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Yumiko Oishi
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Yukiko Kuroda
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Ryota Tamura
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Masahiro Toda
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Koichi Matsuo
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan.
| |
Collapse
|
22
|
Bowling GC, Alex Albright J, Maloney TJ, Quinn MS, Daniels AH, Chesnut GT. Poor Bone Mineral Density Is Associated With Increased Risk of Urological Bone Metastases. Urology 2024; 192:88-96. [PMID: 38710454 DOI: 10.1016/j.urology.2024.04.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/21/2024] [Accepted: 04/24/2024] [Indexed: 05/08/2024]
Abstract
OBJECTIVE To investigate whether a diagnosis of precancer poor bone mineral density (PBMD) is associated with higher risk of urological cancer bone metastasis. METHODS The PearlDiver Database was utilized to conduct a retrospective, propensity-matched cohort analysis of adult patients diagnosed with kidney, bladder, prostate, and testicular cancer with and without a prior diagnosis of PBMD, defined as osteopenia or osteoporosis. Unadjusted and adjusted odds ratios (OR) and 95% confidence intervals are used to compare the rate of newly diagnosed bone metastases between 6months and 3years of the initial cancer diagnosis between the experimental and control cohorts. RESULTS Among 685,066 patients with urological cancers, precancer PBMD was associated with increased odds of bone metastasis at various time periods (1week, 6months, 1, 2, and 3years). The strongest association was appreciated within 1week of cancer diagnosis (kidney: adjusted odds ratio [aOR], 2.37, P <.001; bladder: [aOR], 2.37, P <.001; prostate: [aOR], 2.84, P <.001; testicular: [aOR], 4.45, P <.001). Bisphosphonates were associated with reduced risk of kidney ([aOR], 0.46, P <.001), bladder ([aOR], 0.61, P <.001), and prostate ([aOR], 0.66, P <.001) cancer bone metastasis. CONCLUSION Our findings suggest urology patients with PBMD may be predisposed to forming bone metastases as well as presenting with metastatic disease at time of cancer diagnosis. As such, further studies are needed to elucidate whether PBMD plays a role in bone tropism and whether bone health pertains to prolonging bone-free metastasis.
Collapse
Affiliation(s)
- Gartrell C Bowling
- School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD.
| | | | - Trevor J Maloney
- Urology Service, Walter Reed National Military Medical Center, Bethesda, MD
| | - Matthew S Quinn
- Department of Orthopaedics, Brown University Warren Alpert Medical School, Providence, RI
| | - Alan H Daniels
- Department of Orthopaedics, Brown University Warren Alpert Medical School, Providence, RI
| | - Gregory T Chesnut
- Urology Service, Walter Reed National Military Medical Center, Bethesda, MD; Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD
| |
Collapse
|
23
|
Xin Z, Qin L, Tang Y, Guo S, Li F, Fang Y, Li G, Yao Y, Zheng B, Zhang B, Wu D, Xiao J, Ni C, Wei Q, Zhang T. Immune mediated support of metastasis: Implication for bone invasion. Cancer Commun (Lond) 2024; 44:967-991. [PMID: 39003618 PMCID: PMC11492328 DOI: 10.1002/cac2.12584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 06/05/2024] [Accepted: 06/18/2024] [Indexed: 07/15/2024] Open
Abstract
Bone is a common organ affected by metastasis in various advanced cancers, including lung, breast, prostate, colorectal, and melanoma. Once a patient is diagnosed with bone metastasis, the patient's quality of life and overall survival are significantly reduced owing to a wide range of morbidities and the increasing difficulty of treatment. Many studies have shown that bone metastasis is closely related to bone microenvironment, especially bone immune microenvironment. However, the effects of various immune cells in the bone microenvironment on bone metastasis remain unclear. Here, we described the changes in various immune cells during bone metastasis and discussed their related mechanisms. Osteoblasts, adipocytes, and other non-immune cells closely related to bone metastasis were also included. This review also summarized the existing treatment methods and potential therapeutic targets, and provided insights for future studies of cancer bone metastasis.
Collapse
Affiliation(s)
- Zengfeng Xin
- Department of Orthopedic SurgerySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Luying Qin
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Yang Tang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Siyu Guo
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Fangfang Li
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Yuan Fang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Gege Li
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Yihan Yao
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Binbin Zheng
- Department of Respiratory MedicineNingbo Hangzhou Bay HospitalNingboZhejiangP. R. China
| | - Bicheng Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Dang Wu
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Jie Xiao
- Department of Orthopedic SurgerySecond Affiliated Hospital (Jiande Branch)Zhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Chao Ni
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Breast SurgerySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Qichun Wei
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Ting Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| |
Collapse
|
24
|
Zhang L, Tan J. Effect of primary osteoblast-derived extracellular vesicles on osteoclast differentiation. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:434-442. [PMID: 39034117 PMCID: PMC11375487 DOI: 10.3724/zdxbyxb-2024-0148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
OBJECTIVES To investigate the effect of osteoblast-derived extracellular vesicles (OB-EVs) on the proliferation and differentiation of osteoclasts, and to explore the possible molecular mechanism of extracellular vesicles involved in the communication between osteoblasts and osteoclasts. METHODS Primary osteoblasts were isolated from newborn mouse calvarial bone and induced by β-glycero phosphate, ascorbic acid and dexamethasone. Osteogenic feature was tested by alkaline phosphatase (ALP) and alizarin red S staining. Extracellular vesicles were isolated by ultracentrifugation from the cell culture supernatant. Vesicle morphology was observed by transmission electron microscopy, and the characteristic markers of tumor susceptibility gene 101 (TSG101), ALG-2 interacting protein X (Alix) and cluster of differentiation 9 (CD9) on the surface of extracellular vesicles were identified by Western blotting. Cell counting kit 8 (CCK-8) assay was used to determine the proliferation effect of OB-EVs on mouse mononuclear macrophage RAW264.7 cells. Furthermore, the expression level of specific markers of osteoclast differentiation in RAW264.7 cells was detected by Western blotting after the combined effect of OB-EVs and receptor activator for nuclear factor κB ligand (RANKL). The number of osteoclasts was observed and compared with OB-EVs-treated mouse bone marrow-derived macrophages (BMMs) by tartrate-resistant acid phosphatase (TRAP) staining, and the effect of OB-EVs on osteoclast differentiation was determined. RESULTS The extracted OB-EVs showed a double-layer cup-like structure with a diameter of 30-150 nm, and TSG101, Alix and CD9 were expressed. RAW264.7 cells were stimulated with OB-EVs, and the results of CCK-8 assay showed that high concentration of OB-EVs (more than 20 μg/mL) inhibited cell proliferation (P<0.05). Western blotting analysis showed that the expression of osteoclast differentiation marker proteins such as c-Fos, activated T cell nuclear factor (NFATc1) and c-Jun N-terminal kinase (JNK) in RAW264.7 cells were significantly increased, and the promoting effect was enhanced with increasing of OB-EVs concentration (P<0.05). In addition, the combination of OB-EVs and RANKL on BMMs showed that the number of TRAP-positive cells was significantly higher than that of the RANKL induction group alone (P<0.05). CONCLUSIONS OB-EVs can promote the differentiation of osteoclast precursor cells into osteoclasts, but high concentration of OB-EVs can inhibit proliferation of RAW264.7 cells.
Collapse
Affiliation(s)
- Lan Zhang
- Department of Stomatology, Zhejiang Hospital, Hangzhou 310030, China.
| | - Jingyi Tan
- Department of Stomatology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| |
Collapse
|
25
|
Dawalibi A, Alosaimi AA, Mohammad KS. Balancing the Scales: The Dual Role of Interleukins in Bone Metastatic Microenvironments. Int J Mol Sci 2024; 25:8163. [PMID: 39125732 PMCID: PMC11311339 DOI: 10.3390/ijms25158163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
Bone metastases, a common and debilitating consequence of advanced cancers, involve a complex interplay between malignant cells and the bone microenvironment. Central to this interaction are interleukins (ILs), a group of cytokines with critical roles in immune modulation and inflammation. This review explores the dualistic nature of pro-inflammatory and anti-inflammatory interleukins in bone metastases, emphasizing their molecular mechanisms, pathological impacts, and therapeutic potential. Pro-inflammatory interleukins, such as IL-1, IL-6, and IL-8, have been identified as key drivers in promoting osteoclastogenesis, tumor proliferation, and angiogenesis. These cytokines create a favorable environment for cancer cell survival and bone degradation, contributing to the progression of metastatic lesions. Conversely, anti-inflammatory interleukins, including IL-4, IL-10, and IL-13, exhibit protective roles by modulating immune responses and inhibiting osteoclast activity. Understanding these opposing effects is crucial for developing targeted therapies aimed at disrupting the pathological processes in bone metastases. Key signaling pathways, including NF-κB, JAK/STAT, and MAPK, mediate the actions of these interleukins, influencing tumor cell survival, immune cell recruitment, and bone remodeling. Targeting these pathways presents promising therapeutic avenues. Current treatment strategies, such as the use of denosumab, tocilizumab, and emerging agents like bimekizumab and ANV419, highlight the potential of interleukin-targeted therapies in mitigating bone metastases. However, challenges such as therapeutic resistance, side effects, and long-term efficacy remain significant hurdles. This review also addresses the potential of interleukins as diagnostic and prognostic biomarkers, offering insights into patient stratification and personalized treatment approaches. Interleukins have multifaceted roles that depend on the context, including the environment, cell types, and cellular interactions. Despite substantial progress, gaps in research persist, particularly regarding the precise mechanisms by which interleukins influence the bone metastatic niche and their broader clinical implications. While not exhaustive, this overview underscores the critical roles of interleukins in bone metastases and highlights the need for continued research to fully elucidate their complex interactions and therapeutic potential. Addressing these gaps will be essential for advancing our understanding and treatment of bone metastases in cancer patients.
Collapse
Affiliation(s)
- Ahmad Dawalibi
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| | - Amal Ahmed Alosaimi
- College of Medicine, Imam Mohammad Ibn Saud Islamic University, Riyadh 11432, Saudi Arabia;
| | - Khalid S. Mohammad
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| |
Collapse
|
26
|
Riquelme MA, Wang X, Acosta FM, Zhang J, Chavez J, Gu S, Zhao P, Xiong W, Zhang N, Li G, Srinivasan S, Ma C, Rao MK, Sun LZ, Zhang N, An Z, Jiang JX. Antibody-activation of connexin hemichannels in bone osteocytes with ATP release suppresses breast cancer and osteosarcoma malignancy. Cell Rep 2024; 43:114377. [PMID: 38889005 PMCID: PMC11380445 DOI: 10.1016/j.celrep.2024.114377] [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: 10/05/2023] [Revised: 05/02/2024] [Accepted: 05/31/2024] [Indexed: 06/20/2024] Open
Abstract
Bone tissue represents the most frequent site of cancer metastasis. We developed a hemichannel-activating antibody, Cx43-M2. Cx43-M2, directly targeting osteocytes in situ, activates osteocytic hemichannels and elevates extracellular ATP, thereby inhibiting the growth and migration of cultured breast and osteosarcoma cancer cells. Cx43-M2 significantly decreases breast cancer metastasis, osteosarcoma growth, and osteolytic activity, while improving survival rates in mice. The antibody's inhibition of breast cancer and osteosarcoma is dose dependent in both mouse and human cancer metastatic models. Furthermore, Cx43-M2 enhances anti-tumor immunity by increasing the population and activation of tumor-infiltrating immune-promoting effector T lymphocytes, while reducing immune-suppressive regulatory T cells. Our results suggest that the Cx43-M2 antibody, by activating Cx43 hemichannels and facilitating ATP release and purinergic signaling, transforms the cancer microenvironment from a supportive to a suppressive state. Collectively, our study underscores the potential of Cx43-M2 as a therapeutic for treating breast cancer bone metastasis and osteosarcoma.
Collapse
Affiliation(s)
- Manuel A Riquelme
- Departments of Biochemistry and Structural Biology, Microbiology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Xuewei Wang
- Departments of Biochemistry and Structural Biology, Microbiology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Francisca M Acosta
- Departments of Biochemistry and Structural Biology, Microbiology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Jingruo Zhang
- Departments of Biochemistry and Structural Biology, Microbiology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Jeffery Chavez
- Departments of Biochemistry and Structural Biology, Microbiology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Sumin Gu
- Departments of Biochemistry and Structural Biology, Microbiology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Peng Zhao
- The Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Wei Xiong
- The Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Ningyan Zhang
- The Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Guo Li
- Immunology & Molecular Genetics, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Saranya Srinivasan
- Immunology & Molecular Genetics, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Chaoyu Ma
- Immunology & Molecular Genetics, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Manjeet K Rao
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA; Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Lu-Zhe Sun
- Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Nu Zhang
- Immunology & Molecular Genetics, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA; South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| | - Zhiqiang An
- The Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA.
| | - Jean X Jiang
- Departments of Biochemistry and Structural Biology, Microbiology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA.
| |
Collapse
|
27
|
Gu C, Chen P, Tian H, Yang Y, Huang Z, Yan H, Tang C, Xiang J, Shangguan L, Pan K, Chen P, Huang Y, Liu Z, Tang R, Fan S, Lin X. Targeting initial tumour-osteoclast spatiotemporal interaction to prevent bone metastasis. NATURE NANOTECHNOLOGY 2024; 19:1044-1054. [PMID: 38499860 DOI: 10.1038/s41565-024-01613-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/16/2024] [Indexed: 03/20/2024]
Abstract
Bone is the most common site of metastasis, and although low proliferation and immunoediting at the early stage make existing treatment modalities less effective, the microenvironment-inducing behaviour could be a target for early intervention. Here we report on a spatiotemporal coupling interaction between tumour cells and osteoclasts, and named the tumour-associated osteoclast 'tumasteoclast'-a subtype of osteoclasts in bone metastases induced by tumour-migrasome-mediated cytoplasmic transfer. We subsequently propose an in situ decoupling-killing strategy in which tetracycline-modified nanoliposomes encapsulating sodium bicarbonate and sodium hydrogen phosphate are designed to specifically release high concentrations of hydrogen phosphate ions triggered by tumasteoclasts, which depletes calcium ions and forms calcium-phosphorus crystals. This can inhibit the formation of migrasomes for decoupling and disrupt cell membrane for killing, thereby achieving early prevention of bone metastasis. This study provides a research model for exploring tumour cell behaviour in detail and a proof-of-concept for behaviour-targeting strategy.
Collapse
Affiliation(s)
- Chenhui Gu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China
| | - Pengfei Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China
| | - Hongsen Tian
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China
| | - Yang Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China
| | - Zhenxiang Huang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China
| | - Huige Yan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China
| | - Chenxi Tang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiajia Xiang
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Liqing Shangguan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China
| | - Kaifeng Pan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China
| | - Pengyu Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China
| | - Yue Huang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China
| | - Zhaoming Liu
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Ruikang Tang
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China.
| | - Xianfeng Lin
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
28
|
Chen X, Gong L, Wang Y, Ye C, Guo H, Gao S, Chen J, Wang Z, Gao Y. IL-23 inhibitor enhances the effects of PTEN DNA-loaded lipid nanoparticles for metastatic CRPC therapy. Front Pharmacol 2024; 15:1388613. [PMID: 38898927 PMCID: PMC11186457 DOI: 10.3389/fphar.2024.1388613] [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: 02/20/2024] [Accepted: 05/09/2024] [Indexed: 06/21/2024] Open
Abstract
Introduction: Metastatic castration-resistant prostate cancer (mCRPC) patients face challenges due to limited treatment options. About 50% of patients with mCRPC have a functional loss of phosphatase and tensin homology deleted on chromosome 10 (PTEN), leading to tumor progression, metastasis, and immune suppression. Moreover, elevated IL-23 produced by myeloid-derived suppressor cells (MDSCs) is found in CRPC patients, driving tumor progression. Therefore, a combination strategy based on PTEN restoration and IL-23 inhibition may block CRPC progression and metastasis. Methods: The antitumor effect of restoring PTEN expression combined with the IL-23 inhibitor Apilimod was studied in a mouse model of bone metastasis CRPC and mouse prostate cancer RM-1 cells. To verify the targeting ability of PTEN DNA coated with lipid nanoparticles (LNP@PTEN) in vitro and in vivo. In addition, RT-qPCR and flow cytometry were used to investigate the related mechanisms of the antitumor effect of LNP@PTEN combined with Apilimod. Results: LNPs exhibited significant tumor-targeting and tumor accumulation capabilities both in vitro and in vivo, enhancing PTEN expression and therapeutic efficacy. Additionally, the combination of LNP@PTEN with the IL-23 inhibitor Apilimod demonstrated enhanced inhibition of tumor growth, invasion, and metastasis (particularly secondary organ metastasis) compared to other groups, and extended the survival of mice to 41 days, providing a degree of bone protection. These effects may be attributed to the PTEN function restoration combined with IL-23 inhibition, which help reverse immune suppression in the tumor microenvironment by reducing MDSCs recruitment and increasing the CD8+/CD4+ T cell ratio. Discussion: In summary, these findings highlight the potential of LNPs for delivering gene therapeutic agents. And the combination of LNP@PTEN with Apilimod could achieve anti-tumor effects and improve tumor microenvironment. This combinational strategy opens new avenues for the treatment of mCRPC.
Collapse
Affiliation(s)
- Xinlu Chen
- School of Pharmacy, Fudan University, Shanghai, China
- Department of Pharmacy, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Luyao Gong
- School of Pharmacy, Fudan University, Shanghai, China
| | - Yuanyuan Wang
- School of Pharmacy, Fudan University, Shanghai, China
| | - Chen Ye
- Department of Pharmacy, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Huanhuan Guo
- Department of Pharmacy, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Shen Gao
- Department of Pharmacy, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jiyuan Chen
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhuo Wang
- School of Pharmacy, Fudan University, Shanghai, China
| | - Yuan Gao
- School of Pharmacy, Fudan University, Shanghai, China
- Department of Pharmacy, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
29
|
Fairfield H, Karam M, Schimelman A, Qiang YW, Reagan MR. Adipocytes and metabolism: Contributions to multiple myeloma. J Bone Oncol 2024; 46:100609. [PMID: 38872708 PMCID: PMC11169464 DOI: 10.1016/j.jbo.2024.100609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 05/03/2024] [Accepted: 05/15/2024] [Indexed: 06/15/2024] Open
Abstract
Obesity contributes to many cancers, including breast cancer and multiple myeloma, two cancers that often colonize the bone marrow (BM). Obesity often causes metabolic disease, but at the cellular level, there is uncertainty regarding how these shifts affect cellular phenotypes. Evidence is building that different types of fuel affect tumor cell metabolism, mitochondrial function, and signaling pathways differently, but tumor cells are also flexible and adapt to less-than ideal metabolic conditions, suggesting that single-pronged attacks on tumor metabolism may not be efficacious enough to be effective clinically. In this review, we describe the newest research at the pre-clinical level on how tumor metabolic pathways and energy sources affect cancer cells, with a special focus on multiple myeloma (MM). We also describe the known forward-feedback loops between bone marrow adipocytes (BMAds) and local tumor cells that support tumor growth. We describe how metabolic targets and transcription factors related to fatty acid (FA) oxidation, FA biosynthesis, glycolysis, oxidative phosphorylation (OXPHOS), and other pathways hold great promise as new vulnerabilities in myeloma cells. Specifically, we describe the importance of the acetyl-CoA synthetase (ACSS) and the acyl-CoA synthetase long chain (ACSL) families, which are both involved in FA metabolism. We also describe new data on the importance of lactate metabolism and lactate transporters in supporting the growth of tumor cells in a hypoxic BM microenvironment. We highlight new data showing the dependency of myeloma cells on the mitochondrial pyruvate carrier (MPC), which transports pyruvate to the mitochondria to fuel the tricarboxylic acid (TCA) cycle and electron transport chain (ETC), boosting OXPHOS. Inhibiting the MPC affects myeloma cell mitochondrial metabolism and growth, and synergizes with proteosome inhibitors in killing myeloma cells. We also describe how metabolic signaling pathways intersect established survival and proliferation pathways; for example, the fatty acid binding proteins (FABPs) affect MYC signaling and support growth, survival, and metabolism of myeloma cells. Our goal is to review the current the field so that novel, metabolic-focused therapeutic interventions and treatments can be imagined, developed and tested to decrease the burden of MM and related cancers.
Collapse
Affiliation(s)
- Heather Fairfield
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
- Tufts University School of Medicine, Boston MA, USA
| | - Michelle Karam
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- Tufts University School of Medicine, Boston MA, USA
| | - Allyson Schimelman
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- Tufts University School of Medicine, Boston MA, USA
- Roux Institute, Northeastern University, Portland, ME, USA
| | - Ya-Wei Qiang
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
- Tufts University School of Medicine, Boston MA, USA
| | - Michaela R. Reagan
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
- Tufts University School of Medicine, Boston MA, USA
- Roux Institute, Northeastern University, Portland, ME, USA
| |
Collapse
|
30
|
Shimoda M, Sato Y, Abe K, Masunaga N, Tsukabe M, Yoshinami T, Sota Y, Miyake T, Tanei T, Shimazu K. Prognostic value of serum tartrate‑resistant acid phosphatase‑5b for bone metastasis in patients with resectable breast cancer. Oncol Lett 2024; 27:250. [PMID: 38638841 PMCID: PMC11024733 DOI: 10.3892/ol.2024.14383] [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: 09/26/2023] [Accepted: 03/12/2024] [Indexed: 04/20/2024] Open
Abstract
Bone metastasis significantly affects the quality of life of patients with metastatic breast cancer, and can shorten overall survival. Identifying patients with early-stage breast cancer at high risk for bone metastasis and preventing bone metastasis may lead to a better quality of life and prolonged survival. The present study investigated whether serum tartrate-resistant acid phosphatase-5b (TRACP-5b), a bone turnover marker, can be a prognostic factor for bone metastasis. Female patients who underwent resectable breast surgery between May 2002 and August 2006 were consecutively investigated. A total of 304 patients with a median follow-up of 3,722 days were retrospectively analyzed. TRACP-5b levels in sera prepared from patients' blood drawn preoperatively without any presurgical treatments were measured using an enzyme-linked immunosorbent assay. The cutoff of TRACP-5b levels, in order to separate patients into high and low TRACP-5b groups, was set at median (347 mU/dl). The associations of clinicopathological factors, including TRACP-5b, with bone metastasis-free interval (BMFI), which was defined as the duration between surgery and the diagnosis of bone metastasis at any time point, were examined. Multivariate analysis of various clinicopathological features revealed that lymph node metastasis and histological grade were independent factors associated with BMFI (P=0.017 and 0.030, respectively). In patients with node-positive breast cancer (n=114), a high TRACP-5b level and a high grade were significantly and independently associated with worse BMFI (log-rank P=0.041 and 0.011, respectively). In conclusion, these findings indicated that TRACP-5b may predict bone metastasis in patients with node-positive breast cancer.
Collapse
Affiliation(s)
- Masafumi Shimoda
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yasufumi Sato
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Kaori Abe
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Nanae Masunaga
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Masami Tsukabe
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Tetsuhiro Yoshinami
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yoshiaki Sota
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Tomohiro Miyake
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Tomonori Tanei
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Kenzo Shimazu
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| |
Collapse
|
31
|
Arakil N, Akhund SA, Elaasser B, Mohammad KS. Intersecting Paths: Unraveling the Complex Journey of Cancer to Bone Metastasis. Biomedicines 2024; 12:1075. [PMID: 38791037 PMCID: PMC11117796 DOI: 10.3390/biomedicines12051075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 04/27/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
The phenomenon of bone metastases presents a significant challenge within the context of advanced cancer treatments, particularly pertaining to breast, prostate, and lung cancers. These metastatic occurrences stem from the dissemination of cancerous cells into the bone, thereby interrupting the equilibrium between osteoblasts and osteoclasts. Such disruption results in skeletal complications, adversely affecting patient morbidity and quality of life. This review discusses the intricate interplay between cancer cells and the bone microenvironment, positing the bone not merely as a passive recipient of metastatic cells but as an active contributor to cancer progression through its distinctive biochemical and cellular makeup. A thorough examination of bone structure and the dynamics of bone remodeling is undertaken, elucidating how metastatic cancer cells exploit these processes. This review explores the genetic and molecular pathways that underpin the onset and development of bone metastases. Particular emphasis is placed on the roles of cytokines and growth factors in facilitating osteoclastogenesis and influencing osteoblast activity. Additionally, this paper offers a meticulous critique of current diagnostic methodologies, ranging from conventional radiography to advanced molecular imaging techniques, and discusses the implications of a nuanced understanding of bone metastasis biology for therapeutic intervention. This includes the development of targeted therapies and strategies for managing bone pain and other skeletal-related events. Moreover, this review underscores the imperative of ongoing research efforts aimed at identifying novel therapeutic targets and refining management approaches for bone metastases. It advocates for a multidisciplinary strategy that integrates advancements in medical oncology and radiology with insights derived from molecular biology and genetics, to enhance prognostic outcomes and the quality of life for patients afflicted by this debilitating condition. In summary, bone metastases constitute a complex issue that demands a comprehensive and informed approach to treatment. This article contributes to the ongoing discourse by consolidating existing knowledge and identifying avenues for future investigation, with the overarching objective of ameliorating patient care in the domain of oncology.
Collapse
Affiliation(s)
| | | | | | - Khalid S. Mohammad
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 1153, Saudi Arabia; (N.A.); (S.A.A.); (B.E.)
| |
Collapse
|
32
|
Yang R, Jia L, Cui J. Mechanism and clinical progression of solid tumors bone marrow metastasis. Front Pharmacol 2024; 15:1390361. [PMID: 38770000 PMCID: PMC11102981 DOI: 10.3389/fphar.2024.1390361] [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: 02/23/2024] [Accepted: 04/22/2024] [Indexed: 05/22/2024] Open
Abstract
The rich blood supply of the bone marrow provides favorable conditions for tumor cell proliferation and growth. In the disease's early stages, circulating tumor cells can escape to the bone marrow and form imperceptible micro metastases. These tumor cells may be reactivated to regain the ability to grow aggressively and eventually develop into visible metastases. Symptomatic bone marrow metastases with abnormal hematopoiesis solid tumor metastases are rare and have poor prognoses. Treatment options are carefully chosen because of the suppression of bone marrow function. In this review, we summarized the mechanisms involved in developing bone marrow metastases from tumor cells and the clinical features, treatment options, and prognosis of patients with symptomatic bone marrow metastases from different solid tumors reported in the literature.
Collapse
|
33
|
Park-Min KH, Mun SH, Bockman R, McDonald MM. New Horizons: Translational Aspects of Osteomorphs. J Clin Endocrinol Metab 2024; 109:e1373-e1378. [PMID: 38060842 PMCID: PMC11031245 DOI: 10.1210/clinem/dgad711] [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: 07/06/2023] [Indexed: 04/21/2024]
Abstract
Osteomorphs are a newly described osteoclast lineage cell in mice, which are suggested to play a significant role in the maintenance of bone resorption. Preclinical investigations revealed that osteomorphs are generated through the fission of multinucleated bone-resorbing osteoclasts and can also re-fuse with existing osteoclasts. Modifications to RANKL signaling have been shown to alter cycles of fission and re-fusion of osteomorphs in mice. These novel findings were also shown to contribute to the rebound phenomenon after cessation of anti-RANKL therapy in mice. Moreover, the absence of osteomorph-specific genes in mice exhibits bone structural and quality phenotypes. Given these insights, it could be speculated that osteomorphs play a significant role in bone homeostasis, bone metabolic diseases, and response to therapeutics. In this review, we discuss these potential translational roles for osteomorphs. Importantly, we highlight the need for future preclinical and clinical studies to verify the presence of osteomorphs in humans and explore further the translational implications of this discovery.
Collapse
Affiliation(s)
- Kyung-Hyun Park-Min
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
- BCMB Allied Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10021, USA
| | - Se Hwan Mun
- Research Institute of Women’s Health, Sookmyung Women's University, 140-742 Seoul, Korea
| | - Richard Bockman
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
- Endocrine Service, Hospital for Special Surgery, New York, NY 10021, USA
| | - Michelle M McDonald
- Skeletal Diseases Program, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
34
|
Li X, Chen W, Liu D, Chen P, Wang S, Li F, Chen Q, Lv S, Li F, Chen C, Guo S, Yuan W, Li P, Hu Z. Pathological progression of osteoarthritis: a perspective on subchondral bone. Front Med 2024; 18:237-257. [PMID: 38619691 DOI: 10.1007/s11684-024-1061-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/17/2024] [Indexed: 04/16/2024]
Abstract
Osteoarthritis (OA) is a degenerative bone disease associated with aging. The rising global aging population has led to a surge in OA cases, thereby imposing a significant socioeconomic burden. Researchers have been keenly investigating the mechanisms underlying OA. Previous studies have suggested that the disease starts with synovial inflammation and hyperplasia, advancing toward cartilage degradation. Ultimately, subchondral-bone collapse, sclerosis, and osteophyte formation occur. This progression is deemed as "top to bottom." However, recent research is challenging this perspective by indicating that initial changes occur in subchondral bone, precipitating cartilage breakdown. In this review, we elucidate the epidemiology of OA and present an in-depth overview of the subchondral bone's physiological state, functions, and the varied pathological shifts during OA progression. We also introduce the role of multifunctional signal pathways (including osteoprotegerin (OPG)/receptor activator of nuclear factor-kappa B ligand (RANKL)/receptor activator of nuclear factor-kappa B (RANK), and chemokine (CXC motif) ligand 12 (CXCL12)/CXC motif chemokine receptor 4 (CXCR4)) in the pathology of subchondral bone and their role in the "bottom-up" progression of OA. Using vivid pattern maps and clinical images, this review highlights the crucial role of subchondral bone in driving OA progression, illuminating its interplay with the condition.
Collapse
Affiliation(s)
- Xuefei Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Wenhua Chen
- Research and Development Center of Chinese Medicine Resources and Biotechnology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dan Liu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Pinghua Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Shiyun Wang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Fangfang Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Qian Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Shunyi Lv
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Fangyu Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Chen Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Suxia Guo
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Weina Yuan
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Pan Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zhijun Hu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
35
|
Paindelli C, Parietti V, Barrios S, Shepherd P, Pan T, Wang WL, Satcher RL, Logothetis CJ, Navone N, Campbell MT, Mikos AG, Dondossola E. Bone mimetic environments support engineering, propagation, and analysis of therapeutic response of patient-derived cells, ex vivo and in vivo. Acta Biomater 2024; 178:83-92. [PMID: 38387748 PMCID: PMC12016311 DOI: 10.1016/j.actbio.2024.02.025] [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: 11/21/2023] [Revised: 01/22/2024] [Accepted: 02/15/2024] [Indexed: 02/24/2024]
Abstract
Bone metastases are the most common milestone in the lethal progression of prostate cancer and prominent in a substantial portion of renal malignancies. Interactions between cancer and bone host cells have emerged as drivers of both disease progression and therapeutic resistance. To best understand these central host-epithelial cell interactions, biologically relevant preclinical models are required. To achieve this goal, we here established and characterized tissue-engineered bone mimetic environments (BME) capable of supporting the growth of patient-derived xenograft (PDX) cells, ex vivo and in vivo. The BME consisted of a polycaprolactone (PCL) scaffold colonized by human mesenchymal stem cells (hMSCs) differentiated into osteoblasts. PDX-derived cells were isolated from bone metastatic prostate or renal tumors, engineered to express GFP or luciferase and seeded onto the BMEs. BMEs supported the growth and therapy response of PDX-derived cells, ex vivo. Additionally, BMEs survived after in vivo implantation and further sustained the growth of PDX-derived cells, their serial transplant, and their application to study the response to treatment. Taken together, this demonstrates the utility of BMEs in combination with patient-derived cells, both ex vivo and in vivo. STATEMENT OF SIGNIFICANCE: Our tissue-engineered BME supported the growth of patient-derived cells and proved useful to monitor the therapy response, both ex vivo and in vivo. This approach has the potential to enable co-clinical strategies to monitor bone metastatic tumor progression and therapy response, including identification and prioritization of new targets for patient treatment.
Collapse
Affiliation(s)
- Claudia Paindelli
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Vanessa Parietti
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Sergio Barrios
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States; Rice University, Department of Bioengineering, Houston, TX, 77030, United States
| | - Peter Shepherd
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Tianhong Pan
- Department of Orthopaedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Wei-Lien Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Robert L Satcher
- Department of Orthopaedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Christopher J Logothetis
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Nora Navone
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Matthew T Campbell
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States
| | - Antonios G Mikos
- Rice University, Department of Bioengineering, Houston, TX, 77030, United States
| | - Eleonora Dondossola
- Department of Genitourinary Medical Oncology and David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, United States.
| |
Collapse
|
36
|
Guo J, Ma RY, Qian BZ. Macrophage heterogeneity in bone metastasis. J Bone Oncol 2024; 45:100598. [PMID: 38585688 PMCID: PMC10997910 DOI: 10.1016/j.jbo.2024.100598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/16/2024] [Accepted: 03/20/2024] [Indexed: 04/09/2024] Open
Abstract
Previous studies illustrated that macrophage, a type of innate immune cell, plays critical roles in tumour progression and metastasis. Bone is the most frequent site of metastasis for several cancer types including breast, prostate, and lung. In bone metastasis, osteoclast, a macrophage subset specialized in bone resorption, was heavily investigated in the past. Recent studies illustrated that other macrophage subsets, e.g. monocyte-derived macrophages, and bone resident macrophages, promoted bone metastasis independent of osteoclast function. These novel mechanisms further improved our understanding of macrophage heterogeneity in the context of bone metastasis and illustrated new opportunities for future studies.
Collapse
Affiliation(s)
| | | | - Bin-Zhi Qian
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, The Human Phenome Institute, Zhangjiang-Fudan International Innovation Center, Fudan University, Shanghai 200438, China
| |
Collapse
|
37
|
Sandor LF, Huh JB, Benko P, Hiraga T, Poliska S, Dobo-Nagy C, Simpson JP, Homer NZM, Mahata B, Gyori DS. De novo steroidogenesis in tumor cells drives bone metastasis and osteoclastogenesis. Cell Rep 2024; 43:113936. [PMID: 38489269 PMCID: PMC10995766 DOI: 10.1016/j.celrep.2024.113936] [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] [Revised: 11/08/2023] [Accepted: 02/23/2024] [Indexed: 03/17/2024] Open
Abstract
Osteoclasts play a central role in cancer-cell-induced osteolysis, but the molecular mechanisms of osteoclast activation during bone metastasis formation are incompletely understood. By performing RNA sequencing on a mouse breast carcinoma cell line with higher bone-metastatic potential, here we identify the enzyme CYP11A1 strongly upregulated in osteotropic tumor cells. Genetic deletion of Cyp11a1 in tumor cells leads to a decreased number of bone metastases but does not alter primary tumor growth and lung metastasis formation in mice. The product of CYP11A1 activity, pregnenolone, increases the number and function of mouse and human osteoclasts in vitro but does not alter osteoclast-specific gene expression. Instead, tumor-derived pregnenolone strongly enhances the fusion of pre-osteoclasts via prolyl 4-hydroxylase subunit beta (P4HB), identified as a potential interaction partner of pregnenolone. Taken together, our results demonstrate that Cyp11a1-expressing tumor cells produce pregnenolone, which is capable of promoting bone metastasis formation and osteoclast development via P4HB.
Collapse
Affiliation(s)
- Luca F Sandor
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary
| | - Joon B Huh
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary
| | - Peter Benko
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary
| | - Toru Hiraga
- Department of Histology and Cell Biology, Matsumoto Dental University, Shiojiri, Nagano 399-0781, Japan
| | - Szilard Poliska
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Csaba Dobo-Nagy
- Department of Oral Diagnostics, Semmelweis University School of Dentistry, 1088 Budapest, Hungary
| | - Joanna P Simpson
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK
| | - Natalie Z M Homer
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK; University of Edinburgh/BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK
| | - Bidesh Mahata
- Department of Pathology, University of Cambridge, Cambridge CB21QP Cambridgeshire, UK
| | - David S Gyori
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary.
| |
Collapse
|
38
|
Sun K, Yuan L, Chen S, Sun Y, Wei D. Alendronate Pt IV Prodrug Amphiphile for Enhanced Chemotherapy Targeting and Bone Destruction Inhibition in Osteosarcoma. Adv Healthc Mater 2024; 13:e2302746. [PMID: 37988194 DOI: 10.1002/adhm.202302746] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/16/2023] [Indexed: 11/23/2023]
Abstract
Chemotherapy remains the primary treatment method for osteosarcoma after surgery. However, the lack of selectivity of chemotherapy for osteosarcoma leads to unpredictable therapeutic effects, undesirable side effects, and drug resistance. A platinum(IV) (PtIV ) prodrug amphiphile (ALN-PtIV -Lipo) covalently bound to alendronate (ALN) and a lipid tail is designed to overcome these limitations. ALN-PtIV -Lipo can self-assemble into PtIV lipid nanoparticles (APtIV ) for osteosarcoma targeting chemotherapy and bone destruction inhibition. It is demonstrated that APtIV achieved an eightfold increase in the eradication of osteosarcoma cells compared to cisplatin and threefold selective inhibition of osteosarcoma cells over breast cancer cells via APtIV in vitro. After intravenous injection, APtIV effectively accumulates at the osteosarcoma site in vivo, resulting in significantly suppressed primary osteosarcoma growth, and alleviation of bone destruction. Therefore, APtIV delivers a promising solution for enhanced chemotherapy targeting and bone destruction inhibition in osteosarcoma.
Collapse
Affiliation(s)
- Kaichuang Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266021, China
| | - Lu Yuan
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266021, China
| | - Shen Chen
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266021, China
| | - Dengshuai Wei
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
39
|
Monteran L, Zait Y, Erez N. It's all about the base: stromal cells are central orchestrators of metastasis. Trends Cancer 2024; 10:208-229. [PMID: 38072691 DOI: 10.1016/j.trecan.2023.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 03/16/2024]
Abstract
The tumor microenvironment (TME) is an integral part of tumors and plays a central role in all stages of carcinogenesis and progression. Each organ has a unique and heterogeneous microenvironment, which affects the ability of disseminated cells to grow in the new and sometimes hostile metastatic niche. Resident stromal cells, such as fibroblasts, osteoblasts, and astrocytes, are essential culprits in the modulation of metastatic progression: they transition from being sentinels of tissue integrity to being dysfunctional perpetrators that support metastatic outgrowth. Therefore, better understanding of the complexity of their reciprocal interactions with cancer cells and with other components of the TME is essential to enable the design of novel therapeutic approaches to prevent metastatic relapse.
Collapse
Affiliation(s)
- Lea Monteran
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yael Zait
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Neta Erez
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
40
|
Elaasser B, Arakil N, Mohammad KS. Bridging the Gap in Understanding Bone Metastasis: A Multifaceted Perspective. Int J Mol Sci 2024; 25:2846. [PMID: 38474093 PMCID: PMC10932255 DOI: 10.3390/ijms25052846] [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: 01/30/2024] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
The treatment of patients with advanced cancer poses clinical problems due to the complications that arise as the disease progresses. Bone metastases are a common problem that cancer patients may face, and currently, there are no effective drugs to treat these individuals. Prostate, breast, and lung cancers often spread to the bone, causing significant and disabling health conditions. The bone is a highly active and dynamic tissue and is considered a favorable environment for the growth of cancer. The role of osteoblasts and osteoclasts in the process of bone remodeling and the way in which their interactions change during the progression of metastasis is critical to understanding the pathophysiology of this disease. These interactions create a self-perpetuating loop that stimulates the growth of metastatic cells in the bone. The metabolic reprogramming of both cancer cells and cells in the bone microenvironment has serious implications for the development and progression of metastasis. Insight into the process of bone remodeling and the systemic elements that regulate this process, as well as the cellular changes that occur during the progression of bone metastases, is critical to the discovery of a cure for this disease. It is crucial to explore different therapeutic options that focus specifically on malignancy in the bone microenvironment in order to effectively treat this disease. This review will focus on the bone remodeling process and the effects of metabolic disorders as well as systemic factors like hormones and cytokines on the development of bone metastases. We will also examine the various therapeutic alternatives available today and the upcoming advances in novel treatments.
Collapse
Affiliation(s)
| | | | - Khalid S. Mohammad
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 1153, Saudi Arabia; (B.E.); (N.A.)
| |
Collapse
|
41
|
Liu H, Liu L, Rosen CJ. PTH and the Regulation of Mesenchymal Cells within the Bone Marrow Niche. Cells 2024; 13:406. [PMID: 38474370 PMCID: PMC10930661 DOI: 10.3390/cells13050406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Parathyroid hormone (PTH) plays a pivotal role in maintaining calcium homeostasis, largely by modulating bone remodeling processes. Its effects on bone are notably dependent on the duration and frequency of exposure. Specifically, PTH can initiate both bone formation and resorption, with the outcome being influenced by the manner of PTH administration: continuous or intermittent. In continuous administration, PTH tends to promote bone resorption, possibly by regulating certain genes within bone cells. Conversely, intermittent exposure generally favors bone formation, possibly through transient gene activation. PTH's role extends to various aspects of bone cell activity. It directly influences skeletal stem cells, osteoblastic lineage cells, osteocytes, and T cells, playing a critical role in bone generation. Simultaneously, it indirectly affects osteoclast precursor cells and osteoclasts, and has a direct impact on T cells, contributing to its role in bone resorption. Despite these insights, the intricate mechanisms through which PTH acts within the bone marrow niche are not entirely understood. This article reviews the dual roles of PTH-catabolic and anabolic-on bone cells, highlighting the cellular and molecular pathways involved in these processes. The complex interplay of these factors in bone remodeling underscores the need for further investigation to fully comprehend PTH's multifaceted influence on bone health.
Collapse
Affiliation(s)
- Hanghang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China;
- Maine Medical Center, MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA;
| | - Linyi Liu
- Maine Medical Center, MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA;
| | - Clifford J. Rosen
- Maine Medical Center, MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA;
| |
Collapse
|
42
|
Young SAE, Heller AD, Garske DS, Rummler M, Qian V, Ellinghaus A, Duda GN, Willie BM, Grüneboom A, Cipitria A. From breast cancer cell homing to the onset of early bone metastasis: The role of bone (re)modeling in early lesion formation. SCIENCE ADVANCES 2024; 10:eadj0975. [PMID: 38381833 PMCID: PMC10881061 DOI: 10.1126/sciadv.adj0975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 01/18/2024] [Indexed: 02/23/2024]
Abstract
Breast cancer often metastasizes to bone, causing osteolytic lesions. Structural and biophysical changes are rarely studied yet are hypothesized to influence metastasis. We developed a mouse model of early bone metastasis and multimodal imaging to quantify cancer cell homing, bone (re)modeling, and onset of metastasis. Using tissue clearing and three-dimensional (3D) light sheet fluorescence microscopy, we located enhanced green fluorescent protein-positive cancer cells and small clusters in intact bones and quantified their size and spatial distribution. We detected early bone lesions using in vivo microcomputed tomography (microCT)-based time-lapse morphometry and revealed altered bone (re)modeling in the absence of detectable lesions. With a new microCT image analysis tool, we tracked the growth of early lesions over time. We showed that cancer cells home in all bone compartments, while osteolytic lesions are only detected in the metaphysis, a region of high (re)modeling. Our study suggests that higher rates of (re)modeling act as a driver of lesion formation during early metastasis.
Collapse
Affiliation(s)
- Sarah A. E. Young
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Anna-Dorothea Heller
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Daniela S. Garske
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Maximilian Rummler
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Research Centre, Shriners Hospital for Children–Canada, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| | - Victoria Qian
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Agnes Ellinghaus
- Julius Wolff Institute, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Germany
| | - Georg N. Duda
- Julius Wolff Institute, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Germany
| | - Bettina M. Willie
- Research Centre, Shriners Hospital for Children–Canada, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| | - Anika Grüneboom
- Leibniz-Institute for Advancing Analytics – ISAS – e.V., Dortmund, Germany
| | - Amaia Cipitria
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Group of Bioengineering in Regeneration and Cancer, Biodonostia Health Research Institute, San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
43
|
Searcy MB, Johnson RW. Epigenetic control of the vicious cycle. J Bone Oncol 2024; 44:100524. [PMID: 38304486 PMCID: PMC10830514 DOI: 10.1016/j.jbo.2024.100524] [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: 11/28/2023] [Accepted: 01/09/2024] [Indexed: 02/03/2024] Open
Abstract
Epigenetic alterations, including DNA methylation and post translational modifications to histones, drive tumorigenesis and metastatic progression. In the context of bone metastasis, epigenetic modifications in tumor cells can modulate dissemination of cancer cells to the bone, tumor progression in the bone marrow, and may be associated with patient survival rates. Bone disseminated tumor cells may enter a dormant state or stimulate osteolysis through the "vicious cycle" of bone metastasis where bone disseminated tumor cells disrupt the bone microenvironment, which fuels tumor progression. Epigenetic alterations may either exacerbate or abrogate the vicious cycle by regulating tumor suppressors and oncogenes, which alter proliferation of bone-metastatic cancer cells. This review focuses on the specific epigenetic alterations that regulate bone metastasis, including DNA methylation, histone methylation, and histone acetylation. Here, we summarize key findings from researchers identifying epigenetic changes that drive tumor progression in the bone, along with pre-clinical and clinical studies investigating the utility of targeting aberrant epigenetic alterations to treat bone metastatic cancer.
Collapse
Affiliation(s)
- Madeline B. Searcy
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachelle W. Johnson
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
44
|
Yang J, Cheng Y, Nie Y, Tian B, Huang J, Gong R, Li Z, Zhu J, Gong Y. TRPC5 expression promotes the proliferation and invasion of papillary thyroid carcinoma through the HIF-1α/Twist pathway. Transl Oncol 2024; 39:101809. [PMID: 37918167 PMCID: PMC10638037 DOI: 10.1016/j.tranon.2023.101809] [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: 07/14/2023] [Revised: 09/30/2023] [Accepted: 10/15/2023] [Indexed: 11/04/2023] Open
Abstract
OBJECTIVE This study aimed to investigate the effect of TRPC5 on PTC (papillary thyroid carcinoma) proliferation and invasion. METHODS Immunofluorescence and western blot were used to evaluate the expression of TRPC5 in paraffin sections and clinical tissues. Overexpression and silencing of TRPC5 to generate the cells for in vitro experiments. Wound-healing assay, transwell invasion assay, MTT assay, and in vivo tumorigenicity assay were used to determine cell proliferation and cell migration in vitro and in vivo. Real-time PCR was used to test the expression of TRPC5. Western blot was used to test the expression of downstream factors: E-cadherin, Vimentin, MMP-9, MMP-2, TRPC5, ZEB, Snail, and Twist. RESULTS The level of TRPC5 protein expression was higher in PTC than in adjacent normal thyroid tissue. TPC-1 cells overexpressing TRPC5 were more proliferative, had longer migration distances, and increased the number of invading cells. TPC-1 cells silenced with TRPC5 had a weaker proliferation capacity, shorter migration distances, and a reduced number of invading cells. Overexpression and silencing of TRPC5 modulated E-cadherin, Vimentin, MMP-9, MMP-2, TRPC5, and Twist, but did not affect ZEB and Snail. The results of tumor formation experiments in nude mice showed that inhibition of TRPC5 expression suppressed the volume and weight of transplanted tumors. CONCLUSION TRPC5 induced papillary thyroid cancer metastasis and progression via up-regulated HIF-1α signaling in vivo and in vitro. High TRPC5 expression is a biomarker for lymph node metastasis at its early stages.
Collapse
Affiliation(s)
- Jing Yang
- Department of Thyroid Surgery, West China Hospital, Sichuan University, China; Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yue Cheng
- Department of Otorhinolaryngology-Head and Neck Surgery, Sichuan Electric Power Hospital, China
| | - Yan Nie
- West China School of Medicine, Sichuan University, China
| | - Bole Tian
- Department of pancreatic Surgery, West China Hospital, Sichuan University, China
| | - Jing Huang
- Department of Thyroid Surgery, West China Hospital, Sichuan University, China; Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Rixiang Gong
- Department of Thyroid Surgery, West China Hospital, Sichuan University, China; Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhihui Li
- Department of Thyroid Surgery, West China Hospital, Sichuan University, China; Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingqiang Zhu
- Department of Thyroid Surgery, West China Hospital, Sichuan University, China; Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yanping Gong
- Department of Thyroid Surgery, West China Hospital, Sichuan University, China; Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
45
|
Xu K, Li J, Wen R, Chang B, Cheng Y, Yi X. Role of SIRT3 in bone homeostasis and its application in preventing and treating bone diseases. Front Pharmacol 2023; 14:1248507. [PMID: 38192409 PMCID: PMC10773770 DOI: 10.3389/fphar.2023.1248507] [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: 06/27/2023] [Accepted: 12/11/2023] [Indexed: 01/10/2024] Open
Abstract
Bone homeostasis refers to the balance between osteoblast-mediated bone formation and osteoclast-mediated bone resorption and the maintenance of stable bone mass. SIRT3 is a class of mitochondrial protein deacetylase that influences various mitochondrial functions and is involved in the mechanisms underlying resistance to aging; regulation of bone marrow mesenchymal stem cells, osteoblasts, and osteoclasts; and development of osteoporosis, osteoarthritis, and other bone diseases. Moreover, exercise affects bones through SIRT3. Thus, studies on SIRT3 may provide insights for the treatment of bone diseases. Although SIRT3 can exert multiple effects on bone, the specific mechanism by which it regulates bone homeostasis remains unclear. By evaluating the relevant literature, this review discusses the structure and function of SIRT3, reveals the role and associated mechanisms of SIRT3 in regulating bone homeostasis and mediating bone health during exercise, and highlights the potential pharmacological value of SIRT3 in treating bone diseases.
Collapse
Affiliation(s)
- Ke Xu
- School of Sports Health, Shenyang Sport University, Shenyang, China
| | - Jing Li
- School of Physical Education, Liaoning Normal University, Dalian, China
| | - Ruiming Wen
- School of Sports Health, Shenyang Sport University, Shenyang, China
| | - Bo Chang
- School of Sports Health, Shenyang Sport University, Shenyang, China
| | - Yang Cheng
- School of Sports Health, Shenyang Sport University, Shenyang, China
| | - Xuejie Yi
- School of Sports Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
46
|
Zhang G, Lu Y, Song J, Huang D, An M, Chen W, Han P, Yao X, Zhang X. A multifunctional nano-hydroxyapatite/MXene scaffold for the photothermal/dynamic treatment of bone tumours and simultaneous tissue regeneration. J Colloid Interface Sci 2023; 652:1673-1684. [PMID: 37666199 DOI: 10.1016/j.jcis.2023.08.176] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/13/2023] [Accepted: 08/28/2023] [Indexed: 09/06/2023]
Abstract
After resection of bone tumour, the risk of cancer recurrence and numerous bone defects continues to threaten the health of patients. To overcome the challenge, we developed a novel multifunctional scaffold material consisting mainly of nano-hydroxyapatite particles (n-HA), MXene nanosheets and g-C3N4 to prevent tumour recurrence and promote bone formation. N-HA has the potential to restrict the growth of osteosarcoma cells, and the combination of MXene and g-C3N4 enables the scaffolds to produce photodynamic and photothermal effects simultaneously under near infrared (NIR) irradiation. Surprisingly, n-HA can further enhance the synergistic anti-tumour function of photodynamic and photothermal, and the scaffolds can eradicate osteosarcoma cells in only 10 min at a mild temperature of 45 ℃. Moreover, the scaffold exhibit exceptional cytocompatibility and possesses the capacity to induce osteogenic differentiation of bone marrow mesenchymal stem cells. Therefore, this multifunctional scaffold can not only inhibits the proliferation of bone tumour cells and rapidly eradicate bone tumour through NIR irradiation, but also enhances osteogenic activity. This promising measure can be used to treat tissue damage after bone tumour resection.
Collapse
Affiliation(s)
- Guannan Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China; Shanxi Provincial Key Laboratory for Translational Nuclear Medicine and Precision Protection, Taiyuan 030006, China
| | - Ying Lu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China; Shanxi Provincial Key Laboratory for Translational Nuclear Medicine and Precision Protection, Taiyuan 030006, China
| | - Jianbo Song
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China; Shanxi Provincial Key Laboratory for Translational Nuclear Medicine and Precision Protection, Taiyuan 030006, China.
| | - Di Huang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, China
| | - Meiwen An
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China.
| | - Weiyi Chen
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, China
| | - Peide Han
- College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Xiaohong Yao
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Xiangyu Zhang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China.
| |
Collapse
|
47
|
Langsten KL, Shi L, Wilson AS, Lumia S, Westwood B, Skeen AM, Xie MT, Surratt VE, Turner J, Langefeld CD, Singh R, Cook KL, Kerr BA. A Novel Metastatic Estrogen Receptor-Expressing Breast Cancer Model with Antiestrogen Responsiveness. Cancers (Basel) 2023; 15:5773. [PMID: 38136319 PMCID: PMC10742098 DOI: 10.3390/cancers15245773] [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: 07/31/2023] [Revised: 11/28/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Most women diagnosed with breast cancer (BC) have estrogen receptor alpha-positive (ER+) disease. The current mouse models of ER+ BC often rely on exogenous estrogen to encourage metastasis, which modifies the immune system and the function of some tissues like bone. Other studies use genetically modified or immunocompromised mouse strains, which do not accurately replicate the clinical disease. To create a model of antiestrogen responsive BC with spontaneous metastasis, we developed a mouse model of 4T1.2 triple-negative (TN) breast cancer with virally transduced ER expression that metastasizes spontaneously without exogenous estrogen stimulation and is responsive to antiestrogen drugs. Our mouse model exhibited upregulated ER-responsive genes and multi-organ metastasis without exogenous estrogen administration. Additionally, we developed a second TN BC cell line, E0771/bone, to express ER, and while it expressed ER-responsive genes, it lacked spontaneous metastasis to clinically important tissues. Following antiestrogen treatment (tamoxifen, ICI 182,780, or vehicle control), 4T1.2- and E0771/bone-derived tumor volumes and weights were significantly decreased, exemplifying antiestrogen responsivity in both cell lines. This 4T1.2 tumor model, which expresses the estrogen receptor, metastasizes spontaneously, and responds to antiestrogen treatment, will allow for further investigation into the biology and potential treatment of metastasis.
Collapse
Affiliation(s)
- Kendall L. Langsten
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (K.L.L.); (L.S.); (S.L.); (A.M.S.); (M.T.X.); (V.E.S.); (J.T.); (R.S.); (K.L.C.)
| | - Lihong Shi
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (K.L.L.); (L.S.); (S.L.); (A.M.S.); (M.T.X.); (V.E.S.); (J.T.); (R.S.); (K.L.C.)
| | - Adam S. Wilson
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (A.S.W.); (B.W.)
| | - Salvatore Lumia
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (K.L.L.); (L.S.); (S.L.); (A.M.S.); (M.T.X.); (V.E.S.); (J.T.); (R.S.); (K.L.C.)
| | - Brian Westwood
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (A.S.W.); (B.W.)
| | - Alexandra M. Skeen
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (K.L.L.); (L.S.); (S.L.); (A.M.S.); (M.T.X.); (V.E.S.); (J.T.); (R.S.); (K.L.C.)
| | - Maria T. Xie
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (K.L.L.); (L.S.); (S.L.); (A.M.S.); (M.T.X.); (V.E.S.); (J.T.); (R.S.); (K.L.C.)
| | - Victoria E. Surratt
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (K.L.L.); (L.S.); (S.L.); (A.M.S.); (M.T.X.); (V.E.S.); (J.T.); (R.S.); (K.L.C.)
| | - JoLyn Turner
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (K.L.L.); (L.S.); (S.L.); (A.M.S.); (M.T.X.); (V.E.S.); (J.T.); (R.S.); (K.L.C.)
| | - Carl D. Langefeld
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA;
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA
| | - Ravi Singh
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (K.L.L.); (L.S.); (S.L.); (A.M.S.); (M.T.X.); (V.E.S.); (J.T.); (R.S.); (K.L.C.)
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA
| | - Katherine L. Cook
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (K.L.L.); (L.S.); (S.L.); (A.M.S.); (M.T.X.); (V.E.S.); (J.T.); (R.S.); (K.L.C.)
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (A.S.W.); (B.W.)
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA
| | - Bethany A. Kerr
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; (K.L.L.); (L.S.); (S.L.); (A.M.S.); (M.T.X.); (V.E.S.); (J.T.); (R.S.); (K.L.C.)
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA
| |
Collapse
|
48
|
Zhang F, Chen F, Wang C, Zhou FH. The functional roles of m6A modification in prostate cancer. Proteomics Clin Appl 2023; 17:e2200108. [PMID: 37070355 DOI: 10.1002/prca.202200108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/19/2023]
Abstract
Prostate cancer (PCa) is the most prevalent malignancy of the male genitourinary system, and its etiology suggests that genetics is an essential risk factor for its development and progression, while exogenous factors may have an significant impact on this risk. Initial diagnosis of advanced PCa is relatively frequent, and androgen deprivation therapy (ADT) is the predominant standard of care for PCa and the basis for various novel combination therapy regimens, and is often required throughout the patient's subsequent treatment. Although diagnostic modalities and treatment options are evolving, some patients suffer from complications, including biochemical relapse, metastasis and treatment resistance. Mechanisms of PCa pathogenesis and progression have been the focus of research. N6-methyladenosine (m6A) is an RNA modification involved in cell physiology and tumor metabolism. It has been observed to affect the evolution of diverse cancers through the regulation of gene expression. Genes associated with m6A are prominent in PCa and are involved in multiple aspects of desmoresistant PCa occurrence, progression, PCa bone metastasis (BM), and treatment resistance. Here, we explore the role of m6A modifications in promoting PCa.
Collapse
Affiliation(s)
- Fa Zhang
- Department of Urology, Gansu Provincial People's Hospital, Lanzhou, Gansu, China
| | - Feng Chen
- Department of Anaesthesiology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Chao Wang
- Department of Anaesthesiology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Feng-Hai Zhou
- Department of Urology, Gansu Provincial People's Hospital, Lanzhou, Gansu, China
| |
Collapse
|
49
|
Choi S, Whitman MA, Shimpi AA, Sempertegui ND, Chiou AE, Druso JE, Verma A, Lux SC, Cheng Z, Paszek M, Elemento O, Estroff LA, Fischbach C. Bone-matrix mineralization dampens integrin-mediated mechanosignalling and metastatic progression in breast cancer. Nat Biomed Eng 2023; 7:1455-1472. [PMID: 37550422 DOI: 10.1038/s41551-023-01077-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/30/2023] [Indexed: 08/09/2023]
Abstract
In patients with breast cancer, lower bone mineral density increases the risk of bone metastasis. Although the relationship between bone-matrix mineralization and tumour-cell phenotype in breast cancer is not well understood, mineralization-induced rigidity is thought to drive metastatic progression via increased cell-adhesion forces. Here, by using collagen-based matrices with adjustable intrafibrillar mineralization, we show that, unexpectedly, matrix mineralization dampens integrin-mediated mechanosignalling and induces a less proliferative stem-cell-like phenotype in breast cancer cells. In mice with xenografted decellularized physiological bone matrices seeded with human breast tumour cells, the presence of bone mineral reduced tumour growth and upregulated a gene-expression signature that is associated with longer metastasis-free survival in patients with breast cancer. Our findings suggest that bone-matrix changes in osteogenic niches regulate metastatic progression in breast cancer and that in vitro models of bone metastasis should integrate organic and inorganic matrix components to mimic physiological and pathologic mineralization.
Collapse
Affiliation(s)
- Siyoung Choi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Matthew A Whitman
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Adrian A Shimpi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Nicole D Sempertegui
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Aaron E Chiou
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Joseph E Druso
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Akanksha Verma
- Caryl and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Stephanie C Lux
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Zhu Cheng
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA
| | - Matthew Paszek
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY, USA
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Lara A Estroff
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY, USA.
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, USA.
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
50
|
Singh N, Marak J, Singh DK, Verma S. Follicular carcinoma of the thyroid presenting as metastasis in the wall of an arachnoid cyst. BMJ Case Rep 2023; 16:e255865. [PMID: 37907313 PMCID: PMC10618989 DOI: 10.1136/bcr-2023-255865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023] Open
Abstract
The brain is an uncommon site for metastases of differentiated thyroid carcinoma with the most common location being cerebral hemispheres, followed by cerebellum and pituitary gland. Metastasis in the wall of an arachnoid cyst is exceedingly rare with single case report available in the published literature. Arachnoid cyst metastasis from an extraneuraxial malignancy has not been published until. We present a unique case of thyroid carcinoma metastasizing to the wall of an intracranial arachnoid cyst and the most interesting fact is that it was the first clinical manifestation of her malignancy.
Collapse
Affiliation(s)
- Neha Singh
- Radiodiagnosis & Imaging, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow, India
| | - James Marak
- Radiodiagnosis & Imaging, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow, India
| | - Deepak Kumar Singh
- Neurosurgery, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow, India
| | - Shashwat Verma
- Nuclear Medicine, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow, India
| |
Collapse
|