1
|
Wasi M, Chu T, Guerra RM, Kooker R, Maldonado K, Li X, Lin CY, Song X, Xiong J, You L, Wang L. Mitigating aging and doxorubicin induced bone loss in mature mice via mechanobiology based treatments. Bone 2024; 188:117235. [PMID: 39147353 PMCID: PMC11475016 DOI: 10.1016/j.bone.2024.117235] [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: 06/13/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Aging leads to a reduced anabolic response to mechanical stimuli and a loss of bone mass and structural integrity. Chemotherapy agents such as doxorubicin exacerbate the degeneration of aging skeleton and further subject older cancer patients to a higher fracture risk. To alleviate this clinical problem, we proposed and tested a novel mechanobiology-based therapy. Building upon prior findings that i) Yoda1, the Piezo1 agonist, promoted bone growth in young adult mice and suppressed bone resorption markers in aged mice, and ii) moderate tibial loading protected bone from breast cancer-induced osteolysis, we hypothesized that combined Yoda1 and moderate loading would improve the structural integrity of adult and aged skeletons in vivo and protect bones from deterioration after chemotherapy. We first examined the effects of 4-week Yoda1 (dose 5 mg/kg, 5 times/week) and moderate tibial loading (4.5 N peak load, 4 Hz, 300 cycles for 5 days/week), individually and combined, on mature mice (∼50 weeks of age). Combined Yoda1 and loading was found to mitigate age-associated cortical and trabecular bone loss better than individual interventions. As expected, the non-treated controls experienced an average drop of cortical polar moment of inertia (Ct.pMOI) by -4.3 % over four weeks and the bone deterioration occurred in the majority (64 %) of the samples. Relative to no treatment, loading alone, Yoda1 alone, and combined Yoda1 and loading increased Ct.pMOI by +7.3 %, +9.5 %, +12.0 % and increased the % of samples with positive Ct.pMOI changes by +32 %, +26 %, and +43 %, respectively, suggesting an additive protection of aging-related bone loss for the combined therapy. We further tested if the treatment efficacy was preserved in mature mice following two weeks (six injections) of doxorubicin at the dose of 2.5 or 5 mg/kg. As expected, doxorubicin increased osteocyte apoptosis, altered bone remodeling, and impaired bone structure. However, the effects induced by DOX were too severe to be rescued by Yoda1 and loading, alone or combined, although loading and Yoda1 individually, or combined, increased the number of mice showing positive responsiveness by 0 %, +15 %, and +29 % relative to no intervention after doxorubicin exposure. Overall, this study supported the potentials and challenges of the Yoda1-based strategy in mitigating the detrimental skeletal effects caused by aging and doxorubicin.
Collapse
Affiliation(s)
- Murtaza Wasi
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Tiankuo Chu
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Rosa M Guerra
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - Rory Kooker
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Kenneth Maldonado
- Department of Biomedical Engineering, Kansas State University, Manhattan, KS, USA
| | - Xuehua Li
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Chun-Yu Lin
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Xin Song
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Jinhu Xiong
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Lidan You
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA; Department of Biomedical Engineering, University of Delaware, Newark, DE, USA.
| |
Collapse
|
2
|
Ouyang P, Cheng B, He X, Lou J, Li X, Guo H, Xu F. Navigating the biophysical landscape: how physical cues steer the journey of bone metastatic tumor cells. Trends Cancer 2024; 10:792-808. [PMID: 39127608 DOI: 10.1016/j.trecan.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 08/12/2024]
Abstract
Many tumors prefer to metastasize to bone, but the underlying mechanisms remain elusive. The human skeletal system has unique physical properties, that are distinct from other organs, which play a key role in directing the behavior of tumor cells within bone. Understanding the physical journey of tumor cells within bone is crucial. In this review we discuss bone metastasis in the context of how physical cues in the bone vasculature and bone marrow niche regulate the fate of tumor cells. Our objective is to inspire innovative diagnostic and therapeutic approaches for bone metastasis from a mechanobiological perspective.
Collapse
Affiliation(s)
- Pengrong Ouyang
- Department of Orthopedic Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P.R. China; Bioinspired Engineering and Biomechanics Center (BEBC), Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China
| | - Bo Cheng
- Bioinspired Engineering and Biomechanics Center (BEBC), Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China; TFX Group-Xi'an Jiaotong University Institute of Life Health, Xi'an 710049, P.R. China
| | - Xijing He
- Department of Orthopedic Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P.R. China; Xi'an International Medical Center Hospital, Xi'an 710061, P.R. China.
| | - Jiatao Lou
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China.
| | - Xiaokang Li
- Department of Orthopedics, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, P.R. China.
| | - Hui Guo
- Department of Medical Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, P.R. China.
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China.
| |
Collapse
|
3
|
Creecy A, Segvich D, Metzger C, Kohler R, Wallace JM. Combining anabolic loading and raloxifene improves bone quantity and some quality measures in a mouse model of osteogenesis imperfecta. Bone 2024; 184:117106. [PMID: 38641232 PMCID: PMC11130993 DOI: 10.1016/j.bone.2024.117106] [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: 01/25/2024] [Revised: 03/20/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
Osteogenesis imperfecta (OI) increases fracture risk due to changes in bone quantity and quality caused by mutations in collagen and its processing proteins. Current therapeutics improve bone quantity, but do not treat the underlying quality deficiencies. Male and female G610C+/- mice, a murine model of OI, were treated with a combination of raloxifene and in vivo axial tibial compressive loading starting at 10 weeks of age and continuing for 6 weeks to improve bone quantity and quality. Bone geometry and mechanical properties were measured to determine whole bone and tissue-level material properties. A colocalized Raman/nanoindentation system was used to measure chemical composition and nanomechanical properties in newly formed bone compared to old bone to determine if bone formed during the treatment regimen differed in quality compared to bone formed prior to treatment. Lastly, lacunar geometry and osteocyte apoptosis were assessed. OI mice were able to build bone in response to the loading, but this response was less robust than in control mice. Raloxifene improved some bone material properties in female but not male OI mice. Raloxifene did not alter nanomechanical properties, but loading did. Lacunar geometry was largely unchanged with raloxifene and loading. However, osteocyte apoptosis was increased with loading in raloxifene treated female mice. Overall, combination treatment with raloxifene and loading resulted in positive but subtle changes to bone quality.
Collapse
Affiliation(s)
- Amy Creecy
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, United States of America.
| | - Dyann Segvich
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, United States of America
| | - Corinne Metzger
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Rachel Kohler
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, United States of America
| | - Joseph M Wallace
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, United States of America
| |
Collapse
|
4
|
Wei C, Shi M, Wang Z, Lan W, Feng N, Zhang F, Liu J, Lang JY, Lin W, Ma W. Epiberberine inhibits bone metastatic breast cancer-induced osteolysis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 327:118039. [PMID: 38479545 DOI: 10.1016/j.jep.2024.118039] [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: 01/08/2024] [Revised: 02/20/2024] [Accepted: 03/09/2024] [Indexed: 03/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The anti-tumor related diseases of Coptidis Rhizoma (Huanglian) were correlated with its traditional use of removing damp-heat, clearing internal fire, and counteracting toxicity. In the recent years, Coptidis Rhizoma and its components have drawn extensive attention toward their anti-tumor related diseases. Besides, Coptidis Rhizoma is traditionally used as an anti-inflammatory herb. Epiberberine (EPI) is a significant alkaloid isolated from Coptidis Rhizoma, and exhibits multiple pharmacological activities including anti-inflammatory. However, the effect of epiberberine on breast cancer and the inflammatory factors of metastatic breast cancer-induced osteolysis has not been demonstrated clearly. AIM OF THE STUDY Bone metastatic breast cancer can lead to osteolysis via inflammatory factors-induced osteoclast differentiation and function. In this study, we try to analyze the effect of epiberberine on breast cancer and the inflammatory factors of metastatic breast cancer-induced osteolysis. METHODS To evaluate whether epiberberine could suppress bone metastatic breast cancer-induced osteolytic damage, healthy female Balb/c mice were intratibially injected with murine triple-negative breast cancer 4T1 cells. Then, we examined the inhibitory effect and underlying mechanism of epiberberine on breast cancer-induced osteoclastogenesis in vitro. Xenograft assay was used to study the effect of epiberberine on breast cancer cells in vivo. Moreover, we also studied the inhibitory effects and underlying mechanisms of epiberberine on RANKL-induced osteoclast differentiation and function in vitro. RESULTS The results show that epiberberine displayed potential therapeutic effects on breast cancer-induced osteolytic damage. Besides, our results show that epiberberine inhibited breast cancer cells-induced osteoclast differentiation and function by inhibiting secreted inflammatory cytokines such as IL-8. Importantly, we found that epiberberine directly inhibited RANKL-induced differentiation and function of osteoclast without cytotoxicity. Mechanistically, epiberberine inhibited RANKL-induced osteoclastogensis via Akt/c-Fos signaling pathway. Furthermore, epiberberine combined with docetaxel effectively protected against bone loss induced by metastatic breast cancer cells. CONCLUSIONS Our findings suggested that epiberberine may be a promising natural compound for treating bone metastatic breast cancer-induced osteolytic damage by inhibiting IL-8 and is worthy of further exploration in preclinical and clinical trials.
Collapse
Affiliation(s)
- Chengming Wei
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, 999078, Macau
| | - Meina Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, 999078, Macau
| | - Zi Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, 999078, Macau
| | - Wenjian Lan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Na Feng
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, Guangdong, 529020, China
| | - Fuming Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, 999078, Macau
| | - Jiachen Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, 999078, Macau
| | - Jing-Yu Lang
- The CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wanjun Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, 999078, Macau
| | - Wenzhe Ma
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, 999078, Macau.
| |
Collapse
|
5
|
Kumar V, Naqvi SM, Verbruggen A, McEvoy E, McNamara LM. A mechanobiological model of bone metastasis reveals that mechanical stimulation inhibits the pro-osteolytic effects of breast cancer cells. Cell Rep 2024; 43:114043. [PMID: 38642336 DOI: 10.1016/j.celrep.2024.114043] [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: 12/01/2023] [Accepted: 03/19/2024] [Indexed: 04/22/2024] Open
Abstract
Bone is highly susceptible to cancer metastasis, and both tumor and bone cells enable tumor invasion through a "vicious cycle" of biochemical signaling. Tumor metastasis into bone also alters biophysical cues to both tumor and bone cells, which are highly sensitive to their mechanical environment. However, the mechanobiological feedback between these cells that perpetuate this cycle has not been studied. Here, we develop highly advanced in vitro and computational models to provide an advanced understanding of how tumor growth is regulated by the synergistic influence of tumor-bone cell signaling and mechanobiological cues. In particular, we develop a multicellular healthy and metastatic bone model that can account for physiological mechanical signals within a custom bioreactor. These models successfully recapitulated mineralization, mechanobiological responses, osteolysis, and metastatic activity. Ultimately, we demonstrate that mechanical stimulus provided protective effects against tumor-induced osteolysis, confirming the importance of mechanobiological factors in bone metastasis development.
Collapse
Affiliation(s)
- Vatsal Kumar
- Mechanobiology and Medical Device Research Group (MMDRG), Biomedical Engineering, College of Science and Engineering, University of Galway, H91 HX31 Galway, Ireland
| | - Syeda M Naqvi
- Mechanobiology and Medical Device Research Group (MMDRG), Biomedical Engineering, College of Science and Engineering, University of Galway, H91 HX31 Galway, Ireland
| | - Anneke Verbruggen
- Mechanobiology and Medical Device Research Group (MMDRG), Biomedical Engineering, College of Science and Engineering, University of Galway, H91 HX31 Galway, Ireland
| | - Eoin McEvoy
- Biomedical Engineering, College of Science and Engineering, University of Galway, H91 HX31 Galway, Ireland
| | - Laoise M McNamara
- Mechanobiology and Medical Device Research Group (MMDRG), Biomedical Engineering, College of Science and Engineering, University of Galway, H91 HX31 Galway, Ireland.
| |
Collapse
|
6
|
Xie J, Xu Y, Liu X, Long L, Chen J, Huang C, Shao Y, Cai Z, Zhang Z, Zhou R, Leng J, Bai X, Song Q. Mechanically stimulated osteocytes maintain tumor dormancy in bone metastasis of non-small cell lung cancer by releasing small extracellular vesicles. eLife 2024; 12:RP89613. [PMID: 38547196 PMCID: PMC10977966 DOI: 10.7554/elife.89613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024] Open
Abstract
Although preclinical and clinical studies have shown that exercise can inhibit bone metastasis progression, the mechanism remains poorly understood. Here, we found that non-small cell lung cancer (NSCLC) cells adjacent to bone tissue had a much lower proliferative capacity than the surrounding tumor cells in patients and mice. Subsequently, it was demonstrated that osteocytes, sensing mechanical stimulation generated by exercise, inhibit NSCLC cell proliferation and sustain the dormancy thereof by releasing small extracellular vesicles with tumor suppressor micro-RNAs, such as miR-99b-3p. Furthermore, we evaluated the effects of mechanical loading and treadmill exercise on the bone metastasis progression of NSCLC in mice. As expected, mechanical loading of the tibia inhibited the bone metastasis progression of NSCLC. Notably, bone metastasis progression of NSCLC was inhibited by moderate exercise, and combinations with zoledronic acid had additive effects. Moreover, exercise preconditioning effectively suppressed bone metastasis progression. This study significantly advances the understanding of the mechanism underlying exercise-afforded protection against bone metastasis progression.
Collapse
Affiliation(s)
- Jing Xie
- General Practice Centre, The Seventh Affiliated Hospital, Southern Medical UniversityFoshanChina
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Yafei Xu
- General Practice Centre, The Seventh Affiliated Hospital, Southern Medical UniversityFoshanChina
| | - Xuhua Liu
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Li Long
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Ji Chen
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Chunyan Huang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Yan Shao
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Zhiqing Cai
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Zhimin Zhang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Ruixin Zhou
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Jiarong Leng
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical UniversityGuangzhouChina
| | - Xiaochun Bai
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Qiancheng Song
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical UniversityGuangzhouChina
| |
Collapse
|
7
|
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 DOI: 10.3390/ijms25052846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [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)
- Basant Elaasser
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 1153, Saudi Arabia
| | - Nour Arakil
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 1153, Saudi Arabia
| | - Khalid S Mohammad
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 1153, Saudi Arabia
| |
Collapse
|
8
|
Verbruggen SW. Role of the osteocyte in bone metastasis - The importance of networking. J Bone Oncol 2024; 44:100526. [PMID: 38304485 PMCID: PMC10831278 DOI: 10.1016/j.jbo.2024.100526] [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: 11/18/2023] [Revised: 12/19/2023] [Accepted: 01/16/2024] [Indexed: 02/03/2024] Open
Abstract
Metastatic bone disease is a complex condition resulting from the migration and colonization of cancer cells from their primary site to the bone microenvironment, where they typically develop a metastatic niche. Osteocytes, the most abundant cells in bone tissue and the master regulators of bone remodelling, are increasingly thought to play a crucial role in this process through intricate interactions with cancer cells. This review covers the recent progress made in exploring the multifaceted interactions between osteocytes and cancer cells in the metastatic microenvironment, highlighting the importance of signalling networks in bone metastases. Though these interactions are particularly complex, the renewed focus of researchers on osteocytes within the last 5 years has uncovered multiple new potential molecular mechanisms underlying osteocyte-mediated regulation of cancer cell survival, proliferation, and invasion. A number of key papers will be discussed in detail, emphasizing the significance of signalling pathways and molecular crosstalk, and exploring potential therapeutic strategies targeting osteocyte-cancer cell interactions to improve patient treatment and outcomes.
Collapse
Affiliation(s)
- Stefaan W. Verbruggen
- Centre for Predictive in vitro Models and Centre for Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, United Kingdom
- Digital Environment Research Institute, Queen Mary University of London, United Kingdom
- INSIGNEO Institute for in silico Medicine and Department of Mechanical Engineering, University of Sheffield, United Kingdom
| |
Collapse
|
9
|
Sarazin BA, Liu B, Goldman E, Whitefield AN, Lynch ME. Bone-homing metastatic breast cancer cells impair osteocytes' mechanoresponse in a 3D loading model. Heliyon 2023; 9:e20248. [PMID: 37767467 PMCID: PMC10520780 DOI: 10.1016/j.heliyon.2023.e20248] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/23/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Breast cancer predominantly metastasizes to the skeleton. Mechanical loading is reliably anabolic in bone, and also inhibits bone metastatic tumor formation and bone loss in vivo. To study the underlying mechanisms, we developed a 3D culture model for osteocytes, the primary bone mechanosensor. We verified that MLO-Y4s responded to perfusion by reducing their rankl and rankl:opg gene expression. We next cultured MLO-Y4s with tumor-conditioned media (TCM) collected from human breast cancer cells (MDA-MB-231s) and a corresponding bone-homing subclone to test the impacts on osteocytes' mechanosensation. We found that TCM from the bone-homing subclone was more detrimental to MLO-Y4 growth and viability, and it abrogated loading-induced changes to rankl:opg. Our studies demonstrate that MLO-Y4s, including their mechanoresponse to perfusion, were more negatively impacted by soluble factors from bone-homing breast cancer cells compared to those from parental cells.
Collapse
Affiliation(s)
- Blayne A. Sarazin
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, 80309, USA
| | - Boyuan Liu
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, 80309, USA
| | - Elaine Goldman
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, 80309, USA
| | - Ashlyn N. Whitefield
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, 80309, USA
| | - Maureen E. Lynch
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, 80309, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO, 80309, USA
| |
Collapse
|
10
|
Zhao B, Lv Y. Suspension state and shear stress enhance breast tumor cells EMT through YAP by microRNA-29b. Cell Biol Toxicol 2023; 39:1037-1052. [PMID: 34618275 DOI: 10.1007/s10565-021-09661-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/24/2021] [Indexed: 12/11/2022]
Abstract
Except for biochemical effects, suspension state (Sus) is proved to induce epithelial-mesenchymal transition (EMT) of circulating tumor cells (CTCs) mechanically. However, the difference between the effects of the mechanical microenvironment in capillaries (simplified as shear stress (SS) and Sus) and single Sus on EMT is unclear, nor the underlying mechanism. Here, breast tumor cells (BTCs) were loaded with Sus and SS to mimic the situation of CTCs stimulated by these two kinds of mechanics. It was demonstrated that the EMT of BTCs was enhanced by Sus and SS and the mechanotransductor yes-associated protein (YAP) was partially cytoplasmic stored with microRNA (miR)-29b decreased, which was detected by miR sequencing. Though it couldn't possess a feedback regulation, YAP promoted miR-29b expression and posttranscriptionally regulated BTCs EMT through miR-29b, where transforming growth factor β involved. Analysis of clinical database showed that high miR-29b expression was beneficial to high survival rate stabilizing its role of tumor suppressor. This study discovers the mechanism that Sus and SS promote BTCs EMT by YAP through miR-29b posttranscriptionally and highlight the potential of YAP and miR-29b in tumor therapy. The combination of suspension state and shear stress promotes transforming growth factor β involved epithelial-mesenchymal transition by yes-associated protein through microRNA-29b.
Collapse
Affiliation(s)
- Boyuan Zhao
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China
| | - Yonggang Lv
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China.
| |
Collapse
|
11
|
Anloague A, Delgado-Calle J. Osteocytes: New Kids on the Block for Cancer in Bone Therapy. Cancers (Basel) 2023; 15:2645. [PMID: 37174109 PMCID: PMC10177382 DOI: 10.3390/cancers15092645] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
The tumor microenvironment plays a central role in the onset and progression of cancer in the bone. Cancer cells, either from tumors originating in the bone or from metastatic cancer cells from other body systems, are located in specialized niches where they interact with different cells of the bone marrow. These interactions transform the bone into an ideal niche for cancer cell migration, proliferation, and survival and cause an imbalance in bone homeostasis that severely affects the integrity of the skeleton. During the last decade, preclinical studies have identified new cellular mechanisms responsible for the dependency between cancer cells and bone cells. In this review, we focus on osteocytes, long-lived cells residing in the mineral matrix that have recently been identified as key players in the spread of cancer in bone. We highlight the most recent discoveries on how osteocytes support tumor growth and promote bone disease. Additionally, we discuss how the reciprocal crosstalk between osteocytes and cancer cells provides the opportunity to develop new therapeutic strategies to treat cancer in the bone.
Collapse
Affiliation(s)
- Aric Anloague
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Jesus Delgado-Calle
- Department of Physiology and Cell Biology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
12
|
Li K, Huo Q, Dimmitt NH, Qu G, Bao J, Pandya PH, Saadatzadeh MR, Bijangi-Vishehsaraei K, Kacena MA, Pollok KE, Lin CC, Li BY, Yokota H. Osteosarcoma-enriched transcripts paradoxically generate osteosarcoma-suppressing extracellular proteins. eLife 2023; 12:e83768. [PMID: 36943734 PMCID: PMC10030111 DOI: 10.7554/elife.83768] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 03/01/2023] [Indexed: 03/23/2023] Open
Abstract
Osteosarcoma (OS) is the common primary bone cancer that affects mostly children and young adults. To augment the standard-of-care chemotherapy, we examined the possibility of protein-based therapy using mesenchymal stem cells (MSCs)-derived proteomes and OS-elevated proteins. While a conditioned medium (CM), collected from MSCs, did not present tumor-suppressing ability, the activation of PKA converted MSCs into induced tumor-suppressing cells (iTSCs). In a mouse model, the direct and hydrogel-assisted administration of CM inhibited tumor-induced bone destruction, and its effect was additive with cisplatin. CM was enriched with proteins such as calreticulin, which acted as an extracellular tumor suppressor by interacting with CD47. Notably, the level of CALR transcripts was elevated in OS tissues, together with other tumor-suppressing proteins, including histone H4, and PCOLCE. PCOLCE acted as an extracellular tumor-suppressing protein by interacting with amyloid precursor protein, a prognostic OS marker with poor survival. The results supported the possibility of employing a paradoxical strategy of utilizing OS transcriptomes for the treatment of OS.
Collapse
Affiliation(s)
- Kexin Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical UniversityHarbinChina
- Department of Biomedical Engineering, Indiana University Purdue University IndianapolisIndianapolisUnited States
| | - Qingji Huo
- Department of Pharmacology, School of Pharmacy, Harbin Medical UniversityHarbinChina
- Department of Biomedical Engineering, Indiana University Purdue University IndianapolisIndianapolisUnited States
| | - Nathan H Dimmitt
- Department of Biomedical Engineering, Indiana University Purdue University IndianapolisIndianapolisUnited States
| | - Guofan Qu
- Department of Orthopedic Surgery, Harbin Medical University Cancer HospitalHarbinChina
| | - Junjie Bao
- Department of Orthopedic Surgery, Harbin Medical University Cancer HospitalHarbinChina
| | - Pankita H Pandya
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of MedicineIndianapolisUnited States
- Department of Pediatrics, Indiana University School of MedicineIndianapolisUnited States
| | - M Reza Saadatzadeh
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of MedicineIndianapolisUnited States
- Department of Pediatrics, Indiana University School of MedicineIndianapolisUnited States
| | | | - Melissa A Kacena
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of MedicineIndianapolisUnited States
- Department of Orthopaedic Surgery, Indiana University School of MedicineIndianapolisUnited States
- Indiana Center for Musculoskeletal Health, Indiana University School of MedicineIndianapolisUnited States
| | - Karen E Pollok
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of MedicineIndianapolisUnited States
- Department of Pediatrics, Indiana University School of MedicineIndianapolisUnited States
| | - Chien-Chi Lin
- Department of Biomedical Engineering, Indiana University Purdue University IndianapolisIndianapolisUnited States
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of MedicineIndianapolisUnited States
| | - Bai-Yan Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical UniversityHarbinChina
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University Purdue University IndianapolisIndianapolisUnited States
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of MedicineIndianapolisUnited States
- Indiana Center for Musculoskeletal Health, Indiana University School of MedicineIndianapolisUnited States
| |
Collapse
|
13
|
Lin CY, Song X, Seaman K, You L. Microfluidic Co-culture Platforms for Studying Osteocyte Regulation of Other Cell Types under Dynamic Mechanical Stimulation. Curr Osteoporos Rep 2022; 20:478-492. [PMID: 36149593 DOI: 10.1007/s11914-022-00748-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/26/2022] [Indexed: 01/30/2023]
Abstract
PURPOSE OF REVIEW Osteocytes are the most abundant cell type in bone. These unique cells act primarily as mechanosensors and play crucial roles in the functional adaptation of bone tissue. This review aims to summarize the recent microfluidic studies on mechanically stimulated osteocytes in regulating other cell types. RECENT FINDINGS Microfluidics is a powerful technology that has been widely employed in recent years. With the advantages of microfluidic platforms, researchers can mimic multicellular environments and integrate dynamic systems to study osteocyte regulation under mechanical stimulation. Microfluidic platforms have been developed to investigate mechanically stimulated osteocytes in the direct regulation of multiple cell types, including osteoclasts, osteoblasts, and cancer cells, and in the indirect regulation of cancer cells via endothelial cells. Overall, these microfluidic studies foster the development of treatment approaches targeting osteocytes under mechanical stimulation.
Collapse
Affiliation(s)
- Chun-Yu Lin
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Xin Song
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Kimberly Seaman
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Lidan You
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
14
|
Ihle CL, Wright-Hobart SJ, Owens P. Therapeutics targeting the metastatic breast cancer bone microenvironment. Pharmacol Ther 2022; 239:108280. [DOI: 10.1016/j.pharmthera.2022.108280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/30/2022] [Accepted: 09/12/2022] [Indexed: 11/27/2022]
|
15
|
Matsumoto T, Mukohara A. Effects of Whole-Body Vibration on Breast Cancer Bone Metastasis and Vascularization in Mice. Calcif Tissue Int 2022; 111:535-545. [PMID: 35896728 DOI: 10.1007/s00223-022-01009-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/07/2022] [Indexed: 11/12/2022]
Abstract
We evaluated whether whole-body vibration (WBV) prevented bone loss induced by breast cancer (BC) metastasis and the involvement of bone marrow vasculature. One day after orthotopic transplantation of mammary 4T1 tumor cells, 8-week-old BALB/c mice were subjected to 0.3 g/90 Hz vertical vibration for 20 min/day for 5 days/week (BC-WBV) or sham-handled (BC-Sham) over 3 weeks. Age-matched intact mice (Intact) were also sham-handled. Both tibiae were harvested from BC-WBV (n = 7), BC-Sham (n = 9), and Intact (n = 5) mice for bone structure imaging by synchrotron radiation-based computed tomography (SRCT) and hematoxylin and eosin staining, whereas right tibiae were harvested from other BC-WBV and BC-Sham (n = 6 each) mice for vascular imaging by SRCT. Tumor cells were similarly widespread in the marrow in BC-WBV and BC-Sham mice. In BC-Sham mice, cortical bone volume, trabecular volume fraction, trabecular thickness, trabecular number density, and bone mineral density were smaller, and marrow volume and trabecular separation were larger than in Intact mice. However, although trabecular thickness was smaller in BC-WBV than Intact mice, the others did not differ between the two groups. Serum osteocalcin tended to be higher in BC-WBV than BC-Sham mice. Compared with BC-Sham mice, BC-WBV mice had a smaller vessel diameter, a trend of a larger vessel number density, and smaller vessel diameter heterogeneity. In conclusion, WBV mitigates bone loss in BC bone metastasis, which may be partly due to increased bone anabolism. The alteration of marrow vasculature appears to be favorable for anti-tumor drug delivery. Further studies are needed to clarify the multiple actions of WBV on bone, tumor, and marrow vasculature and how they contribute to bone protection in BC metastasis.
Collapse
Affiliation(s)
- Takeshi Matsumoto
- Biomedical Engineering Laboratory, Tokushima University Graduate School of Technology, Industrial and Social Sciences, 770-8506, Tokushima, Japan.
| | - Akihiro Mukohara
- Biomedical Engineering Laboratory, Tokushima University Faculty of Science and Technology, 770-8506, Tokushima, Japan
| |
Collapse
|
16
|
Feng Y, Liu S, Zha R, Sun X, Li K, Wu D, Aryal UK, Koch M, Li BY, Yokota H. Prostate cancer-associated urinary proteomes differ before and after prostatectomy. Ther Adv Med Oncol 2022; 14:17588359221131532. [PMID: 36324734 PMCID: PMC9618752 DOI: 10.1177/17588359221131532] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/22/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND A wide range of disorders can be detected in the urine. Tumor-modifying proteins in the urine may serve as a diagnostic tool for cancer patients and the alterations in their profiles may indicate efficacies of chemotherapy, radiotherapy, and surgery. METHODS We focused on urinary proteomes of patients with prostate cancer and identified tumor-modifying proteins in the samples before and after prostatectomy. Protein array analysis was conducted to evaluate a differential profile of tumor-promoting cytokines, while mass spectrometry-based global proteomics was conducted to identify tumor-suppressing proteins. RESULTS The result revealed striking differences by prostatectomy. Notably, the urine from the post-prostatectomy significantly decreased the tumorigenic behaviors of prostate tumor cells as well as breast cancer cells. We observed that angiogenin, a stimulator of blood vessel formation, was reduced in the post-prostatectomy urine. By contrast, the levels of three cell-membrane proteins such as prostasin (PRSS8), nectin 2 (PVRL2), and nidogen 1 (NID1) were elevated and they acted as extracellular tumor-suppressing proteins. These three proteins, given extracellularly, downregulated tumorigenic genes such as Runx2, Snail, and transforming growth factor beta and induced apoptosis of tumor cells. However, the role of NID1 differed depending on the location, and intracellular NID1 was tumorigenic and reduced the percent survival. CONCLUSIONS This study demonstrated that prostatectomy remarkably altered the profile of urinary proteomes, and the post-prostatectomy urine provided tumor-suppressive proteomes. The result sheds novel light on the dynamic nature of the urinary proteomes and a unique strategy for predicting tumor suppressors.
Collapse
Affiliation(s)
| | | | - Rongrong Zha
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China,Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Xun Sun
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China,Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Kexin Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China,Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Di Wu
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China,Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Uma K. Aryal
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | - Michael Koch
- Department of Urology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bai-Yan Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, #157 Baojian Road, Harbin, Heilongjiang 150081, China
| | | |
Collapse
|
17
|
Zheng A, Zhang L, Yang J, Yin X, Zhang T, Wu X, Ma X. Physical activity prevents tumor metastasis through modulation of immune function. Front Pharmacol 2022; 13:1034129. [PMID: 36313283 PMCID: PMC9596782 DOI: 10.3389/fphar.2022.1034129] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 09/20/2022] [Indexed: 01/10/2023] Open
Abstract
Metastasis is responsible for 90% of deaths in cancer patients. Most patients diagnosed with metastatic cancer will die within 5 years. PA is good for health and has become an emerging adjuvant therapy for cancer survivors. Regular moderate exercise substantially lowers the incidence and recurrence of several cancers, alleviates cancer-related adverse events, enhances the efficacy of anti-cancer treatments, and improves the quality of life of cancer patients. Revealing the mechanisms of PA inhibiting tumor metastasis could upgrade our understanding of cancer biology and help researchers explore new therapeutic strategies to improve survival in cancer patients. However, it remains poorly understood how physical activity prevents metastasis by modulating tumor behavior. The immune system is involved in each step of tumor metastasis. From invasion to colonization, immune cells interact with tumor cells to secret cytokines and proteases to remodel the tumor microenvironment. Substantial studies demonstrated the ability of physical activity to induce antitumor effects of immune cells. This provides the possibility that physical activity can modulate immune cells behavior to attenuate tumor metastasis. The purpose of this review is to discuss and summarize the critical link between immune function and exercise in metastasis prevention.
Collapse
Affiliation(s)
- Aiping Zheng
- Division of Biotherapy, Cancer Center, West China Hospital, Cancer Center, Sichuan University, Chengdu, China
- Head and Neck Oncology Ward, Cancer Center, West China Hospital, Cancer Center, Sichuan University, Chengdu, China
| | - Lei Zhang
- Department of Obstetrics & Gynecology, Chengdu First People’s Hospital & Chengdu Integrated TCM & Western Medicine Hospital, Chengdu, China
| | - Jiaqing Yang
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaomeng Yin
- Division of Biotherapy, Cancer Center, West China Hospital, Cancer Center, Sichuan University, Chengdu, China
| | - Tao Zhang
- Division of Biotherapy, Cancer Center, West China Hospital, Cancer Center, Sichuan University, Chengdu, China
| | - Xin Wu
- Head and Neck Oncology Ward, Cancer Center, West China Hospital, Cancer Center, Sichuan University, Chengdu, China
- Head and Neck Oncology Ward, Division of Radiotherapy Oncology, Cancer Center, West China Hospital, Chengdu, China
- *Correspondence: Xin Wu, ; Xuelei Ma,
| | - Xuelei Ma
- Division of Biotherapy, Cancer Center, West China Hospital, Cancer Center, Sichuan University, Chengdu, China
- *Correspondence: Xin Wu, ; Xuelei Ma,
| |
Collapse
|
18
|
Ma C, Du T, Niu X, Fan Y. Biomechanics and mechanobiology of the bone matrix. Bone Res 2022; 10:59. [PMID: 36042209 PMCID: PMC9427992 DOI: 10.1038/s41413-022-00223-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 05/13/2022] [Accepted: 05/27/2022] [Indexed: 11/23/2022] Open
Abstract
The bone matrix plays an indispensable role in the human body, and its unique biomechanical and mechanobiological properties have received much attention. The bone matrix has unique mechanical anisotropy and exhibits both strong toughness and high strength. These mechanical properties are closely associated with human life activities and correspond to the function of bone in the human body. None of the mechanical properties exhibited by the bone matrix is independent of its composition and structure. Studies on the biomechanics of the bone matrix can provide a reference for the preparation of more applicable bone substitute implants, bone biomimetic materials and scaffolds for bone tissue repair in humans, as well as for biomimetic applications in other fields. In providing mechanical support to the human body, bone is constantly exposed to mechanical stimuli. Through the study of the mechanobiology of the bone matrix, the response mechanism of the bone matrix to its surrounding mechanical environment can be elucidated and used for the health maintenance of bone tissue and defect regeneration. This paper summarizes the biomechanical properties of the bone matrix and their biological significance, discusses the compositional and structural basis by which the bone matrix is capable of exhibiting these mechanical properties, and studies the effects of mechanical stimuli, especially fluid shear stress, on the components of the bone matrix, cells and their interactions. The problems that occur with regard to the biomechanics and mechanobiology of the bone matrix and the corresponding challenges that may need to be faced in the future are also described.
Collapse
Affiliation(s)
- Chunyang Ma
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Tianming Du
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China
| | - Xufeng Niu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China. .,Research Institute of Beihang University in Shenzhen, Shenzhen, 518057, China.
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China. .,School of Engineering Medicine, Beihang University, Beijing, 100083, China.
| |
Collapse
|
19
|
Sun X, Li K, Li BY, Yokota H. Wnt signaling: a double-edged sword in protecting bone from cancer. J Bone Miner Metab 2022; 41:365-370. [PMID: 36040520 DOI: 10.1007/s00774-022-01363-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/29/2022] [Indexed: 11/30/2022]
Abstract
Wnt signaling plays a critical role in loading-driven bone formation and bone homeostasis, whereas its activation in cancer cells promotes their progression. Currently, major research efforts in cancer treatment have been directed to the development of Wnt inhibitors. Recent studies on tumor-bone interactions, however, presented multiple lines of evidence that support a tumor-suppressive role of Lrp5, a Wnt co-receptor, and β-catenin, in Wnt signaling. This review describes the action of Wnt signaling as a double-edged sword in the bone microenvironment and suggests the possibility of a novel option for protecting bone from cancer.
Collapse
Affiliation(s)
- Xun Sun
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, 723 West Michigan Street, SL220, Indianapolis, IN, 46202, USA
| | - Kexin Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, 723 West Michigan Street, SL220, Indianapolis, IN, 46202, USA
| | - Bai-Yan Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, 723 West Michigan Street, SL220, Indianapolis, IN, 46202, USA.
- Simon Comprehensive Cancer Center, Indianapolis, IN, 46202, USA.
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
20
|
Zhang Y, Habibovic P. Delivering Mechanical Stimulation to Cells: State of the Art in Materials and Devices Design. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2110267. [PMID: 35385176 DOI: 10.1002/adma.202110267] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/19/2022] [Indexed: 06/14/2023]
Abstract
Biochemical signals, such as growth factors, cytokines, and transcription factors are known to play a crucial role in regulating a variety of cellular activities as well as maintaining the normal function of different tissues and organs. If the biochemical signals are assumed to be one side of the coin, the other side comprises biophysical cues. There is growing evidence showing that biophysical signals, and in particular mechanical cues, also play an important role in different stages of human life ranging from morphogenesis during embryonic development to maturation and maintenance of tissue and organ function throughout life. In order to investigate how mechanical signals influence cell and tissue function, tremendous efforts have been devoted to fabricating various materials and devices for delivering mechanical stimuli to cells and tissues. Here, an overview of the current state of the art in the design and development of such materials and devices is provided, with a focus on their design principles, and challenges and perspectives for future research directions are highlighted.
Collapse
Affiliation(s)
- Yonggang Zhang
- Department of Instructive Biomaterials Engineering, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, Maastricht, 6229 ER, The Netherlands
| | - Pamela Habibovic
- Department of Instructive Biomaterials Engineering, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, Maastricht, 6229 ER, The Netherlands
| |
Collapse
|
21
|
Lin CY, Song X, Ke Y, Raha A, Wu Y, Wasi M, Wang L, Geng F, You L. Yoda1 Enhanced Low-Magnitude High-Frequency Vibration on Osteocytes in Regulation of MDA-MB-231 Breast Cancer Cell Migration. Cancers (Basel) 2022; 14:3395. [PMID: 35884459 PMCID: PMC9324638 DOI: 10.3390/cancers14143395] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 02/05/2023] Open
Abstract
Low-magnitude (≤1 g) high-frequency (≥30 Hz) (LMHF) vibration has been shown to enhance bone mineral density. However, its regulation in breast cancer bone metastasis remains controversial for breast cancer patients and elder populations. Yoda1, an activator of the mechanosensitive Piezo1 channel, could potentially intensify the effect of LMHF vibration by enhancing the mechanoresponse of osteocytes, the major mechanosensory bone cells with high expression of Piezo1. In this study, we treated osteocytes with mono- (Yoda1 only or vibration only) or combined treatment (Yoda1 and LMHF vibration) and examined the further regulation of osteoclasts and breast cancer cells through the conditioned medium. Moreover, we studied the effects of combined treatment on breast cancer cells in regulation of osteocytes. Combined treatment on osteocytes showed beneficial effects, including increasing the nuclear translocation of Yes-associated protein (YAP) in osteocytes (488.0%, p < 0.0001), suppressing osteoclastogenesis (34.3%, p = 0.004), and further reducing migration of MDA-MB-231 (15.1%, p = 0.02) but not Py8119 breast cancer cells (4.2%, p = 0.66). Finally, MDA-MB-231 breast cancer cells subjected to the combined treatment decreased the percentage of apoptotic osteocytes (34.5%, p = 0.04) but did not affect the intracellular calcium influx. This study showed the potential of stimulating Piezo1 in enhancing the mechanoresponse of osteocytes to LMHF vibration and further suppressing breast cancer migration via osteoclasts.
Collapse
Affiliation(s)
- Chun-Yu Lin
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; (C.-Y.L.); (Y.K.)
| | - Xin Song
- Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada;
| | - Yaji Ke
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; (C.-Y.L.); (Y.K.)
| | - Arjun Raha
- W Booth School of Engineering Practice and Technology, McMaster University, Hamilton, ON L8S 4L7, Canada; (A.R.); (Y.W.); (F.G.)
| | - Yuning Wu
- W Booth School of Engineering Practice and Technology, McMaster University, Hamilton, ON L8S 4L7, Canada; (A.R.); (Y.W.); (F.G.)
| | - Murtaza Wasi
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA; (M.W.); (L.W.)
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA; (M.W.); (L.W.)
| | - Fei Geng
- W Booth School of Engineering Practice and Technology, McMaster University, Hamilton, ON L8S 4L7, Canada; (A.R.); (Y.W.); (F.G.)
| | - Lidan You
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; (C.-Y.L.); (Y.K.)
- Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada;
| |
Collapse
|
22
|
Sun X, Li K, Hase M, Zha R, Feng Y, Li BY, Yokota H. Suppression of breast cancer-associated bone loss with osteoblast proteomes via Hsp90ab1/moesin-mediated inhibition of TGFβ/FN1/CD44 signaling. Am J Cancer Res 2022; 12:929-943. [PMID: 34976221 PMCID: PMC8692912 DOI: 10.7150/thno.66148] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/22/2021] [Indexed: 01/04/2023] Open
Abstract
Background: Bone is a frequent site of metastases from breast cancer, but existing therapeutic options are not satisfactory. Although osteoblasts have active roles in cancer progression by assisting the vicious bone-destructive cycle, we employed a counterintuitive approach of activating pro-tumorigenic Wnt signaling and examined the paradoxical possibility of developing osteoblast-derived tumor-suppressive, bone-protective secretomes. Methods: Wnt signaling was activated by the overexpression of Lrp5 and β-catenin in osteoblasts as well as a pharmacological agent (BML284), and the therapeutic effects of their conditioned medium (CM) were evaluated using in vitro cell cultures, ex vivo breast cancer tissues, and a mouse model of osteolysis. To explore the unconventional regulatory mechanism of the action of Wnt-activated osteoblasts, whole-genome proteomics analysis was conducted, followed by immunoprecipitation and gain- and loss-of-function assays. Results: While osteoblasts did not present any innate tumor-suppressing ability, we observed that the overexpression of Lrp5 and β-catenin in Wnt signaling made their CM tumor-suppressive and bone-protective. The growth of breast cancer cells and tissues was inhibited by Lrp5-overexpressing CM (Lrp5 CM), which suppressed mammary tumors and tumor-driven bone destruction in a mouse model. Lrp5 CM also inhibited the differentiation and maturation of bone-resorbing osteoclasts by downregulating NFATc1 and cathepsin K. The overexpression of Lrp5 upregulated osteopontin that enriched Hsp90ab1 (Hsp90 beta) and moesin (MSN) in Lrp5 CM. Hsp90ab1 and MSN are atypical tumor-suppressing proteins since they are multi-tasking, moonlighting proteins that promote tumorigenesis in tumor cells. Importantly, Hsp90ab1 immuno-precipitated latent TGFβ and inactivated TGFβ, whereas MSN interacted with CD44, a cancer stem-cell marker, as well as fibronectin 1, an ECM protein. Furthermore, Hsp90ab1 and MSN downregulated KDM3A that demethylated histones, together with PDL1 that inhibited immune responses. Conclusion: In contrast to inducing tumor-enhancing secretomes and chemoresistance in general by inhibiting varying oncogenic pathways in chemotherapy, this study presented the unexpected outcome of generation tumor-suppressive secretomes by activating the pro-tumorigenic Wnt pathway. The results shed light on the contrasting role of oncogenic signaling in tumor cells and osteoblast-derived secretomes, suggesting a counterintuitive option for the treatment of breast cancer-associated bone metastasis.
Collapse
Affiliation(s)
- Xun Sun
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China,Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Kexin Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China,Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Misato Hase
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA,Graduate School of Engineering, Mie University, Mie 514, Japan
| | - Rongrong Zha
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China,Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Yan Feng
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China,Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Bai-Yan Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China,✉ Corresponding authors: Bai-Yan Li and Hiroki Yokota
| | - Hiroki Yokota
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China,Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA,Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA,✉ Corresponding authors: Bai-Yan Li and Hiroki Yokota
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW While the function of osteocytes under physiologic conditions is well defined, their role and involvement in cancer disease remains relatively unexplored, especially in a context of non-bone metastatic cancer. This review will focus on describing the more advanced knowledge regarding the interactions between osteocytes and cancer. RECENT FINDINGS We will discuss the involvement of osteocytes in the onset and progression of osteosarcoma, with the common bone cancers, as well as the interaction that is established between osteocytes and multiple myeloma. Mechanisms responsible for cancer dissemination to bone, as frequently occur with advanced breast and prostate cancers, will be reviewed. While a role for osteocytes in the stimulation and proliferation of cancer cells has been reported, protective effects of osteocytes against bone colonization have been described as well, thus increasing ambiguity regarding the role of osteocytes in cancer progression and dissemination. Lastly, supporting the idea that skeletal defects can occur also in the absence of direct cancer dissemination or osteolytic lesions directly adjacent to the bone, our recent findings will be presented showing that in the absence of bone metastases, the bone microenvironment and, particularly, osteocytes, can manifest a clear and dramatic response to the distant, non-metastatic tumor. Our observations support new studies to clarify whether treatments designed to preserve the osteocytes can be combined with traditional anticancer therapies, even when bone is not directly affected by tumor growth.
Collapse
Affiliation(s)
- Fabrizio Pin
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Matt Prideaux
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lynda F Bonewald
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrea Bonetto
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Otolaryngology-Head & Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Surgery, Indiana University School of Medicine, 980 W Walnut Street, R3-C522, Indianapolis, IN, 46202, USA.
- Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
24
|
Wang S, Pei S, Wasi M, Parajuli A, Yee A, You L, Wang L. Moderate tibial loading and treadmill running, but not overloading, protect adult murine bone from destruction by metastasized breast cancer. Bone 2021; 153:116100. [PMID: 34246808 PMCID: PMC8478818 DOI: 10.1016/j.bone.2021.116100] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 06/18/2021] [Accepted: 06/29/2021] [Indexed: 12/15/2022]
Abstract
Osteolytic bone lesions, which develop in many metastatic breast cancer patients, impair bone integrity and lead to adverse skeletal related events that are difficult to treat and sometimes fatal. Moderate mechanical loading has been shown to suppress osteolysis in young mice with breast cancer. In this study, we aimed to investigate the dose-dependent effects of mechanical loading on protecting the integrity of adult skeletons with breast cancer. Localized tibial loading and aerobic treadmill running with three levels of varying intensity were tested in a syngeneic mammary tumor bone metastasis model. Adult C57BL/6J female mice (14-week-old, N = 88 mice) received intra-tibial injections of Py8119 triple-negative murine breast cancer cells or PBS and underwent 4 to 5 weeks of exercise or acted as sedentary/non-loaded controls. The bone structure was monitored longitudinally with weekly in vivo micro-computed tomography imaging, while the cellular responses in bone and marrow were examined using immunohistochemistry. Moderate treadmill running (16 m/min, 50 min/day, 5 days/week, and 5 weeks) and tibial loading (4.5 N, 630 με, 4 Hz, 300 cycles/day, 5 days/week, and 4 weeks) suppressed tumor-induced bone destruction, as evaluated by full-thickness perforation of tibial cortex and the volume of osteolytic lesions in the cortex. In contrast, tibial loading at higher magnitude (8 N, 1100 με) induced woven bone and accelerated bone destruction, compared with the non-loaded controls. The three exercise regimens differentially affected osteocyte apoptosis, osteocyte hypoxia, osteoclast activity, bone marrow vasculature, and tumor proliferation. In conclusion, the relationship between exercise intensity and the risk of breast cancer-induced osteolysis was found to follow a J-shaped curve in a preclinical model, suggesting the need to optimize exercise parameters in order to harness the skeletal benefits of exercise in metastatic breast cancers.
Collapse
Affiliation(s)
- Shubo Wang
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Shaopeng Pei
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Murtaza Wasi
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Ashutosh Parajuli
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Albert Yee
- Division of Orthopaedics, Department of Surgery, Sunnybrook Health Sciences Centre and the University of Toronto, Toronto, Ontario, Canada
| | - Lidan You
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Liyun Wang
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA.
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW In this review, we provide an overview of what is currently known about the impacts of mechanical stimuli on metastatic tumor-induced bone disease (TIBD). Further, we focus on the role of the osteocyte, the skeleton's primary mechanosensory cell, which is central to the skeleton's mechanoresponse, sensing and integrating local mechanical stimuli, and then controlling the downstream remodeling balance as appropriate. RECENT FINDINGS Exercise and controlled mechanical loading have anabolic effects on bone tissue in models of bone metastasis. They also have anti-tumorigenic properties, in part due to offsetting the vicious cycle of osteolytic bone loss as well as regulating inflammatory signals. The impacts of metastatic cancer on the mechanosensory function of osteocytes remains unclear. Increased mechanical stimuli are a potential method for mitigating TIBD.
Collapse
Affiliation(s)
- Blayne A Sarazin
- Department of Mechanical Engineering, University of Colorado, 427 UCB, Boulder, CO, 80309, USA
| | - Claire L Ihle
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Philip Owens
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Veterans Affairs, Research Service, Eastern Colorado Health Care System, Aurora, CO, 80045, USA
| | - Maureen E Lynch
- Department of Mechanical Engineering, University of Colorado, 427 UCB, Boulder, CO, 80309, USA.
- Biofrontiers Institute, University of Colorado, Boulder, CO, 80309, USA.
| |
Collapse
|
26
|
Liu S, Sun X, Li K, Zha R, Feng Y, Sano T, Dong C, Liu Y, Aryal UK, Sudo A, Li BY, Yokota H. Generation of the tumor-suppressive secretome from tumor cells. Am J Cancer Res 2021; 11:8517-8534. [PMID: 34373756 PMCID: PMC8344019 DOI: 10.7150/thno.61006] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 07/09/2021] [Indexed: 12/13/2022] Open
Abstract
Rationale: The progression of cancer cells depends on the soil and building an inhibitory soil might be a therapeutic option. We previously created tumor-suppressive secretomes by activating Wnt signaling in MSCs. Here, we examined whether the anti-tumor secretomes can be produced from tumor cells. Methods: Wnt signaling was activated in tumor cells by overexpressing β-catenin or administering BML284, a Wnt activator. Their conditioned medium (CM) was applied to cancer cells or tissues, and the effects of CM were evaluated. Tumor growth in the mammary fat pad and tibia in C57BL/6 female mice was also evaluated through μCT imaging and histology. Whole-genome proteomics analysis was conducted to determine and characterize novel tumor-suppressing proteins, which were enriched in CM. Results: The overexpression of β-catenin or the administration of BML284 generated tumor-suppressive secretomes from breast, prostate and pancreatic cancer cells. In the mouse model, β-catenin-overexpressing CM reduced tumor growth and tumor-driven bone destruction. This inhibition was also observed with BML284-treated CM. Besides p53 and Trail, proteomics analysis revealed that CM was enriched with enolase 1 (Eno1) and ubiquitin C (Ubc) that presented notable tumor-suppressing actions. Importantly, Eno1 immunoprecipitated CD44, a cell-surface adhesion receptor, and its silencing suppressed Eno1-driven tumor inhibition. A pan-cancer survival analysis revealed that the downregulation of MMP9, Runx2 and Snail by CM had a significant impact on survival outcomes (p < 0.00001). CM presented a selective inhibition of tumor cells compared to non-tumor cells, and it downregulated PD-L1, an immune escape modulator. Conclusions: The tumor-suppressive secretome can be generated from tumor cells, in which β-catenin presented two opposing roles, as an intracellular tumor promoter in tumor cells and a generator of extracellular tumor suppressor in CM. Eno1 was enriched in CM and its interaction with CD44 was involved in Eno1's anti-tumor action. Besides presenting a potential option for treating primary cancers and metastases, the result indicates that aggressive tumors may inhibit the growth of less aggressive tumors via tumor-suppressive secretomes.
Collapse
|
27
|
Han S, Currier T, Edraki M, Liu B, Lynch ME, Modarres-Sadeghi Y. Flow inside a bone scaffold: Visualization using 3D phase contrast MRI and comparison with numerical simulations. J Biomech 2021; 126:110625. [PMID: 34293601 DOI: 10.1016/j.jbiomech.2021.110625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 10/20/2022]
Abstract
We report on results of experimental flow measurements inside a bone scaffold model, subjected to a uniform incoming flow (applied perfusion). Understanding the flow behavior inside a tissue engineered scaffold is essential for mechanistic studies of mechanobiology, particularly flow-sensitive bone cells. Nearly all existing studies that quantify interstitial flow inside engineered bone scaffolds have been based on numerical results, in part due to the difficulties associated with quantitative measurements and visualization of flow inside large, opaque bone or bone mimics. Thus, an experimental platform to complement and validate in silico studies is needed. Therefore, we developed a flow visualization method using Phase-Contrast Magnetic Resonance Imaging (PC-MRI) to measure flow velocities within a 3D-printed microCT-based rendering of a bone scaffold. We designed and built a non-magnetic recirculating water tunnel to apply uniform perfusion to the 3D-printed model and we measured flow distribution within the scaffold and compared these experimental results with CFD results. Both magnitude and distribution of flow velocities observed at different slices of the scaffold were in quantitative agreement numerically and experimentally. This experimental approach can be used to both validate numerical studies and provide insight into the flow behavior inside tissue-engineered scaffolds for a range of applications, including fundamental mechanobiology of healthy cells, and in the context of diseases, such as cancer.
Collapse
Affiliation(s)
- Suyue Han
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA, USA
| | - Todd Currier
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA, USA
| | - Mahdiar Edraki
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA, USA
| | - Boyuan Liu
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA, USA
| | - Maureen E Lynch
- Department of Mechanical Engineering, University of Boulder, CO, USA
| | - Yahya Modarres-Sadeghi
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
28
|
Adhikari M, Delgado-Calle J. Role of Osteocytes in Cancer Progression in the Bone and the Associated Skeletal Disease. Curr Osteoporos Rep 2021; 19:247-255. [PMID: 33818732 PMCID: PMC8486016 DOI: 10.1007/s11914-021-00679-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2021] [Indexed: 11/29/2022]
Abstract
PURPOSE OF REVIEW The goal of this manuscript is to review the current knowledge on the role of osteocytes in cancer in the bone, discuss the potential of osteocytes as a therapeutic target, and propose future research needed to understand the crosstalk between cancer cells and osteocytes in the tumor niche. RECENT FINDINGS Numerous studies have established that cancer cells manipulate osteocytes to facilitate invasion and tumor progression in bone. Moreover, cancer cells dysregulate osteocyte function to disrupt physiological bone remodeling, leading to the development of bone disease. Targeting osteocytes and their derived factors has proven to effectively interfere with the progression of cancer in the bone and the associated bone disease. Osteocytes communicate with cancer cells and are also part of the vicious cycle of cancer in the bone. Additional studies investigating the role of osteocytes on metastases to the bone and the development of drug resistance are needed.
Collapse
Affiliation(s)
- Manish Adhikari
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Jesús Delgado-Calle
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| |
Collapse
|
29
|
Mechanical tibial loading remotely suppresses brain tumors by dopamine-mediated downregulation of CCN4. Bone Res 2021; 9:26. [PMID: 34031366 PMCID: PMC8144433 DOI: 10.1038/s41413-021-00144-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 12/09/2020] [Accepted: 12/20/2020] [Indexed: 11/08/2022] Open
Abstract
Mechanical loading to the bone is known to be beneficial for bone homeostasis and for suppressing tumor-induced osteolysis in the loaded bone. However, whether loading to a weight-bearing hind limb can inhibit distant tumor growth in the brain is unknown. We examined the possibility of bone-to-brain mechanotransduction using a mouse model of a brain tumor by focusing on the response to Lrp5-mediated Wnt signaling and dopamine in tumor cells. The results revealed that loading the tibia with elevated levels of tyrosine hydroxylase, a rate-limiting enzyme in dopamine synthesis, markedly reduced the progression of the brain tumors. The simultaneous application of fluphenazine (FP), an antipsychotic dopamine modulator, enhanced tumor suppression. Dopamine and FP exerted antitumor effects through the dopamine receptors DRD1 and DRD2, respectively. Notably, dopamine downregulated Lrp5 via DRD1 in tumor cells. A cytokine array analysis revealed that the reduction in CCN4 was critical for loading-driven, dopamine-mediated tumor suppression. The silencing of Lrp5 reduced CCN4, and the administration of CCN4 elevated oncogenic genes such as MMP9, Runx2, and Snail. In summary, this study demonstrates that mechanical loading regulates dopaminergic signaling and remotely suppresses brain tumors by inhibiting the Lrp5-CCN4 axis via DRD1, indicating the possibility of developing an adjuvant bone-mediated loading therapy.
Collapse
|
30
|
Woollam M, Wang L, Grocki P, Liu S, Siegel AP, Kalra M, Goodpaster JV, Yokota H, Agarwal M. Tracking the Progression of Triple Negative Mammary Tumors over Time by Chemometric Analysis of Urinary Volatile Organic Compounds. Cancers (Basel) 2021; 13:1462. [PMID: 33806757 PMCID: PMC8004946 DOI: 10.3390/cancers13061462] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 01/06/2023] Open
Abstract
Previous studies have shown that volatile organic compounds (VOCs) are potential biomarkers of breast cancer. An unanswered question is how urinary VOCs change over time as tumors progress. To explore this, BALB/c mice were injected with 4T1.2 triple negative murine tumor cells in the tibia. This typically causes tumor progression and osteolysis in 1-2 weeks. Samples were collected prior to tumor injection and from days 2-19. Samples were analyzed by headspace solid phase microextraction coupled to gas chromatography-mass spectrometry. Univariate analysis identified VOCs that were biomarkers for breast cancer; some of these varied significantly over time and others did not. Principal component analysis was used to distinguish Cancer (all Weeks) from Control and Cancer Week 1 from Cancer Week 3 with over 90% accuracy. Forward feature selection and linear discriminant analysis identified a unique panel that could identify tumor presence with 94% accuracy and distinguish progression (Cancer Week 1 from Cancer Week 3) with 97% accuracy. Principal component regression analysis also demonstrated that a VOC panel could predict number of days since tumor injection (R2 = 0.71 and adjusted R2 = 0.63). VOC biomarkers identified by these analyses were associated with metabolic pathways relevant to breast cancer.
Collapse
Affiliation(s)
- Mark Woollam
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (M.W.); (P.G.); (A.P.S.); (J.V.G.)
- Integrated Nanosystems Development Institute, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (L.W.); (S.L.); (H.Y.)
| | - Luqi Wang
- Integrated Nanosystems Development Institute, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (L.W.); (S.L.); (H.Y.)
- Department of Biomedical Engineering, Indiana University-Purdue University, Indianapolis, IN 46202, USA
| | - Paul Grocki
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (M.W.); (P.G.); (A.P.S.); (J.V.G.)
- Integrated Nanosystems Development Institute, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (L.W.); (S.L.); (H.Y.)
| | - Shengzhi Liu
- Integrated Nanosystems Development Institute, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (L.W.); (S.L.); (H.Y.)
- Department of Biomedical Engineering, Indiana University-Purdue University, Indianapolis, IN 46202, USA
| | - Amanda P. Siegel
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (M.W.); (P.G.); (A.P.S.); (J.V.G.)
- Integrated Nanosystems Development Institute, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (L.W.); (S.L.); (H.Y.)
| | - Maitri Kalra
- Hematology and Oncology, Ball Memorial Hospital, Indiana University Health, Muncie, IN 47303, USA;
| | - John V. Goodpaster
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (M.W.); (P.G.); (A.P.S.); (J.V.G.)
| | - Hiroki Yokota
- Integrated Nanosystems Development Institute, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (L.W.); (S.L.); (H.Y.)
- Department of Biomedical Engineering, Indiana University-Purdue University, Indianapolis, IN 46202, USA
- Biomechanics and Biomaterials Research Center, Indiana University-Purdue University, Indianapolis, IN 46202, USA
| | - Mangilal Agarwal
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (M.W.); (P.G.); (A.P.S.); (J.V.G.)
- Integrated Nanosystems Development Institute, Indiana University-Purdue University, Indianapolis, IN 46202, USA; (L.W.); (S.L.); (H.Y.)
- Department of Mechanical & Energy Engineering, Indiana University-Purdue University, Indianapolis, IN 46202, USA
| |
Collapse
|
31
|
Sano T, Sun X, Feng Y, Liu S, Hase M, Fan Y, Zha R, Wu D, Aryal UK, Li BY, Sudo A, Yokota H. Inhibition of the Growth of Breast Cancer-Associated Brain Tumors by the Osteocyte-Derived Conditioned Medium. Cancers (Basel) 2021; 13:1061. [PMID: 33802279 PMCID: PMC7959137 DOI: 10.3390/cancers13051061] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/19/2021] [Accepted: 02/24/2021] [Indexed: 12/12/2022] Open
Abstract
The brain is a common site of metastasis from advanced breast cancer but few effective treatments are available. We examined a therapeutic option with a conditioned medium (CM), focusing on the role of Lrp5 and β-catenin in Wnt signaling, and IL1ra in osteocytes. Osteocytes presented the innate anti-tumor effect and the overexpression of the above genes strengthened their action. In a mouse model, the injection of their CM inhibited mammary tumors and tumor-driven osteolysis. Importantly, Lrp5- and/or IL1ra-overexpressing osteocytes or the local administration of β-catenin-overexpressing CM markedly inhibited brain tumors. In the transport analysis, tumor-suppressing factors in CM were shown to diffuse through the skull. Mechanistically, the CM with overexpression of the above genes downregulated oncogenic genes such as MMP9, Runx2, TGFβ, and Snail in breast cancer cells. Also, the CM with β-catenin overexpression downregulated CXCL1 and CXCL5 and upregulated tumor suppressors such as LIMA1, DSP, p53, and TRAIL in breast cancer cells. Notably, whole-genome proteomics revealed that histone H4 was enriched in CM and acted as an atypical tumor suppressor. Lrp5-overexpressing MSCs were also shown to act as anti-tumor agents. Collectively, this study demonstrated the therapeutic role of engineered CM in brain tumors and the tumor-suppressing action of extracellular histone H4. The result sheds light on the potential CM-based therapy for breast cancer-associated brain metastases in a minimally invasive manner.
Collapse
Affiliation(s)
- Tomohiko Sano
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA; (T.S.); (X.S.); (Y.F.); (S.L.); (M.H.); (Y.F.); (R.Z.); (D.W.)
- Department of Orthopedic Surgery, Mie University Graduate School of Medicine, Edobashi Tsu 2-174, Japan;
| | - Xun Sun
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA; (T.S.); (X.S.); (Y.F.); (S.L.); (M.H.); (Y.F.); (R.Z.); (D.W.)
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China;
| | - Yan Feng
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA; (T.S.); (X.S.); (Y.F.); (S.L.); (M.H.); (Y.F.); (R.Z.); (D.W.)
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China;
| | - Shengzhi Liu
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA; (T.S.); (X.S.); (Y.F.); (S.L.); (M.H.); (Y.F.); (R.Z.); (D.W.)
| | - Misato Hase
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA; (T.S.); (X.S.); (Y.F.); (S.L.); (M.H.); (Y.F.); (R.Z.); (D.W.)
- Graduate School of Engineering, Mie University, Edobashi Tsu 2-174, Japan
| | - Yao Fan
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA; (T.S.); (X.S.); (Y.F.); (S.L.); (M.H.); (Y.F.); (R.Z.); (D.W.)
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China;
| | - Rongrong Zha
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA; (T.S.); (X.S.); (Y.F.); (S.L.); (M.H.); (Y.F.); (R.Z.); (D.W.)
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China;
| | - Di Wu
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA; (T.S.); (X.S.); (Y.F.); (S.L.); (M.H.); (Y.F.); (R.Z.); (D.W.)
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China;
| | - Uma K. Aryal
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA;
| | - Bai-Yan Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China;
| | - Akihiro Sudo
- Department of Orthopedic Surgery, Mie University Graduate School of Medicine, Edobashi Tsu 2-174, Japan;
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA; (T.S.); (X.S.); (Y.F.); (S.L.); (M.H.); (Y.F.); (R.Z.); (D.W.)
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China;
- Simon Cancer Research Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
32
|
Riehl BD, Kim E, Bouzid T, Lim JY. The Role of Microenvironmental Cues and Mechanical Loading Milieus in Breast Cancer Cell Progression and Metastasis. Front Bioeng Biotechnol 2021; 8:608526. [PMID: 33585411 PMCID: PMC7874074 DOI: 10.3389/fbioe.2020.608526] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/22/2020] [Indexed: 01/08/2023] Open
Abstract
Cancer can disrupt the microenvironments and mechanical homeostatic actions in multiple scales from large tissue modification to altered cellular signaling pathway in mechanotransduction. In this review, we highlight recent progresses in breast cancer cell mechanobiology focusing on cell-microenvironment interaction and mechanical loading regulation of cells. First, the effects of microenvironmental cues on breast cancer cell progression and metastasis will be reviewed with respect to substrate stiffness, chemical/topographic substrate patterning, and 2D vs. 3D cultures. Then, the role of mechanical loading situations such as tensile stretch, compression, and flow-induced shear will be discussed in relation to breast cancer cell mechanobiology and metastasis prevention. Ultimately, the substrate microenvironment and mechanical signal will work together to control cancer cell progression and metastasis. The discussions on breast cancer cell responsiveness to mechanical signals, from static substrate and dynamic loading, and the mechanotransduction pathways involved will facilitate interdisciplinary knowledge transfer, enabling further insights into prognostic markers, mechanically mediated metastasis pathways for therapeutic targets, and model systems required to advance cancer mechanobiology.
Collapse
Affiliation(s)
- Brandon D Riehl
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Eunju Kim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Tasneem Bouzid
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Jung Yul Lim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States.,Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
33
|
Conceição F, Sousa DM, Paredes J, Lamghari M. Sympathetic activity in breast cancer and metastasis: partners in crime. Bone Res 2021; 9:9. [PMID: 33547275 PMCID: PMC7864971 DOI: 10.1038/s41413-021-00137-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 11/16/2020] [Accepted: 11/20/2020] [Indexed: 01/30/2023] Open
Abstract
The vast majority of patients with advanced breast cancer present skeletal complications that severely compromise their quality of life. Breast cancer cells are characterized by a strong tropism to the bone niche. After engraftment and colonization of bone, breast cancer cells interact with native bone cells to hinder the normal bone remodeling process and establish an osteolytic "metastatic vicious cycle". The sympathetic nervous system has emerged in recent years as an important modulator of breast cancer progression and metastasis, potentiating and accelerating the onset of the vicious cycle and leading to extensive bone degradation. Furthermore, sympathetic neurotransmitters and their cognate receptors have been shown to promote several hallmarks of breast cancer, such as proliferation, angiogenesis, immune escape, and invasion of the extracellular matrix. In this review, we assembled the current knowledge concerning the complex interactions that take place in the tumor microenvironment, with a special emphasis on sympathetic modulation of breast cancer cells and stromal cells. Notably, the differential action of epinephrine and norepinephrine, through either α- or β-adrenergic receptors, on breast cancer progression prompts careful consideration when designing new therapeutic options. In addition, the contribution of sympathetic innervation to the formation of bone metastatic foci is highlighted. In particular, we address the remarkable ability of adrenergic signaling to condition the native bone remodeling process and modulate the bone vasculature, driving breast cancer cell engraftment in the bone niche. Finally, clinical perspectives and developments on the use of β-adrenergic receptor inhibitors for breast cancer management and treatment are discussed.
Collapse
Affiliation(s)
- Francisco Conceição
- grid.5808.50000 0001 1503 7226I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal ,grid.5808.50000 0001 1503 7226INEB—Instituto Nacional de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal ,grid.5808.50000 0001 1503 7226ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Daniela M. Sousa
- grid.5808.50000 0001 1503 7226I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal ,grid.5808.50000 0001 1503 7226INEB—Instituto Nacional de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Joana Paredes
- grid.5808.50000 0001 1503 7226I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal ,grid.5808.50000 0001 1503 7226IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, 4200-135 Porto, Portugal ,grid.5808.50000 0001 1503 7226FMUP—Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal
| | - Meriem Lamghari
- grid.5808.50000 0001 1503 7226I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal ,grid.5808.50000 0001 1503 7226INEB—Instituto Nacional de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal ,grid.5808.50000 0001 1503 7226ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| |
Collapse
|
34
|
Liu B, Han S, Modarres-Sadeghi Y, Lynch ME. Multiphysics simulation of a compression-perfusion combined bioreactor to predict the mechanical microenvironment during bone metastatic breast cancer loading experiments. Biotechnol Bioeng 2021; 118:1779-1792. [PMID: 33491767 DOI: 10.1002/bit.27692] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 01/15/2021] [Accepted: 01/22/2021] [Indexed: 01/12/2023]
Abstract
Incurable breast cancer bone metastasis causes widespread bone loss, resulting in fragility, pain, increased fracture risk, and ultimately increased patient mortality. Increased mechanical signals in the skeleton are anabolic and protect against bone loss, and they may also do so during osteolytic bone metastasis. Skeletal mechanical signals include interdependent tissue deformations and interstitial fluid flow, but how metastatic tumor cells respond to each of these individual signals remains underinvestigated, a barrier to translation to the clinic. To delineate their respective roles, we report computed estimates of the internal mechanical field of a bone mimetic scaffold undergoing combinations of high and low compression and perfusion using multiphysics simulations. Simulations were conducted in advance of multimodal loading bioreactor experiments with bone metastatic breast cancer cells to ensure that mechanical stimuli occurring internally were physiological and anabolic. Our results show that mechanical stimuli throughout the scaffold were within the anabolic range of bone cells in all loading configurations, were homogenously distributed throughout, and that combined high magnitude compression and perfusion synergized to produce the largest wall shear stresses within the scaffold. These simulations, when combined with experiments, will shed light on how increased mechanical loading in the skeleton may confer anti-tumorigenic effects during metastasis.
Collapse
Affiliation(s)
- Boyuan Liu
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, Massachusetts, USA
| | - Suyue Han
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, Massachusetts, USA
| | - Yahya Modarres-Sadeghi
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, Massachusetts, USA
| | - Maureen E Lynch
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, Massachusetts, USA.,Department of Mechanical Engineering, University of Colorado, Boulder, Colorado, USA
| |
Collapse
|
35
|
Feng Y, Liu S, Zha R, Sun X, Li K, Robling A, Li B, Yokota H. Mechanical Loading-Driven Tumor Suppression Is Mediated by Lrp5-Dependent and Independent Mechanisms. Cancers (Basel) 2021; 13:cancers13020267. [PMID: 33450808 PMCID: PMC7828232 DOI: 10.3390/cancers13020267] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Advanced breast cancer and prostate cancer metastasize to varying organs including the bone. We show here that mechanical loading to the knee suppresses tumor growth in the loaded bone and the non-loaded mammary pad. Although lipoprotein receptor-related protein 5 (Lrp5) in osteocytes is necessary to induce loading-driven bone formation, loading-driven tumor suppression is regulated by Lrp5-dependent and independent mechanisms. Lrp5 overexpression in osteocytes enhances tumor suppression, but without Lrp5 in osteocytes, mechanical loading elevates dopamine, chemerin, p53, and TNF-related apoptosis-inducing ligand (TRAIL) and reduces cholesterol and nexin. Their systemic changes contribute to inhibiting tumors without Lrp5. Osteoclast development is also inhibited by the load-driven regulation of chemerin and nexin. Abstract Bone is mechanosensitive and lipoprotein receptor-related protein 5 (Lrp5)-mediated Wnt signaling promotes loading-driven bone formation. While mechanical loading can suppress tumor growth, the question is whether Lrp5 mediates loading-driven tumor suppression. Herein, we examined the effect of Lrp5 using osteocyte-specific Lrp5 conditional knockout mice. All mice presented noticeable loading-driven tumor suppression in the loaded tibia and non-loaded mammary pad. The degree of suppression was more significant in wild-type than knockout mice. In all male and female mice, knee loading reduced cholesterol and elevated dopamine. It reduced tumor-promoting nexin, which was elevated by cholesterol and reduced by dopamine. By contrast, it elevated p53, TNF-related apoptosis-inducing ligand (TRAIL), and chemerin, and they were regulated reversely by dopamine and cholesterol. Notably, Lrp5 overexpression in osteocytes enhanced tumor suppression, and osteoclast development was inhibited by chemerin. Collectively, this study identified Lrp5-dependent and independent mechanisms for tumor suppression. Lrp5 in osteocytes contributed to the loaded bone, while the Lrp5-independent regulation of dopamine- and cholesterol-induced systemic suppression.
Collapse
Affiliation(s)
- Yan Feng
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China; (Y.F.); (R.Z.); (X.S.); (K.L.)
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA;
| | - Shengzhi Liu
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA;
| | - Rongrong Zha
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China; (Y.F.); (R.Z.); (X.S.); (K.L.)
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA;
| | - Xun Sun
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China; (Y.F.); (R.Z.); (X.S.); (K.L.)
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA;
| | - Kexin Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China; (Y.F.); (R.Z.); (X.S.); (K.L.)
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA;
| | - Alexander Robling
- Department of Anatomy Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Baiyan Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China; (Y.F.); (R.Z.); (X.S.); (K.L.)
- Correspondence: (B.L.); (H.Y.); Tel.: +86-451-8667-1354 (B.L.); +317-278-5177 (H.Y.); Fax: +86-451-8667-1354 (B.L.); +317-278-2455 (H.Y.)
| | - Hiroki Yokota
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China; (Y.F.); (R.Z.); (X.S.); (K.L.)
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA;
- Department of Anatomy Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Correspondence: (B.L.); (H.Y.); Tel.: +86-451-8667-1354 (B.L.); +317-278-5177 (H.Y.); Fax: +86-451-8667-1354 (B.L.); +317-278-2455 (H.Y.)
| |
Collapse
|
36
|
Rummler M, Ziouti F, Bouchard AL, Brandl A, Duda GN, Bogen B, Beilhack A, Lynch ME, Jundt F, Willie BM. Mechanical loading prevents bone destruction and exerts anti-tumor effects in the MOPC315.BM.Luc model of myeloma bone disease. Acta Biomater 2021; 119:247-258. [PMID: 33130307 DOI: 10.1016/j.actbio.2020.10.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/05/2020] [Accepted: 10/27/2020] [Indexed: 02/08/2023]
Abstract
Bone continually adapts to changing external loading conditions via (re)modeling (modeling and remodeling) processes. While physical activity is known to beneficially enhance bone mass in healthy individuals, little is known in how physical stimuli affect osteolytic bone destruction in patients suffering from multiple myeloma bone disease. Multiple myeloma (MM) is caused by malignant plasma cells in the bone marrow, shifting the balance in bone remodeling towards massive resorption. We hypothesized that in vivo tibial mechanical loading has anabolic effects in mice with locally injected MOPC315.BM.Luc cells. Conventional microCT analysis revealed enhanced cortical bone mass and microstructure in loaded compared to nonloaded mice. State-of-the-art time-lapse microCT based image analysis demonstrated bone (re)modeling processes at the endosteal and periosteal surfaces as the underlying causes of increased bone mass. Loading prevented the progression and development of osteolytic destruction. Physical stimuli also diminished local MM cell growth and dissemination evidenced by quantification of MM cell-specific immunoglobulin A levels in the serum of mice and by bioluminescence analysis. These data indicate that mechanical loading not only rescues the bone phenotype, but also exerts cell-extrinsic anti-myeloma effects in the MOPC315.BM.Luc model. In conclusion, the use of physical stimuli should be further investigated as an anabolic treatment for osteolytic bone destruction in patients with MM.
Collapse
|
37
|
Bone, a Secondary Growth Site of Breast and Prostate Carcinomas: Role of Osteocytes. Cancers (Basel) 2020; 12:cancers12071812. [PMID: 32640686 PMCID: PMC7408809 DOI: 10.3390/cancers12071812] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023] Open
Abstract
Bone is the primarily preferred site for breast and prostate cancer to metastasize. Bone metastases are responsible for most deaths related to breast and prostate cancer. The bone's particular microenvironment makes it conducive for the growth of cancer cells. Studies on bone metastasis have focused on the interaction between cancer cells and the bone microenvironment. Osteocytes, the most common cell type of bone tissue, have received little attention in bone metastasis, although they are master signal sensors, integrators, and skeleton transducers. They play an important role in regulating bone mass by acting on both osteoblasts and osteoclasts, through the release of proteins such as sclerostin, Dickkopf-1 (DKK-1), and fibroblast growth factor 23 (FGF23). Osteocytes have been extensively re-evaluated, in light of their multiple functions: with different experimental approaches, it has been shown that, indeed, osteocytes are actively involved in the colonization of bone tissue by cancer cells. The present review focuses on recent research on the role that osteocytes play in bone metastasis of breast and prostate cancers. Moreover, the studies here summarized open up perspectives for new therapeutic approaches focused on modulating the activity of osteocytes to improve the condition of the bone metastatic patients. A better understanding of the complex interactions between cancer cells and bone-resident cells is indispensable for identifying potential therapeutic targets to stop tumor progression and prevent bone metastases.
Collapse
|
38
|
Wu D, Fan Y, Liu S, Woollam MD, Sun X, Murao E, Zha R, Prakash R, Park C, Siegel AP, Liu J, Agarwal M, Li BY, Yokota H. Loading-induced antitumor capability of murine and human urine. FASEB J 2020; 34:7578-7592. [PMID: 32293076 DOI: 10.1096/fj.202000096r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/09/2020] [Accepted: 03/23/2020] [Indexed: 12/15/2022]
Abstract
While urine has been considered as a useful bio-fluid for health monitoring, its dynamic changes to physical activity are not well understood. We examined urine's possible antitumor capability in response to medium-level, loading-driven physical activity. Urine was collected from mice subjected to 5-minute skeletal loading and human individuals before and after 30-minute step aerobics. Six cancer cell lines (breast, prostate, and pancreas) and a mouse model of the mammary tumor were employed to evaluate the effect of urine. Compared to urine collected prior to loading, urine collected post-activity decreased the cellular viability, proliferation, migration, and invasion of tumor cells, as well as tumor weight in the mammary fat pad. Detection of urinary volatile organic compounds and ELISA assays showed that the loading-conditioned urine reduced cholesterol and elevated dopamine and melatonin. Immunohistochemical fluorescent images presented upregulation of the rate-limiting enzymes for the production of dopamine and melatonin in the brain. Molecular analysis revealed that the antitumor effect was linked to the reduction in molecular vinculin-linked molecular force as well as the downregulation of the Lrp5-CSF1-CD105 regulatory axis. Notably, the survival rate for the high expression levels of Lrp5, CSF1, and CD105 in tumor tissues was significantly lowered in the Cancer Genome Atlas database. Collectively, this study revealed that 5- or 10-minute loading-driven physical activity was sufficient to induce the striking antitumor effect by activating the neuronal signaling and repressing cholesterol synthesis. The result supported the dual role of loading-conditioned urine as a potential tumor suppressor and a source of diagnostic biomarkers.
Collapse
Affiliation(s)
- Di Wu
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin, China.,Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Yao Fan
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin, China.,Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Shengzhi Liu
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Mark D Woollam
- Department of Chemistry and Chemical Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA.,Integrative Nanosystems Development Institute, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Xun Sun
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin, China.,Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Eiji Murao
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA.,Graduate School of Engineering, Mie University, Mie, Japan
| | - Rongrong Zha
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin, China.,Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Rahul Prakash
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Charles Park
- Department of Physics, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Amanda P Siegel
- Department of Chemistry and Chemical Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA.,Integrative Nanosystems Development Institute, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Jing Liu
- Department of Physics, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA.,Simon Cancer Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mangilal Agarwal
- Integrative Nanosystems Development Institute, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Bai-Yan Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin, China
| | - Hiroki Yokota
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin, China.,Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA.,Integrative Nanosystems Development Institute, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA.,Simon Cancer Research Center, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
39
|
Abstract
Bone is the most frequent site of breast cancer and prostate cancer metastasis, and one of the most common sites of metastasis for many solid tumors. Once cancer cells colonize in the bone, it imposes a major clinical challenge for the treatment of the disease, and fatality rates increase drastically. Bone, the largest organ in the body, provides a fertile microenvironment enriched with nutrients, growth factors and hormones, a generous reward for cancer cells. Dependent on cancer type, cancer cells can cause osteoblastic (bone forming) or osteolytic lesions to promote the net resorption and/or release of growth factors from the bone extracellular matrix. These processes activate a "vicious cycle", leading to disruption of bone integrity and promoting cancer cell growth and migration. Cancer cells influence the bone microenvironment favoring their colonization and growth. In order to metastasize to the bone, cancer cells must first migrate from the site of origin, and once established within the bone, they must overcome the dormant inducing effects of resident cells. If successful, cancer cells can then colonize and continually disrupt bone homeostasis that is primarily maintained by osteocytes, the most abundant bone cell type. For example, it has been shown that exercise induces osteocytes to release anabolic factors that inhibit osteoclast resorptive activity, promote dormancy and the release of anti-cancer factors that inhibit breast cancer cell metastasis. In this review, we will summarize recent research findings and provide mechanistic insights related to the role of osteocytes in osteolytic metastasis.
Collapse
|