1
|
Anloague A, Sabol HM, Kaur J, Khan S, Ashby C, Schinke C, Barnes CL, Alturkmani F, Ambrogini E, Gundesen MT, Lund T, Amstrup AK, Andersen TL, Diaz-delCastillo M, Roodman GD, Bellido T, Delgado-Calle J. A novel CCL3-HMGB1 signaling axis regulating osteocyte RANKL expression in multiple myeloma. Haematologica 2025; 110:952-966. [PMID: 39605211 PMCID: PMC11959238 DOI: 10.3324/haematol.2024.286484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Multiple myeloma (MM) is a clonal plasma cell proliferative malignancy characterized by a debilitating bone disease. Osteolytic destruction, a hallmark of MM, is driven by increased osteoclast number and exacerbated bone resorption, primarily fueled by the excessive production of RANKL, the master regulator of osteoclast formation, within the tumor niche. We previously reported that osteocytes, the most abundant cells in the bone niche, promote tumor progression and support MM bone disease by overproducing RANKL. However, the molecular mechanisms underlying RANKL dysregulation in osteocytes in the context of MM bone disease are not entirely understood. Here, we present evidence that MM-derived CCL3 induces upregulation of RANKL expression in both human and murine osteocytes. Through a combination of in vitro, ex vivo, and in vivo models and clinical data, we demonstrate that genetic or pharmacologic inhibition of CCL3 prevents RANKL upregulation in osteocytes and attenuates the bone loss induced by MM cells. Mechanistic studies revealed that MM-derived CCL3 triggers the secretion of HMGB1 by osteocytes, a process required for osteocytic RANKL upregulation by MM cells. These findings identify a previously unknown CCL3-HMGB1 signaling axis in the MM tumor niche that drives bone resorption by promoting RANKL overproduction in osteocytes.
Collapse
Affiliation(s)
- Aric Anloague
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Hayley M Sabol
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Japneet Kaur
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US; Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock
| | - Sharmin Khan
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Cody Ashby
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, US; Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Carolina Schinke
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, US; Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR
| | - C Lowry Barnes
- Department of Orthopedic Surgery; University of Arkansas for Medical Sciences, Little Rock, AR
| | - Farah Alturkmani
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US; Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences Little Rock, AR
| | - Elena Ambrogini
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US; Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences Little Rock, AR, US; Central Arkansas Veterans Healthcare System, Little Rock, AR
| | - Michael Tveden Gundesen
- Department of Hematology, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Thomas Lund
- Department of Hematology, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Centre for Innovative Medical Technology, Odense University Hospital, Odense, Denmark
| | - Anne Kristine Amstrup
- Department of Endocrinology and Internal Medicine (MEA), THG, Aarhus University Hospital, Aarhus, Denmark
| | - Thomas Levin Andersen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Pathology, Odense University Hospital, Odense, Denmark; Department of Forensic Medicine, University of Aarhus, Aarhus, Denmark
| | | | - G David Roodman
- Division of Hematology and Oncology, Department of Medicine, Indiana University, Indianapolis, IN
| | - Teresita Bellido
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US; Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, US; Department of Orthopedic Surgery; University of Arkansas for Medical Sciences, Little Rock, AR, US; Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences Little Rock, AR, US; Central Arkansas Veterans Healthcare System, Little Rock, AR
| | - Jesus Delgado-Calle
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US; Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, US; Department of Orthopedic Surgery; University of Arkansas for Medical Sciences, Little Rock, AR, US; Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences Little Rock, AR.
| |
Collapse
|
2
|
Bishop RT, Delgado-Calle J, Reagan MR, Marino S. 2D and 3D In Vitro Co-culture for Cancer and Bone Cell Interaction Studies. Methods Mol Biol 2025; 2885:113-141. [PMID: 40448759 DOI: 10.1007/978-1-0716-4306-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2025]
Abstract
Co-culture assays are used to study the mutual interaction between cells in vitro. This chapter describes 2D and 3D co-culture systems used to study cell-cell signaling crosstalk between cancer cells and bone marrow adipocytes, osteoblasts, osteoclasts, and osteocytes. The chapter provides a step-by-step guide to the most used cell culture techniques, functional assays, and gene expression.
Collapse
Affiliation(s)
- Ryan T Bishop
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Research Center and Institute, Tampa, FL, USA
| | - Jesus Delgado-Calle
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Winthrop P. Rockefeller Cancer Institute, Little Rock, AR, USA
| | - Michaela R Reagan
- Center for Molecular Medicine, Maine Medical Centre Research Institute, Scarborough, ME, USA
| | - Silvia Marino
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
3
|
Kaur J, Delgado-Calle J. Ex Vivo Model Systems of Cancer-Bone Cell Interactions. Methods Mol Biol 2025; 2885:291-302. [PMID: 40448766 DOI: 10.1007/978-1-0716-4306-8_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2025]
Abstract
Ex vivo bone organ cultures preserve cell-cell and cell-matrix interactions and allow the study of cancer and bone cells in their natural tumor environment. This chapter is a comprehensive guide detailing the methods employed to establish and characterize ex vivo bone-cancer organ cultures. The chapter begins by outlining the materials and step-by-step procedures for successfully initiating and maintaining these cultures, emphasizing the advantage of modeling cancer within the bone microenvironment. Additionally, the chapter provides a detailed account of the techniques utilized to analyze key endpoints, such as tumor growth, microenvironmental changes, and therapeutic responses.
Collapse
Affiliation(s)
- Japneet Kaur
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jesus Delgado-Calle
- Department of Physiology and Cell Biology, The Winthrop P Rockefeller Cancer Institute, Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
4
|
Kaur J, Adhikari M, Sabol HM, Anloague A, Khan S, Kurihara N, Diaz-delCastillo M, Andreasen CM, Barnes CL, Stambough JB, Palmieri M, Reyes-Castro O, Zarrer J, Taipaleenmäki H, Ambrogini E, Almeida M, O’Brien CA, Nookaw I, Delgado-Calle J. Single-Cell Transcriptomic Analysis Identifies Senescent Osteocytes That Trigger Bone Destruction in Breast Cancer Metastasis. Cancer Res 2024; 84:3936-3952. [PMID: 39312185 PMCID: PMC11611663 DOI: 10.1158/0008-5472.can-24-0857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/22/2024] [Accepted: 09/19/2024] [Indexed: 09/25/2024]
Abstract
Breast cancer bone metastases increase fracture risk and are a major cause of morbidity and mortality among women. Upon colonization by tumor cells, the bone microenvironment undergoes profound reprogramming to support cancer progression, which disrupts the balance between osteoclasts and osteoblasts and leads to bone lesions. A deeper understanding of the processes mediating this reprogramming could help develop interventions for treating patients with bone metastases. Here, we demonstrated that osteocytes (Ot) in established breast cancer bone metastasis develop premature senescence and a distinctive senescence-associated secretory phenotype (SASP) that favors bone destruction. Single-cell RNA sequencing identified Ots from mice with breast cancer bone metastasis enriched in senescence, SASP markers, and pro-osteoclastogenic genes. Multiplex in situ hybridization and artificial intelligence-assisted analysis depicted Ots with senescence-associated satellite distension, telomere dysfunction, and p16Ink4a expression in mice and patients with breast cancer bone metastasis. Breast cancer cells promoted Ot senescence and enhanced their osteoclastogenic potential in in vitro and ex vivo organ cultures. Clearance of senescent cells with senolytics suppressed bone resorption and preserved bone mass in mice with breast cancer bone metastasis. These results demonstrate that Ots undergo pathological reprogramming by breast cancer cells and identify Ot senescence as an initiating event triggering lytic bone disease in breast cancer metastases. Significance: Breast cancer cells remodel the bone microenvironment by promoting premature cellular senescence and SASP in osteocytes, which can be targeted with senolytics to alleviate bone loss induced by metastatic breast cancer. See related commentary by Frieling and Lynch, p. 3917.
Collapse
Affiliation(s)
- Japneet Kaur
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Manish Adhikari
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Hayley M. Sabol
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Aric Anloague
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Sharmin Khan
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Noriyoshi Kurihara
- Division of Hematology and Oncology, Department of Medicine, Indiana University, Indianapolis, IN, US
| | | | - Christina Møller Andreasen
- Molecular Bone Histology lab, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Clinical Pathology, Odense University Hospital, Odense University Hospital, Odense, Denmark
| | - C. Lowry Barnes
- Department of Orthopedic Surgery; University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Jeffrey B. Stambough
- Department of Orthopedic Surgery; University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Michela Palmieri
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US
| | - Olivia Reyes-Castro
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US
| | - Jennifer Zarrer
- Institute of Musculoskeletal Medicine, Musculoskeletal University Center Munich, University Hospital, LMU Munich, Germany
| | - Hanna Taipaleenmäki
- Institute of Musculoskeletal Medicine, Musculoskeletal University Center Munich, University Hospital, LMU Munich, Germany
| | - Elena Ambrogini
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US
| | - Maria Almeida
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US
| | - Charles A. O’Brien
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Intawat Nookaw
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, US
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Jesus Delgado-Calle
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US
- Department of Orthopedic Surgery; University of Arkansas for Medical Sciences, Little Rock, AR, US
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, US
| |
Collapse
|
5
|
Roosma J. A comprehensive review of oncogenic Notch signaling in multiple myeloma. PeerJ 2024; 12:e18485. [PMID: 39619207 PMCID: PMC11608568 DOI: 10.7717/peerj.18485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 10/16/2024] [Indexed: 12/13/2024] Open
Abstract
Multiple myeloma remains an incurable plasma cell cancer with radical case-by-case heterogeneity. Because of this, personalized and disease-specific biology of multiple myeloma must be understood for the discovery of effective molecular targets. The highly evolutionarily conserved Notch signaling pathway has been extensively described as a multifaceted driver of the multiple myeloma disease process-contributing to both intrinsic effects of malignant cells and to widespread remodeling of the tumor microenvironment that further facilitates disease progression. Namely, Notch signaling amongst malignant cells promotes increased proliferation, tumor-initiating capacity, drug resistance, and invasiveness. Moreover, Notch signaling between malignant cells and cells of the tumor microenvironment leads to increased osteodegenerative disease and angiogenesis. This comprehensive review will discuss both the intrinsic implications of pathological Notch signaling in multiple myeloma and the extrinsic implications of Notch signaling in the multiple myeloma tumor microenvironment. Additionally, the genetic origins of Notch signaling dysregulation in multiple myeloma and current attempts at targeting Notch therapeutically will be reviewed. While the subject has been reviewed previously, recent developments in the intervening years demand a revised synthesis of the literature. The aim of this work is to introduce and thoroughly synthesize the current state of knowledge in this vein of research and to highlight future directions for both new and in-the-field scientists.
Collapse
Affiliation(s)
- Justin Roosma
- Biology, Eastern Washington University, Cheney, Washington, United States
| |
Collapse
|
6
|
Sabol HM, Ashby C, Adhikari M, Anloague A, Kaur J, Khan S, Choudhury SR, Schinke C, Palmieri M, Barnes CL, Ambrogini E, Nookaew I, Delgado-Calle J. A NOTCH3-CXCL12-driven myeloma-tumor niche signaling axis promotes chemoresistance in multiple myeloma. Haematologica 2024; 109:2606-2618. [PMID: 38385272 PMCID: PMC11290536 DOI: 10.3324/haematol.2023.284443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 02/13/2024] [Indexed: 02/23/2024] Open
Abstract
Multiple myeloma (MM) remains incurable due to disease relapse and drug resistance. Notch signals from the tumor microenvironment (TME) confer chemoresistance, but the cellular and molecular mechanisms are not entirely understood. Using clinical and transcriptomic datasets, we found that NOTCH3 is upregulated in CD138+ cells from newly diagnosed MM (NDMM) patients compared to healthy individuals and increased in progression/relapsed MM (PRMM) patients. Further, NDMM patients with high NOTCH3 expression exhibited worse responses to bortezomib (BOR)-based therapies. Cells of the TME, including osteocytes, upregulated NOTCH3 in MM cells and protected them from apoptosis induced by BOR. NOTCH3 activation (NOTCH3OE) in MM cells decreased BOR anti-MM efficacy and its ability to improve survival in in vivo myeloma models. Molecular analyses revealed that NDMM and PRMM patients with high NOTCH3 exhibit CXCL12 upregulation. TME cells upregulated CXCL12 and activated the CXCR4 pathway in MM cells in a NOTCH3-dependent manner. Moreover, genetic or pharmacologic inhibition of CXCL12 in NOTCH3OE MM cells restored sensitivity to BOR regimes in vitro and in human bones bearing NOTCH3OE MM tumors cultured ex vivo. Our clinical and preclinical data unravel a novel NOTCH3-CXCL12 pro-survival signaling axis in the TME and suggest that osteocytes transmit chemoresistance signals to MM cells.
Collapse
Affiliation(s)
- Hayley M. Sabol
- Physiology and Cell Biology, University of Arkansas for Medical Sciences
| | - Cody Ashby
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences
| | - Manish Adhikari
- Physiology and Cell Biology, University of Arkansas for Medical Sciences
| | - Aric Anloague
- Physiology and Cell Biology, University of Arkansas for Medical Sciences
| | - Japneet Kaur
- Physiology and Cell Biology, University of Arkansas for Medical Sciences
| | - Sharmin Khan
- Physiology and Cell Biology, University of Arkansas for Medical Sciences
| | - Samrat Roy Choudhury
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences
- Pediatric Hematology-Oncology, Arkansas Children’s Research Institute, University of Arkansas for Medical Sciences
| | - Carolina Schinke
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences
- Myeloma Center, University of Arkansas for Medical Sciences
| | - Michela Palmieri
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System
| | - C. Lowry Barnes
- Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Elena Ambrogini
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases and Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System
| | - Intawat Nookaew
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences
| | - Jesus Delgado-Calle
- Physiology and Cell Biology, University of Arkansas for Medical Sciences
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences
| |
Collapse
|
7
|
Zerlotin R, Oranger A, Pignataro P, Dicarlo M, Sanesi L, Suriano C, Storlino G, Rizzi R, Mestice A, Di Gioia S, Mori G, Grano M, Colaianni G, Colucci S. Irisin prevents trabecular bone damage and tumor invasion in a mouse model of multiple myeloma. JBMR Plus 2024; 8:ziae066. [PMID: 38855797 PMCID: PMC11162589 DOI: 10.1093/jbmrpl/ziae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/29/2024] [Indexed: 06/11/2024] Open
Abstract
Bone disease associated with multiple myeloma (MM) is characterized by osteolytic lesions and pathological fractures, which remain a therapeutic priority despite new drugs improving MM patient survival. Antiresorptive molecules represent the main option for the treatment of MM-associated bone disease (MMBD), whereas osteoanabolic molecules are under investigation. Among these latter, we here focused on the myokine irisin, which is able to enhance bone mass in healthy mice, prevent bone loss in osteoporotic mouse models, and accelerate fracture healing in mice. Therefore, we investigated irisin effect on MMBD in a mouse model of MM induced by intratibial injection of myeloma cells followed by weekly administration of 100 μg/kg of recombinant irisin for 5 wk. By micro-Ct analysis, we demonstrated that irisin improves MM-induced trabecular bone damage by partially preventing the reduction of femur Trabecular Bone Volume/Total Volume (P = .0028), Trabecular Number (P = .0076), Trabecular Fractal Dimension (P = .0044), and increasing Trabecular Separation (P = .0003) in MM mice. In cortical bone, irisin downregulates the expression of Sclerostin, a bone formation inhibitor, and RankL, a pro-osteoclastogenic molecule, while in BM it upregulates Opg, an anti-osteoclastogenic cytokine. We found that in the BM tibia of irisin-treated MM mice, the percentage of MM cells displays a reduction trend, while in the femur it decreases significantly. This is in line with the in vitro reduction of myeloma cell viability after 48 h of irisin stimulation at both 200 and 500 ng/mL and, after 72 h already at 100 ng/mL rec-irisin. These results could be due to irisin ability to downregulate the expression of Notch 3, which is important for cell-to-cell communication in the tumor niche, and Cyclin D1, supporting an inhibitory effect of irisin on MM cell proliferation. Overall, our findings suggest that irisin could be a new promising strategy to counteract MMBD and tumor burden in one shot.
Collapse
Affiliation(s)
- Roberta Zerlotin
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Angela Oranger
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Patrizia Pignataro
- Department of Translational Biomedicine and Neuroscience, University of Bari, 70124 Bari, Italy
| | - Manuela Dicarlo
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Lorenzo Sanesi
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Clelia Suriano
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Giuseppina Storlino
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Rita Rizzi
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Anna Mestice
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Sante Di Gioia
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Giorgio Mori
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Maria Grano
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Graziana Colaianni
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Silvia Colucci
- Department of Translational Biomedicine and Neuroscience, University of Bari, 70124 Bari, Italy
| |
Collapse
|
8
|
Adhikari M, Kaur J, Sabol HM, Anloague A, Khan S, Kurihara N, Diaz-delCastillo M, Andreasen CM, Barnes CL, Stambough JB, Palmieri M, Reyes-Castro O, Ambrogini E, Almeida M, O’Brien CA, Nookaw I, Delgado-Calle J. Single-cell Transcriptome Analysis Identifies Senescent Osteocytes as Contributors to Bone Destruction in Breast Cancer Metastasis. RESEARCH SQUARE 2024:rs.3.rs-4047486. [PMID: 38558984 PMCID: PMC10980159 DOI: 10.21203/rs.3.rs-4047486/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Breast cancer bone metastases increase fracture risk and are a major cause of morbidity and mortality among women. Upon colonization by tumor cells, the bone microenvironment undergoes profound reprogramming to support cancer progression that disrupts the balance between osteoclasts and osteoblasts, leading to bone lesions. Whether such reprogramming affects matrix-embedded osteocytes remains poorly understood. Here, we demonstrate that osteocytes in breast cancer bone metastasis develop premature senescence and a distinctive senescence-associated secretory phenotype (SASP) that favors bone destruction. Single-cell RNA sequencing identified osteocytes from mice with breast cancer bone metastasis enriched in senescence and SASP markers and pro-osteoclastogenic genes. Using multiplex in situ hybridization and AI-assisted analysis, we detected osteocytes with senescence-associated distension of satellites, telomere dysfunction, and p16Ink4a expression in mice and patients with breast cancer bone metastasis. In vitro and ex vivo organ cultures showed that breast cancer cells promote osteocyte senescence and enhance their osteoclastogenic potential. Clearance of senescent cells with senolytics suppressed bone resorption and preserved bone mass in mice with breast cancer bone metastasis. These results demonstrate that osteocytes undergo pathological reprogramming by breast cancer cells and identify osteocyte senescence as an initiating event triggering bone destruction in breast cancer metastases.
Collapse
Affiliation(s)
- Manish Adhikari
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Japneet Kaur
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Hayley M. Sabol
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Aric Anloague
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Sharmin Khan
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Noriyoshi Kurihara
- Division of Hematology and Oncology, Department of Medicine, Indiana University, Indianapolis, IN, US
| | | | - Christina Møller Andreasen
- Molecular Bone Histology lab, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Clinical Pathologyogy, Odense University Hospital, Odense University Hospital, Odense, Denmark
| | - C. Lowry Barnes
- Department of Orthopedic Surgery; University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Jeffrey B. Stambough
- Department of Orthopedic Surgery; University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Michela Palmieri
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US
| | - Olivia Reyes-Castro
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US
| | - Elena Ambrogini
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US
| | - Maria Almeida
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US
| | - Charles A. O’Brien
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Intawat Nookaw
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, US
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, US
| | - Jesus Delgado-Calle
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, US
| |
Collapse
|
9
|
Elaasser B, Arakil N, Mohammad KS. Bridging the Gap in Understanding Bone Metastasis: A Multifaceted Perspective. Int J Mol Sci 2024; 25:2846. [PMID: 38474093 PMCID: PMC10932255 DOI: 10.3390/ijms25052846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
The treatment of patients with advanced cancer poses clinical problems due to the complications that arise as the disease progresses. Bone metastases are a common problem that cancer patients may face, and currently, there are no effective drugs to treat these individuals. Prostate, breast, and lung cancers often spread to the bone, causing significant and disabling health conditions. The bone is a highly active and dynamic tissue and is considered a favorable environment for the growth of cancer. The role of osteoblasts and osteoclasts in the process of bone remodeling and the way in which their interactions change during the progression of metastasis is critical to understanding the pathophysiology of this disease. These interactions create a self-perpetuating loop that stimulates the growth of metastatic cells in the bone. The metabolic reprogramming of both cancer cells and cells in the bone microenvironment has serious implications for the development and progression of metastasis. Insight into the process of bone remodeling and the systemic elements that regulate this process, as well as the cellular changes that occur during the progression of bone metastases, is critical to the discovery of a cure for this disease. It is crucial to explore different therapeutic options that focus specifically on malignancy in the bone microenvironment in order to effectively treat this disease. This review will focus on the bone remodeling process and the effects of metabolic disorders as well as systemic factors like hormones and cytokines on the development of bone metastases. We will also examine the various therapeutic alternatives available today and the upcoming advances in novel treatments.
Collapse
Affiliation(s)
| | | | - Khalid S. Mohammad
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 1153, Saudi Arabia; (B.E.); (N.A.)
| |
Collapse
|
10
|
Tang G, Huang S, Luo J, Wu Y, Zheng S, Tong R, Zhong L, Shi J. Advances in research on potential inhibitors of multiple myeloma. Eur J Med Chem 2023; 262:115875. [PMID: 37879169 DOI: 10.1016/j.ejmech.2023.115875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/03/2023] [Accepted: 10/12/2023] [Indexed: 10/27/2023]
Abstract
Multiple myeloma (MM) is a common hematological malignancy. Although recent clinical applications of immunomodulatory drugs, proteasome inhibitors and CD38-targeting antibodies have significantly improved the outcome of MM patient with increased survival, the incidence of drug resistance and severe treatment-related complications is gradually on the rise. This review article summarizes the characteristics and clinical investigations of several MM drugs in clinical trials, including their structures, mechanisms of action, structure-activity relationships, and clinical study progress. Furthermore, the application potentials of the drugs that have not yet entered clinical trials are also reviewed. The review also outlines the future directions of MM drug development.
Collapse
Affiliation(s)
- Guoyuan Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Shan Huang
- Cancer Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Ji Luo
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Yingmiao Wu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Shuai Zheng
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Rongsheng Tong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.
| | - Ling Zhong
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China; Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, Sichuan, 610044, China.
| | - Jianyou Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.
| |
Collapse
|
11
|
Platonova N, Lazzari E, Colombo M, Falleni M, Tosi D, Giannandrea D, Citro V, Casati L, Ronchetti D, Bolli N, Neri A, Torricelli F, Crews LA, Jamieson CHM, Chiaramonte R. The Potential of JAG Ligands as Therapeutic Targets and Predictive Biomarkers in Multiple Myeloma. Int J Mol Sci 2023; 24:14558. [PMID: 37834003 PMCID: PMC10572399 DOI: 10.3390/ijms241914558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/03/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
The NOTCH ligands JAG1 and JAG2 have been correlated in vitro with multiple myeloma (MM) cell proliferation, drug resistance, self-renewal and a pathological crosstalk with the tumor microenvironment resulting in angiogenesis and osteoclastogenesis. These findings suggest that a therapeutic approach targeting JAG ligands might be helpful for the care of MM patients and lead us to explore the role of JAG1 and JAG2 in a MM in vivo model and primary patient samples. JAG1 and JAG2 protein expression represents a common feature in MM cell lines; therefore, we assessed their function through JAG1/2 conditional silencing in a MM xenograft model. We observed that JAG1 and JAG2 showed potential as therapeutic targets in MM, as their silencing resulted in a reduction in the tumor burden. Moreover, JAG1 and JAG2 protein expression in MM patients was positively correlated with the presence of MM cells in patients' bone marrow biopsies. Finally, taking advantage of the Multiple Myeloma Research Foundation (MMRF) CoMMpass global dataset, we showed that JAG2 gene expression level was a predictive biomarker associated with patients' overall survival and progression-free survival, independently from other main molecular or clinical features. Overall, these results strengthened the rationale for the development of a JAG1/2-tailored approach and the use of JAG2 as a predictive biomarker in MM.
Collapse
Affiliation(s)
- Natalia Platonova
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (N.P.); (E.L.); (M.C.); (M.F.); (D.T.); (D.G.); (V.C.); (L.C.)
| | - Elisa Lazzari
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (N.P.); (E.L.); (M.C.); (M.F.); (D.T.); (D.G.); (V.C.); (L.C.)
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, La Jolla, CA 92093, USA; (L.A.C.); (C.H.M.J.)
- UC San Diego Sanford, Stem Cell Institute, La Jolla, CA 92037, USA
| | - Michela Colombo
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (N.P.); (E.L.); (M.C.); (M.F.); (D.T.); (D.G.); (V.C.); (L.C.)
| | - Monica Falleni
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (N.P.); (E.L.); (M.C.); (M.F.); (D.T.); (D.G.); (V.C.); (L.C.)
- Unit of Pathology A.O. San Paolo, Via A. Di Rudinì 8, 20142 Milan, Italy
| | - Delfina Tosi
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (N.P.); (E.L.); (M.C.); (M.F.); (D.T.); (D.G.); (V.C.); (L.C.)
- Unit of Pathology A.O. San Paolo, Via A. Di Rudinì 8, 20142 Milan, Italy
| | - Domenica Giannandrea
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (N.P.); (E.L.); (M.C.); (M.F.); (D.T.); (D.G.); (V.C.); (L.C.)
| | - Valentina Citro
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (N.P.); (E.L.); (M.C.); (M.F.); (D.T.); (D.G.); (V.C.); (L.C.)
| | - Lavinia Casati
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (N.P.); (E.L.); (M.C.); (M.F.); (D.T.); (D.G.); (V.C.); (L.C.)
| | - Domenica Ronchetti
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy; (D.R.); (N.B.)
| | - Niccolò Bolli
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy; (D.R.); (N.B.)
- Hematology, Fondazione Cà Granda IRCCS Policlinico, 20122 Milan, Italy
| | - Antonino Neri
- Scientific Directorate, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy;
| | - Federica Torricelli
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy;
| | - Leslie A. Crews
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, La Jolla, CA 92093, USA; (L.A.C.); (C.H.M.J.)
| | - Catriona H. M. Jamieson
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, La Jolla, CA 92093, USA; (L.A.C.); (C.H.M.J.)
- UC San Diego Sanford, Stem Cell Institute, La Jolla, CA 92037, USA
| | - Raffaella Chiaramonte
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (N.P.); (E.L.); (M.C.); (M.F.); (D.T.); (D.G.); (V.C.); (L.C.)
| |
Collapse
|
12
|
Wells KV, Krackeler ML, Jathal MK, Parikh M, Ghosh PM, Leach JK, Genetos DC. Prostate cancer and bone: clinical presentation and molecular mechanisms. Endocr Relat Cancer 2023; 30:e220360. [PMID: 37226936 PMCID: PMC10696925 DOI: 10.1530/erc-22-0360] [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: 11/21/2022] [Accepted: 05/24/2023] [Indexed: 05/26/2023]
Abstract
Prostate cancer (PCa) is an increasingly prevalent health problem in the developed world. Effective treatment options exist for localized PCa, but metastatic PCa has fewer treatment options and shorter patient survival. PCa and bone health are strongly entwined, as PCa commonly metastasizes to the skeleton. Since androgen receptor signaling drives PCa growth, androgen-deprivation therapy whose sequelae reduce bone strength constitutes the foundation of advanced PCa treatment. The homeostatic process of bone remodeling - produced by concerted actions of bone-building osteoblasts, bone-resorbing osteoclasts, and regulatory osteocytes - may also be subverted by PCa to promote metastatic growth. Mechanisms driving skeletal development and homeostasis, such as regional hypoxia or matrix-embedded growth factors, may be subjugated by bone metastatic PCa. In this way, the biology that sustains bone is integrated into adaptive mechanisms for the growth and survival of PCa in bone. Skeletally metastatic PCa is difficult to investigate due to the entwined nature of bone biology and cancer biology. Herein, we survey PCa from origin, presentation, and clinical treatment to bone composition and structure and molecular mediators of PCa metastasis to bone. Our intent is to quickly yet effectively reduce barriers to team science across multiple disciplines that focuses on PCa and metastatic bone disease. We also introduce concepts of tissue engineering as a novel perspective to model, capture, and study complex cancer-microenvironment interactions.
Collapse
Affiliation(s)
- Kristina V Wells
- Department of Anatomy, Physiology, and Cell Biology, University of California Davis School of Veterinary Medicine, Davis, California, USA
| | - Margaret L Krackeler
- Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, California, USA
| | - Maitreyee K Jathal
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, California, USA
- Veterans Affairs-Northern California Health System, Mather, California, USA
| | - Mamta Parikh
- Division of Hematology and Oncology, School of Medicine, University of California Davis, Sacramento, California, USA
| | - Paramita M Ghosh
- Veterans Affairs-Northern California Health System, Mather, California, USA
- Department of Urologic Surgery, School of Medicine, University of California Davis, Sacramento, California, USA
| | - J Kent Leach
- Department of Orthopaedic Surgery, School of Medicine, University of California Davis, Sacramento, California, USA
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Damian C Genetos
- Department of Anatomy, Physiology, and Cell Biology, University of California Davis School of Veterinary Medicine, Davis, California, USA
| |
Collapse
|
13
|
Kitase Y, Prideaux M. Regulation of the Osteocyte Secretome with Aging and Disease. Calcif Tissue Int 2023; 113:48-67. [PMID: 37148298 DOI: 10.1007/s00223-023-01089-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/21/2023] [Indexed: 05/08/2023]
Abstract
As the most numerous and long-lived of all bone cells, osteocytes have essential functions in regulating skeletal health. Through the lacunar-canalicular system, secreted proteins from osteocytes can reach cells throughout the bone. Furthermore, the intimate connectivity between the lacunar-canalicular system and the bone vasculature allows for the transport of osteocyte-secreted factors into the circulation to reach the entire body. Local and endocrine osteocyte signaling regulates physiological processes such as bone remodeling, bone mechanoadaptation, and mineral homeostasis. However, these processes are disrupted by impaired osteocyte function induced by aging and disease. Dysfunctional osteocyte signaling is now associated with the pathogenesis of many disorders, including chronic kidney disease, cancer, diabetes mellitus, and periodontitis. In this review, we focus on the targeting of bone and extraskeletal tissues by the osteocyte secretome. In particular, we highlight the secreted osteocyte proteins, which are known to be dysregulated during aging and disease, and their roles during disease progression. We also discuss how therapeutic or genetic targeting of osteocyte-secreted proteins can improve both skeletal and systemic health.
Collapse
Affiliation(s)
- Yukiko Kitase
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Matthew Prideaux
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
14
|
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: 6] [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
|