1
|
Rinella L, Fiorentino G, Compagno M, Grange C, Cedrino M, Marano F, Bosco O, Vissio E, Delsedime L, D'Amelio P, Bussolati B, Arvat E, Catalano MG. Dickkopf-1 (DKK1) drives growth and metastases in castration-resistant prostate cancer. Cancer Gene Ther 2024; 31:1266-1279. [PMID: 38740881 DOI: 10.1038/s41417-024-00783-7] [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: 01/30/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024]
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) is associated with a poor prognosis and remains an incurable fatal disease. Therefore, the identification of molecular markers involved in cancer progression is urgently needed to develop more-effective therapies. The present study investigated the role of the Wnt signaling modulator Dickkopf-1 (DKK1) in the growth and metastatic progression of mCRPC. DKK1 silencing through siRNA and deletion via CRISPR/Cas9 editing were performed in two different metastatic castration-resistant prostate cancer cell lines (PC3 and DU145). A xenograft tumor model was used to assess tumor growth and metastases. In in vitro experiments, both DKK1 silencing and deletion reduced cell growth and migration of both cell lines. DKK1 knockout clones (DKK1-KO) exhibited cell cycle arrest, tubulin reorganization, and modulation of tumor metastasis-associated genes. Furthermore, in DKK1-KO cells, E-cadherin re-expression and its membrane co-localization with β-catenin were observed, contributing to reduced migration; Cadherin-11, known to increase during epithelial-mesenchymal transition, was down-regulated in DKK1-KO cells. In the xenograft mouse model, DKK1 deletion not only reduced tumor growth but also inhibited the formation of lung metastases. In conclusion, our findings support the key role of DKK1 in the growth and metastatic dissemination of mCRPC, both in vitro and in vivo.
Collapse
Affiliation(s)
- Letizia Rinella
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Mara Compagno
- Center for Experimental Research and Medical Studies (CeRMS), Molinette Hospital, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Cristina Grange
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Massimo Cedrino
- Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Francesca Marano
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Ornella Bosco
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Elena Vissio
- Unit of Pathology, Molinette Hospital, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Luisa Delsedime
- Unit of Pathology, Molinette Hospital, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | | | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Emanuela Arvat
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | |
Collapse
|
2
|
Abdullah KM, Sharma G, Takkar S, Kaushal JB, Pothuraju R, Chakravarti B, Batra SK, Siddiqui JA. α-lipoic acid modulates prostate cancer cell growth and bone cell differentiation. Sci Rep 2024; 14:4404. [PMID: 38388663 PMCID: PMC10884017 DOI: 10.1038/s41598-024-54479-x] [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: 07/31/2023] [Accepted: 02/12/2024] [Indexed: 02/24/2024] Open
Abstract
Prostate cancer (PCa) progression leads to bone modulation in approximately 70% of affected men. A nutraceutical, namely, α-lipoic acid (α-LA), is known for its potent anti-cancer properties towards various cancers and has been implicated in treating and promoting bone health. Our study aimed to explore the molecular mechanism behind the role of α-LA as therapeutics in preventing PCa and its associated bone modulation. Notably, α-LA treatment significantly reduced the cell viability, migration, and invasion of PCa cell lines in a dose-dependent manner. In addition, α-LA supplementation dramatically increased reactive oxygen species (ROS) levels and HIF-1α expression, which started the downstream molecular cascade and activated JNK/caspase-3 signaling pathway. Flow cytometry data revealed the arrest of the cell cycle in the S-phase, which has led to apoptosis of PCa cells. Furthermore, the results of ALP (Alkaline phosphatase) and TRAP (tartrate-resistant acid phosphatase) staining signifies that α-LA supplementation diminished the PCa-mediated differentiation of osteoblasts and osteoclasts, respectively, in the MC3T3-E1 and bone marrow macrophages (BMMs) cells. In summary, α-LA supplementation enhanced cellular apoptosis via increased ROS levels, HIF-1α expression, and JNK/caspase-3 signaling pathway in advanced human PCa cell lines. Also, the treatment of α-LA improved bone health by reducing PCa-mediated bone cell modulation.
Collapse
Affiliation(s)
- K M Abdullah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Gunjan Sharma
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Simran Takkar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jyoti B Kaushal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Ramesh Pothuraju
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - Bandana Chakravarti
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, 226014, India
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA.
| | - Jawed A Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA.
| |
Collapse
|
3
|
Askari E, Harsini S. Unraveling the Hypocalcemic Response to 177Lu-Prostate-Specific Membrane Antigen Therapy. J Nucl Med 2024; 65:332-333. [PMID: 37734839 DOI: 10.2967/jnumed.123.266368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 07/14/2023] [Accepted: 07/20/2023] [Indexed: 09/23/2023] Open
|
4
|
Chiarito M, Piacente L, Chaoul N, Pontrelli P, D'Amato G, Grandone A, Russo G, Street ME, Wasniewska MG, Brunetti G, Faienza MF. Role of Wnt-signaling inhibitors DKK-1 and sclerostin in bone fragility associated with Turner syndrome. J Endocrinol Invest 2022; 45:1255-1263. [PMID: 35237949 PMCID: PMC9098532 DOI: 10.1007/s40618-022-01760-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 02/07/2022] [Indexed: 12/22/2022]
Abstract
PURPOSE Girls affected with Turner syndrome (TS) present with low bone mineral density (BMD) and osteopenia/osteoporosis. Thus, they have an increased risk to develop fractures compared to normal population. The aim of this study was to deepen the pathophysiology of skeletal fragility in TS subjects by evaluating the serum levels of Dickkopf-1 (DKK-1) and sclerostin, main regulators of bone mass, as well as the percentage of circulating osteoblast precursors (OCPs). METHODS Thirty-four TS girls and 24 controls were recruited. All subjects underwent anthropometric measures (height, weight, body mass index-BMI). A peripheral venous blood sample was collected to determine serum levels of active intact parathyroid hormone (PTH), 25-OH vitamin D, calcium, phosphorus, bone alkaline phosphatase (bALP), osteocalcin, sclerostin, DKK-1, RANKL and OPG. OCPs were detected by flow cytometry. In TS subjects bone mineralization was measured at lumbar spine by dual energy X-ray absorptiometry (DXA). RESULTS bALP, 25-OH Vitamin D, and osteocalcin levels were significant lower in TS subjects than in the controls. Statistically significant higher levels of sclerostin, DKK-1 and RANKL were measured in patients compared with the controls. The percentage of OCPs did not show significant differences between patients and controls. Sclerostin and DKK-1 levels were related with anthropometric parameters, bone metabolism markers, HRT, rhGH therapy, RANKL and lumbar BMAD-Z-score. CONCLUSION TS patients showed higher levels of sclerostin and DKK-1 than controls which can be related to HRT, and to reduced bone formation markers as well as the increased bone resorption activity.
Collapse
Affiliation(s)
- M Chiarito
- Department of Biomedical Sciences and Human Oncology, Pediatric Unit, University of Bari "A. Moro", Bari, Italy
| | - L Piacente
- Department of Biomedical Sciences and Human Oncology, Pediatric Unit, University of Bari "A. Moro", Bari, Italy
| | - N Chaoul
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro", Bari, Italy
| | - P Pontrelli
- Department of Emergency and Organ Transplantation, Division of Nephrology, University of Bari "A. Moro", Bari, Italy
| | - G D'Amato
- Neonatal Intensive Care Unit, "Di Venere" Hospital, Bari, Italy
| | - A Grandone
- Department of Woman, Child and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - G Russo
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
| | - M E Street
- Department of Mother and Child, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - M G Wasniewska
- Pediatric Unit, Department of Human Pathology in Adulthood and Childhood, University of Messina, Messina, Italy
| | - G Brunetti
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University "A. Moro" of Bari, Bari, Italy
| | - M F Faienza
- Department of Biomedical Sciences and Human Oncology, Pediatric Unit, University of Bari "A. Moro", Bari, Italy.
| |
Collapse
|
5
|
Hinz N, Jücker M. AKT in Bone Metastasis of Solid Tumors: A Comprehensive Review. Cancers (Basel) 2021; 13:cancers13102287. [PMID: 34064589 PMCID: PMC8151478 DOI: 10.3390/cancers13102287] [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: 04/16/2021] [Revised: 05/05/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Bone metastasis is a frequent complication of solid tumors and leads to a reduced overall survival. Although much progress has been made in the field of tumor therapy in the last years, bone metastasis depicts a stage of the disease with a lack of appropriate therapeutical options. Hence, this review aims to present the role of AKT in bone metastasis of solid tumors to place the spotlight on AKT as a possible therapeutical approach for patients with bone metastases. Furthermore, we intended to discuss postulated underlying molecular mechanisms of the bone metastasis-promoting effect of AKT, especially in highly bone-metastatic breast, prostate, and lung cancer. To conclude, this review identified the AKT kinase as a potential therapeutical target in bone metastasis and revealed remaining questions, which need to be addressed in further research projects. Abstract Solid tumors, such as breast cancer and prostate cancer, often form bone metastases in the course of the disease. Patients with bone metastases frequently develop complications, such as pathological fractures or hypercalcemia and exhibit a reduced life expectancy. Thus, it is of vital importance to improve the treatment of bone metastases. A possible approach is to target signaling pathways, such as the PI3K/AKT pathway, which is frequently dysregulated in solid tumors. Therefore, we sought to review the role of the serine/threonine kinase AKT in bone metastasis. In general, activation of AKT signaling was shown to be associated with the formation of bone metastases from solid tumors. More precisely, AKT gets activated in tumor cells by a plethora of bone-derived growth factors and cytokines. Subsequently, AKT promotes the bone-metastatic capacities of tumor cells through distinct signaling pathways and secretion of bone cell-stimulating factors. Within the crosstalk between tumor and bone cells, also known as the vicious cycle, the stimulation of osteoblasts and osteoclasts also causes activation of AKT in these cells. As a consequence, bone metastasis is reduced after experimental inhibition of AKT. In summary, AKT signaling could be a promising therapeutical approach for patients with bone metastases of solid tumors.
Collapse
|
6
|
Henrich SE, McMahon KM, Plebanek MP, Calvert AE, Feliciano TJ, Parrish S, Tavora F, Mega A, De Souza A, Carneiro BA, Thaxton CS. Prostate cancer extracellular vesicles mediate intercellular communication with bone marrow cells and promote metastasis in a cholesterol-dependent manner. J Extracell Vesicles 2020; 10:e12042. [PMID: 33408816 PMCID: PMC7775568 DOI: 10.1002/jev2.12042] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 11/10/2020] [Accepted: 11/28/2020] [Indexed: 12/17/2022] Open
Abstract
Primary tumours can establish long-range communication with distant organs to transform them into fertile soil for circulating tumour cells to implant and proliferate, a process called pre-metastatic niche (PMN) formation. Tumour-derived extracellular vesicles (EV) are potent mediators of PMN formation due to their diverse complement of pro-malignant molecular cargo and their propensity to target specific cell types (Costa-Silva et al., 2015; Hoshino et al., 2015; Peinado et al., 2012; Peinado et al., 2017). While significant progress has been made to understand the mechanisms by which pro-metastatic EVs create tumour-favouring microenvironments at pre-metastatic organ sites, comparatively little attention has been paid to the factors intrinsic to recipient cells that may modify the extent to which pro-metastatic EV signalling is received and transduced. Here, we investigated the role of recipient cell cholesterol homeostasis in prostate cancer (PCa) EV-mediated signalling and metastasis. Using a bone metastatic model of enzalutamide-resistant PCa, we first characterized an axis of EV-mediated communication between PCa cells and bone marrow that is marked by in vitro and in vivo PCa EV uptake by bone marrow myeloid cells, activation of NF-κB signalling, enhanced osteoclast differentiation, and reduced myeloid thrombospondin-1 expression. We then employed a targeted, biomimetic approach to reduce myeloid cell cholesterol in vitro and in vivo prior to conditioning with PCa EVs. Reducing myeloid cell cholesterol prevented the uptake of PCa EVs by recipient myeloid cells, abolished NF-κB activity and osteoclast differentiation, stabilized thrombospondin-1 expression, and reduced metastatic burden by 77%. These results demonstrate that cholesterol homeostasis in bone marrow myeloid cells regulates pro-metastatic EV signalling and metastasis by acting as a gatekeeper for EV signal transduction.
Collapse
Affiliation(s)
- Stephen E. Henrich
- Department of UrologyFeinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
- Simpson Querrey Institute for BioNanotechnologyNorthwestern UniversityChicagoIllinoisUSA
| | - Kaylin M. McMahon
- Department of UrologyFeinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
- Simpson Querrey Institute for BioNanotechnologyNorthwestern UniversityChicagoIllinoisUSA
| | - Michael P. Plebanek
- Department of UrologyFeinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
- Simpson Querrey Institute for BioNanotechnologyNorthwestern UniversityChicagoIllinoisUSA
| | - Andrea E. Calvert
- Department of UrologyFeinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
- Simpson Querrey Institute for BioNanotechnologyNorthwestern UniversityChicagoIllinoisUSA
| | - Timothy J. Feliciano
- Department of UrologyFeinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
- Simpson Querrey Institute for BioNanotechnologyNorthwestern UniversityChicagoIllinoisUSA
| | - Samuel Parrish
- Department of UrologyFeinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Fabio Tavora
- Department of PathologyMessejana Heart and Lung HospitalFortalezaBrazil
| | - Anthony Mega
- Warren Alpert Medical SchoolBrown UniversityProvidenceRhode IslandUSA
- Lifespan Cancer InstituteProvidenceRhode IslandUSA
| | - Andre De Souza
- Warren Alpert Medical SchoolBrown UniversityProvidenceRhode IslandUSA
- Lifespan Cancer InstituteProvidenceRhode IslandUSA
| | - Benedito A. Carneiro
- Warren Alpert Medical SchoolBrown UniversityProvidenceRhode IslandUSA
- Lifespan Cancer InstituteProvidenceRhode IslandUSA
| | - C. Shad Thaxton
- Department of UrologyFeinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
- Simpson Querrey Institute for BioNanotechnologyNorthwestern UniversityChicagoIllinoisUSA
- Robert H. Lurie Comprehensive Cancer CenterNorthwestern UniversityChicagoIllinoisUSA
| |
Collapse
|
7
|
Dai J, Escara-Wilke J, Keller JM, Jung Y, Taichman RS, Pienta KJ, Keller ET. Primary prostate cancer educates bone stroma through exosomal pyruvate kinase M2 to promote bone metastasis. J Exp Med 2019; 216:2883-2899. [PMID: 31548301 PMCID: PMC6888980 DOI: 10.1084/jem.20190158] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/30/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer (PCa) metastasizes selectively to bone through unknown mechanisms. In the current study, we identified exosome-mediated transfer of pyruvate kinase M2 (PKM2) from PCa cells into bone marrow stromal cells (BMSCs) as a novel mechanism through which primary tumor-derived exosomes promote premetastatic niche formation. We found that PKM2 up-regulates BMSC CXCL12 production in a HIF-1α-dependent fashion, which subsequently enhances PCa seeding and growth in the bone marrow. Furthermore, serum-derived exosomes from patients with either primary PCa or PCa metastasis, as opposed to healthy men, reveal that increased exosome PKM2 expression is associated with metastasis, suggesting clinical relevance of exosome PKM2 in PCa. Targeting the exosome-induced CXCL12 axis diminished exosome-mediated bone metastasis. In summary, primary PCa cells educate the bone marrow to create a premetastatic niche through primary PCa exosome-mediated transfer of PKM2 into BMSCs and subsequent up-regulation of CXCL12. This novel mechanism indicates the potential for exosome PKM2 as a biomarker and suggests therapeutic targets for PCa bone metastasis.
Collapse
Affiliation(s)
- Jinlu Dai
- Department of Urology, Medical School, University of Michigan, Ann Arbor, MI
| | - June Escara-Wilke
- Department of Urology, Medical School, University of Michigan, Ann Arbor, MI
| | - Jill M Keller
- Department of Urology, Medical School, University of Michigan, Ann Arbor, MI
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI
| | - Younghun Jung
- Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Russell S Taichman
- Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Kenneth J Pienta
- Department of Urology, Brady Urological Institute, Johns Hopkins University, Baltimore, MD
| | - Evan T Keller
- Department of Urology, Medical School, University of Michigan, Ann Arbor, MI
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI
| |
Collapse
|
8
|
The Mode-of-Action of Targeted Alpha Therapy Radium-223 as an Enabler for Novel Combinations to Treat Patients with Bone Metastasis. Int J Mol Sci 2019; 20:ijms20163899. [PMID: 31405099 PMCID: PMC6720648 DOI: 10.3390/ijms20163899] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 12/12/2022] Open
Abstract
Bone metastasis is a common clinical complication in several cancer types, and it causes a severe reduction in quality of life as well as lowering survival time. Bone metastases proceed through a vicious self-reinforcing cycle that can be osteolytic or osteoblastic in nature. The vicious cycle is characterized by cancer cells residing in bone releasing signal molecules that promote the differentiation of osteoclasts and osteoblasts either directly or indirectly. The increased activity of osteoclasts and osteoblasts then increases bone turnover, which releases growth factors that benefit metastatic cancer cells. In order to improve the prognosis of patients with bone metastases this cycle must be broken. Radium-223 dichloride (radium-223), the first targeted alpha therapy (TAT) approved, is an osteomimetic radionuclide that is incorporated into bone metastases where its high-linear energy transfer alpha radiation disrupts both the activity of bone cells and cancer cells. Therefore, radium-223 treatment has been shown preclinically to directly affect cancer cells in both osteolytic breast cancer and osteoblastic prostate cancer bone metastases as well as to inhibit the differentiation of osteoblasts and osteoclasts. Clinical studies have demonstrated an increase in survival in patients with metastatic castration-resistant prostate cancer. Due to the effectiveness and low toxicity of radium-223, several novel combination treatment strategies are currently eliciting considerable research interest.
Collapse
|
9
|
Interleukin-7 Contributes to the Invasiveness of Prostate Cancer Cells by Promoting Epithelial-Mesenchymal Transition. Sci Rep 2019; 9:6917. [PMID: 31061414 PMCID: PMC6502845 DOI: 10.1038/s41598-019-43294-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 04/17/2019] [Indexed: 01/05/2023] Open
Abstract
Precise mechanisms underlying interleukin-7 (IL-7)-mediated tumor invasion remain unclear. Thus, we investigated the role of IL-7 in tumor invasiveness using metastatic prostate cancer PC-3 cell line derivatives, and assessed the potential of IL-7 as a clinical target using a Janus kinase (JAK) inhibitor and an IL-7-blocking antibody. We found that IL-7 stimulated wound-healing migration and invasion of PC-3 cells, increased phosphorylation of signal transducer and activator of transcription 5, Akt, and extracellular signal-regulated kinase. On the other hand, a JAK inhibitor and an IL-7-blocking antibody decreased the invasiveness of PC-3 cells. IL-7 increased tumor sphere formation and expression of epithelial–mesenchymal transition (EMT) markers. Importantly, lentiviral delivery of IL-7Rα to PC-3 cells significantly increased bone metastasis in an experimental murine metastasis model compared to controls. The gene expression profile of human prostate cancer cells from The Cancer Genome Atlas revealed that EMT pathways are strongly associated with prostate cancers that highly express both IL-7 and IL-7Rα. Collectively, these data suggest that IL-7 and/or IL-7Rα are promising targets of inhibiting tumor metastasis.
Collapse
|
10
|
de Vries TJ, El Bakkali I, Kamradt T, Schett G, Jansen IDC, D'Amelio P. What Are the Peripheral Blood Determinants for Increased Osteoclast Formation in the Various Inflammatory Diseases Associated With Bone Loss? Front Immunol 2019; 10:505. [PMID: 30941138 PMCID: PMC6434996 DOI: 10.3389/fimmu.2019.00505] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 02/25/2019] [Indexed: 01/18/2023] Open
Abstract
Local priming of osteoclast precursors (OCp) has long been considered the main and obvious pathway that takes place in the human body, where local bone lining cells and RANKL-expressing osteocytes may facilitate the differentiation of OCp. However, priming of OCp away from bone, such as in inflammatory tissues, as revealed in peripheral blood, may represent a second pathway, particularly relevant in individuals who suffer from systemic bone loss such as prevalent in inflammatory diseases. In this review, we used a systematic approach to review the literature on osteoclast formation in peripheral blood in patients with inflammatory diseases associated with bone loss. Only studies that compared inflammatory (bone) disease with healthy controls in the same study were included. Using this core collection, it becomes clear that experimental osteoclastogenesis using peripheral blood from patients with bone loss diseases in prevalent diseases such as rheumatoid arthritis, osteoporosis, periodontitis, and cancer-related osteopenia unequivocally point toward an intrinsically increased osteoclast formation and activation. In particular, such increased osteoclastogenesis already takes place without the addition of the classical osteoclastogenesis cytokines M-CSF and RANKL in vitro. We show that T-cells and monocytes as OCp are the minimal demands for such unstimulated osteoclast formation. In search for common and disease-specific denominators of the diseases with inflammation-driven bone loss, we demonstrate that altered T-cell activity and a different composition—such as the CD14+CD16+ vs. CD14+CD16– monocytes—and priming of OCp with increased M-CSF, RANKL, and TNF- α levels in peripheral blood play a role in increased osteoclast formation and activity. Future research will likely uncover the barcodes of the OCp in the various inflammatory diseases associated with bone loss.
Collapse
Affiliation(s)
- Teun J de Vries
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ismail El Bakkali
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Thomas Kamradt
- Institute of Immunology, Universitätsklinikum Jena, Jena, Germany
| | - Georg Schett
- Department of Internal Medicine III, Friedrich-Alexander University Erlangen-Nürnberg and Universitatsklinikum Erlangen, Erlangen, Germany
| | - Ineke D C Jansen
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Patrizia D'Amelio
- Gerontology and Bone Metabolic Diseases Division, Department of Medical Science, University of Turin, Turin, Italy
| |
Collapse
|
11
|
Metformin; an old antidiabetic drug with new potentials in bone disorders. Biomed Pharmacother 2018; 109:1593-1601. [PMID: 30551413 DOI: 10.1016/j.biopha.2018.11.032] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 11/05/2018] [Accepted: 11/07/2018] [Indexed: 01/15/2023] Open
Abstract
The prevalence of diabetes mellitus especially type 2 diabetes mellitus is increasing all over the world. In addition to cardiomyopathy and nephropathy, diabetics are at higher risk of mortality and morbidity due to greater risk of bone fractures and skeletal abnormalities. Patients with diabetes mellitus have lower bone quality in comparison to their non-diabetic counterparts mainly because of hyperglycemia, toxic effects of advanced glycosylation end-products (AGEs) on bone tissue, and impaired bone microvascular system. AGEs may also contribute to the development of osteoarthritis further to osteoporosis. Therefore, glycemic control in diabetic patients is vital for bone health. Metformin, a widely used antidiabetic drug, has been shown to improve bone quality and decrease the risk of fractures in patients with diabetes in addition to glycemic control and improving insulin sensitivity. AMP activated protein kinase (AMPK), the key molecule in metformin antidiabetic mechanism of action, is also effective in signaling pathways involved in bone physiology. This review, discusses the molecules linking diabetes and bone turnover, role of AMPK in bone metabolism, and the effect of metformin as an activator of AMPK on bone disorders and malignancies.
Collapse
|
12
|
Salamanna F, Borsari V, Contartese D, Nicoli Aldini N, Fini M. Link between estrogen deficiency osteoporosis and susceptibility to bone metastases: A way towards precision medicine in cancer patients. Breast 2018; 41:42-50. [PMID: 30007267 DOI: 10.1016/j.breast.2018.06.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 06/21/2018] [Indexed: 01/18/2023] Open
Abstract
Different fields of cancer management consider bone health to be of increasing clinical importance for patients: 1) presence of bone metastases in many solid tumors, 2) use of bone-targeted treatments in the reduction of bone metastasis, 3) effects of cancer treatment on reproductive hormones, critical for normal bone remodeling maintenance. Additionally, bone microenvironment is further complicated by the decline of ovarian sex steroid production and by the related increase in inflammatory factors linked to menopause, which result in accelerated bone loss and increased risk of osteoporosis (OP). Similarly, cancers and metastasis to bone showed a close relationship with sex hormones (particularly estrogen). Thus, these findings raise a question: Could pre-existing estrogen deficiency OP promote and/or influence cancer cell homing and tumor growth in bone? Although some preclinical and clinical evidence exists, it is mandatory to understand this aspect that would be relevant in the clinical theatre, where physicians need to understand the treatments available to reduce the risk of skeletal disease in cancer patients. This descriptive systematic review summarizes preclinical and clinical studies dealing with bimodal interactions between pre-existing estrogen deficiency OP and bone metastasis development and provides evidence supporting differences in tumor growth and colonization between healthy and OP status. Few studies evaluated the impact of estrogen deficiency OP on the susceptibility to bone metastases. Therefore, implementing biological knowledge, could help researchers and clinicians to have a better comprehension of the importance of pre- and post-menopausal bone microenvironment and its clinical implications for precision medicine in cancer patients.
Collapse
Affiliation(s)
- F Salamanna
- Laboratory of Preclinical and Surgical Studies, Rizzoli RIT Department, IRCCS Rizzoli Orthopedic Institute, Via di Barbiano 1/10, Bologna, 40136, Italy
| | - V Borsari
- Laboratory of Preclinical and Surgical Studies, Rizzoli RIT Department, IRCCS Rizzoli Orthopedic Institute, Via di Barbiano 1/10, Bologna, 40136, Italy.
| | - D Contartese
- Laboratory of Preclinical and Surgical Studies, Rizzoli RIT Department, IRCCS Rizzoli Orthopedic Institute, Via di Barbiano 1/10, Bologna, 40136, Italy
| | - N Nicoli Aldini
- Laboratory of Preclinical and Surgical Studies, Rizzoli RIT Department, IRCCS Rizzoli Orthopedic Institute, Via di Barbiano 1/10, Bologna, 40136, Italy
| | - M Fini
- Laboratory of Biomechanics and Technology Innovation, Rizzoli RIT Department, IRCCS Rizzoli Orthopedic Institute, Via di Barbiano 1/10, Bologna, 40136, Italy
| |
Collapse
|
13
|
Farhat A, Jiang D, Cui D, Keller ET, Jackson TL. An integrative model of prostate cancer interaction with the bone microenvironment. Math Biosci 2017; 294:1-14. [PMID: 28919575 DOI: 10.1016/j.mbs.2017.09.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 08/27/2017] [Accepted: 09/11/2017] [Indexed: 12/13/2022]
Abstract
Despite advanced efforts in early diagnosis, aggressive surgical treatment, and use of targeted chemotherapies, the prognosis for many cancers is still dismal. This emphasizes the necessity to develop new strategies for understanding tumor growth and metastasis. Here we use a systems approach that combines mathematical modeling and numerical simulation to develop a predictive computational model for prostate cancer and its subversion of the bone microenvironment. This model simulates metastatic prostate cancer evolution, progressing from normal bone and hormone levels to quantifiable diseased states. The simulations clearly demonstrate phenomena similar to those found clinically in prostate cancer patients. In addition, the major prediction of this model is the existence of low and high osteogenic states that are markedly different from one another. The existence and potential realization of these steady states appear to be mediated by the Wnt signaling pathway and by the effects of PSA on TGF-β, which encourages the bone microenvironment to evolve. The model is used to explore several potential therapeutic strategies, with some potential drug targets showing more promise than others: in particular, completely blocking Wnt and greatly increasing DKK-1 had significant positive effects, while blocking RANKL did not improve the outcome.
Collapse
Affiliation(s)
- A Farhat
- Department of Mathematics, University of Michigan, Ann Arbor, MI 48109-1043, USA
| | - D Jiang
- Command Post 1412 Broadway, New York, NY 10018, USA
| | - D Cui
- Department of Urology, Shanghai General Hospital, Shanghi, China
| | - E T Keller
- Department of Urology, University of Michigan, Ann Arbor, MI 48109, USA.
| | - T L Jackson
- Department of Mathematics, University of Michigan, Ann Arbor, MI 48109-1043, USA.
| |
Collapse
|
14
|
Buondonno I, Rovera G, Sassi F, Rigoni MM, Lomater C, Parisi S, Pellerito R, Isaia GC, D’Amelio P. Vitamin D and immunomodulation in early rheumatoid arthritis: A randomized double-blind placebo-controlled study. PLoS One 2017; 12:e0178463. [PMID: 28582403 PMCID: PMC5459341 DOI: 10.1371/journal.pone.0178463] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 05/12/2017] [Indexed: 01/11/2023] Open
Abstract
The aim of this study was to evaluate differences in T helper cell sub-types and osteoclast (OCs) precursors in peripheral blood between patients affected by early rheumatoid arthritis (eRA) and healthy controls. The effect of administration of cholecalcipherol on clinical and laboratory parameters was subsequently evaluated, by a parallel, randomized double blind, placebo controlled trial. Thirty nine eRA patients and 31 age-matched controls were enrolled and compared for levels of 25OH vitamin D, T helper cell sub-types, OCs precursors including both classical and non-classical and pro-inflammatory cytokines at baseline. Eligible patients were female ≥18 years of age with a diagnosis of RA, as defined by the American College of Rheumatology 2010 criteria for <6 months prior to inclusion in the study. Patients with auto-immune or inflammatory diseases other than RA were excluded. Patients treated with glucocorticoids (GCs), disease modifying activity drugs and biologic agents within the past 6 months were also excluded. In the second phase of the study, eRA patients were randomly assigned to standard treatment with methotrexate (MTX) and GCs with (21) or without (18) cholecalcipherol (300,000 IU) and followed for 3 months; the randomization was done by computer generated tables to allocate treatments. Three patients didn’t come back to the follow up visit for personal reasons. None of the patients experienced adverse events. The main outcome measures were T cells phenotypes, OCs precursors and inflammatory cytokines. Secondary outcome measure were clinical parameters. In eRA, 25OH vitamin D levels were significantly lower. CD4+/IFNγ+,CD4+/IL4+, CD4+/IL17A+ and CD4+IL17A+IFNγ+, cells were increased in eRA as well as non-classical OCs precursors, whereas T regulatory cells were not altered. TNFα, TGFβ1, RANKL, IL-23 and IL-6 were increased in eRA. Non-classical OCs, IL-23 and IL-6 correlated with disease severity and activity. Standard treatment with MTX and GC ameliorated clinical symptoms and reduced IL-23, whereas it did not affect CD4+ cells sub-sets nor OCs precursors. After 3 months, the combined use of cholecalcipherol significantly ameliorated the effect of treatment on global health. In eRA, a significant imbalance in T CD4+ sub-types accompanied by increased levels of non-classical OCs precursors and pro-inflammatory cytokines was observed. A single dose of cholecalcipherol (300,000 IU) combined with standard treatment significantly ameliorates patients general health.
Collapse
Affiliation(s)
- Ilaria Buondonno
- Department of Medical Science, Gerontology and Bone Metabolic Disease Section, University of Torino, Torino, Italy
| | - Guido Rovera
- Rheumatology Unit, Ospedale Mauriziano, Torino, Italy
| | - Francesca Sassi
- Department of Medical Science, Gerontology and Bone Metabolic Disease Section, University of Torino, Torino, Italy
| | - Micol Maria Rigoni
- Department of Medical Science, Gerontology and Bone Metabolic Disease Section, University of Torino, Torino, Italy
| | | | - Simone Parisi
- Rheumatology Department, AOU Città della Salute e della Scienza di Torino, Torino, Italy
| | | | - Giovanni Carlo Isaia
- Department of Medical Science, Gerontology and Bone Metabolic Disease Section, University of Torino, Torino, Italy
| | - Patrizia D’Amelio
- Department of Medical Science, Gerontology and Bone Metabolic Disease Section, University of Torino, Torino, Italy
- * E-mail:
| |
Collapse
|
15
|
Pivetta E, Wassermann B, Bulian P, Steffan A, Colombatti A, Polesel J, Spessotto P. Functional osteoclastogenesis: the baseline variability in blood donor precursors is not associated with age and gender. Oncotarget 2016; 6:31889-900. [PMID: 26376614 PMCID: PMC4741648 DOI: 10.18632/oncotarget.5575] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 08/15/2015] [Indexed: 02/06/2023] Open
Abstract
Mononuclear osteoclast precursors circulate in the monocyte fraction of peripheral blood and form multinuclear cells with all osteoclastic phenotypic characteristics when cultured in the presence of macrophage colony stimulating factor (M-CSF) and receptor activator of nuclear factor kB ligand (RANKL). The method to obtain osteoclast precursors from peripheral blood is simple but the number of recovered osteoclasts is often largely insufficient for functional analyses. The original aim of this study was to develop a rapid and efficient method that could overcome the donor variability and enrich the osteoclast precursors from a small volume of peripheral blood as a basis for future clinical studies to correlate the differentiation potential of circulating osteoclast precursors with bone lesions in cancer patients. We improved the efficiency of osteoclastogenesis by reducing isolation and purification times and overcame the use of flow cytometry and immunomagnetic purification procedures. In our culture system the osteoclast number was increased several-fold and the precursors were able to reach a full differentiation within seven days of culture. Both age as well as gender differences in osteoclastogenesis efficiency were no longer evident by processing limited volume blood samples with this simple and rapid method.
Collapse
Affiliation(s)
- Eliana Pivetta
- Division of Experimental Oncology 2, Department of Translational Research, CRO-IRCCS, Aviano, Pordenone, Italy
| | - Bruna Wassermann
- Division of Experimental Oncology 2, Department of Translational Research, CRO-IRCCS, Aviano, Pordenone, Italy
| | - Pietro Bulian
- Clinical and Experimental Onco-Hematology Unit, CRO-IRCCS, Aviano, Pordenone, Italy
| | | | - Alfonso Colombatti
- Division of Experimental Oncology 2, Department of Translational Research, CRO-IRCCS, Aviano, Pordenone, Italy
| | - Jerry Polesel
- Unit of Epidemiology and Biostatistics, CRO-IRCSS, Aviano, Pordenone, Italy
| | - Paola Spessotto
- Division of Experimental Oncology 2, Department of Translational Research, CRO-IRCCS, Aviano, Pordenone, Italy
| |
Collapse
|
16
|
Xu W, Pang K, Zhou ZG, Chen YF, Mo T, Li M, Liu CB. Dickkopf 2 promotes proliferation and invasion via Wnt signaling in prostate cancer. Mol Med Rep 2016; 14:2283-8. [PMID: 27431620 DOI: 10.3892/mmr.2016.5502] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 03/01/2016] [Indexed: 11/06/2022] Open
Abstract
Wnt/β-catenin signaling dysregulation is involved in tumorigenesis. Furthermore, epigenetic modification of the Dickkopf (DKK) family (DKK1‑4) has been shown to be important in the regulation of Wnt signaling. However, the functions and mechanism of DKK2 in the development and progression of prostate cancer remain unclear. Therefore, the present study investigated the role of DKK2 in prostate cancer. The mRNA and protein expression levels of DKK2 in prostate cancer tissues and cells were assessed by reverse transcription‑quantitative polymerase chain reaction and western blotting, respectively. The biological function of DKK2 in prostate cancer was investigated using 3‑(4,5‑dimethylthiazol‑2‑yl)-2,5‑diphenyltetrazolium bromide and transwell invasion assays. DKK2 was demonstrated to be upregulated in prostate cancer tissues and cells, and knockdown of DKK2 suppressed cell proliferation and invasion. Furthermore, small interfering RNA targeting DKK2 inhibited the expression of β‑catenin, cyclin D1 and c‑Myc in prostate cancer cells. The present report suggested that DKK2 downregulation suppressed the proliferation and invasion of prostate cancer cells by inhibiting the Wnt/β‑catenin signaling pathway.
Collapse
Affiliation(s)
- Wei Xu
- Department of Urology, The First People's Hospital of Yulin, Yulin, Guangxi 537000, P.R. China
| | - Kuan Pang
- Department of Urology, The First People's Hospital of Yulin, Yulin, Guangxi 537000, P.R. China
| | - Ze-Guang Zhou
- Department of Urology, The First People's Hospital of Yulin, Yulin, Guangxi 537000, P.R. China
| | - Yi-Feng Chen
- Department of Urology, The First People's Hospital of Yulin, Yulin, Guangxi 537000, P.R. China
| | - Ting Mo
- Department of Urology, The First People's Hospital of Yulin, Yulin, Guangxi 537000, P.R. China
| | - Ming Li
- Basic Medicine College, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Cheng-Bei Liu
- Department of Urology, The First People's Hospital of Yulin, Yulin, Guangxi 537000, P.R. China
| |
Collapse
|
17
|
Browne AJ, Göbel A, Thiele S, Hofbauer LC, Rauner M, Rachner TD. p38 MAPK regulates the Wnt inhibitor Dickkopf-1 in osteotropic prostate cancer cells. Cell Death Dis 2016; 7:e2119. [PMID: 26913608 PMCID: PMC4849158 DOI: 10.1038/cddis.2016.32] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 01/05/2016] [Accepted: 01/22/2016] [Indexed: 12/17/2022]
Abstract
The Wnt inhibitor Dickkopf-1 (DKK-1) has been associated with the occurrence of bone metastases in osteotropic prostate cancer by inhibiting osteoblastogenesis. P38 mitogen-activated protein kinase (MAPK) activity is also dysregulated in advanced prostate cancer. However, the impact of p38 MAPK signaling on DKK-1 remains unknown. Inhibition of p38 MAPK signaling in osteolytic PC3 cells by small molecule inhibitors (doramapimod, LY2228820 and SB202190) suppressed DKK-1 expression, whereas activation of p38 MAPK by anisomycin increased DKK-1. Further dissection by targeting individual p38 MAPK isoforms with siRNA revealed a stronger role for MAPK11 than MAPK14 and MAPK12 in the regulation of DKK-1. Moreover, prostate cancer cells with a predominantly osteolytic phenotype produced sufficient amounts of DKK-1 to inhibit Wnt3a-induced osteoblastic differentiation in C2C12 cells. This inhibition was blocked directly by neutralizing DKK-1 using a specific antibody and also indirectly by blocking p38 MAPK. Furthermore, tissue expression in human prostate cancer revealed a correlation between p38 MAPK and DKK-1 expression with higher expression in tumor compared with normal tissues. These results reveal that p38 MAPK regulates DKK-1 in prostate cancer and may present a potential target in osteolytic prostate cancers.
Collapse
Affiliation(s)
- A J Browne
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - A Göbel
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - S Thiele
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - L C Hofbauer
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - M Rauner
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - T D Rachner
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
18
|
The effects of lung and prostate cancer bone metastasis on serum osteoprotegerin levels: a meta-analysis. Sci Rep 2015; 5:18324. [PMID: 26671549 PMCID: PMC4680868 DOI: 10.1038/srep18324] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 11/16/2015] [Indexed: 01/11/2023] Open
Abstract
Bone metastasis leads to skeletal-related events in final-stage cancer patients. The incidence of prostate and lung cancers increases yearly; these cancers readily invade the bone. Some recent studies have found that serum osteoprotegerin (OPG) levels may be altered in patients with bone metastasis, whereas other reports have produced inconsistent findings. Hence, we conducted a meta-analysis to examine the effects of lung and prostate cancer on serum OPG levels. A systematic literature search was conducted using PubMed, Medline, and CNKI to identify relevant studies. A total of 11 studies were included. The standardized mean difference (SMD) and 95% confidence interval (95% CI) of the bone metastasis (BM) group, the non-bone metastasis (BM-) group and healthy controls were calculated. In prostate cancer, serum OPG levels in the BM group were higher than in the BM- group and healthy controls. Additionally, no significant difference in serum OPG levels was found between the BM- group and healthy controls. In lung cancer, serum OPG levels in the BM and BM- groups were significantly increased compared with healthy controls. However, no significant difference in serum OPG levels was found between the BM and BM- groups. Studies with larger sample sizes are required to confirm these findings.
Collapse
|
19
|
Salamanna F, Maglio M, Borsari V, Giavaresi G, Aldini NN, Fini M. Peripheral Blood Mononuclear Cells Spontaneous Osteoclastogenesis: Mechanisms Driving the Process and Clinical Relevance in Skeletal Disease. J Cell Physiol 2015; 231:521-30. [DOI: 10.1002/jcp.25134] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 08/12/2015] [Indexed: 01/13/2023]
Affiliation(s)
- Francesca Salamanna
- Rizzoli Research Innovation Technology Department, Rizzoli Orthopaedic Institute; Laboratory of Biocompatibility, Technological Innovations and Advanced Therapies; Bologna Italy
| | - Melania Maglio
- Rizzoli Orthopaedic Institute; Laboratory of Preclinical and Surgical Studies; Bologna Italy
| | - Veronica Borsari
- Rizzoli Research Innovation Technology Department, Rizzoli Orthopaedic Institute; Laboratory of Biocompatibility, Technological Innovations and Advanced Therapies; Bologna Italy
| | - Gianluca Giavaresi
- Rizzoli Research Innovation Technology Department, Rizzoli Orthopaedic Institute; Laboratory of Biocompatibility, Technological Innovations and Advanced Therapies; Bologna Italy
- Rizzoli Orthopaedic Institute; Laboratory of Preclinical and Surgical Studies; Bologna Italy
- Rizzoli Orthopedic Institute; Laboratory of Tissue Engineering-Innovative Technology Platforms for Tissue Engineering (PON01-00829); Palermo Italy
| | - Nicolò Nicoli Aldini
- Rizzoli Research Innovation Technology Department, Rizzoli Orthopaedic Institute; Laboratory of Biocompatibility, Technological Innovations and Advanced Therapies; Bologna Italy
- Rizzoli Orthopaedic Institute; Laboratory of Preclinical and Surgical Studies; Bologna Italy
| | - Milena Fini
- Rizzoli Research Innovation Technology Department, Rizzoli Orthopaedic Institute; Laboratory of Biocompatibility, Technological Innovations and Advanced Therapies; Bologna Italy
- Rizzoli Orthopaedic Institute; Laboratory of Preclinical and Surgical Studies; Bologna Italy
| |
Collapse
|
20
|
Sudhan DR, Siemann DW. Cathepsin L targeting in cancer treatment. Pharmacol Ther 2015; 155:105-16. [PMID: 26299995 DOI: 10.1016/j.pharmthera.2015.08.007] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 08/17/2015] [Indexed: 12/14/2022]
Abstract
Proteolytic enzymes may serve as promising targets for novel therapeutic treatment strategies seeking to impede cancer progression and metastasis. One such enzyme is cathepsin L (CTSL), a lysosomal cysteine protease. CTSL upregulation, a common occurrence in a variety of human cancers, has been widely correlated with metastatic aggressiveness and poor patient prognosis. In addition, CTSL has been implicated to contribute to cancer-associated osteolysis, a debilitating morbidity affecting both life expectancy and the quality of life. In this review, we highlight the mechanisms by which CTSL contributes to tumor progression and dissemination and discuss the therapeutic utility of CTSL intervention strategies aimed at impeding metastatic progression and bone resorption.
Collapse
Affiliation(s)
- Dhivya R Sudhan
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Dietmar W Siemann
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA; Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
21
|
Mucci JM, Cuello MF, Kisinovsky I, Larroude M, Delpino MV, Rozenfeld PA. Proinflammatory and proosteoclastogenic potential of peripheral blood mononuclear cells from Gaucher patients: Implication for bone pathology. Blood Cells Mol Dis 2015; 55:134-43. [PMID: 26142329 DOI: 10.1016/j.bcmd.2015.05.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 04/04/2015] [Accepted: 05/26/2015] [Indexed: 01/18/2023]
Abstract
Gaucher disease (GD) is caused by mutations in the GBA gene that confer a deficient level of activity of glucocerebrosidase (GCase). This deficiency leads to the accumulation of the glycolipid glucocerebroside in the lysosomes of cells of monocyte/macrophage system. Bone compromise in Gaucher disease patients is the most disabling aspect of the disease. However, pathophysiological aspects of skeletal alterations are still poorly understood. On the other hand it is well known that inflammation is a key player in GD pathology. In this work, we revealed increased levels of the proinflammatory CD14(+)CD16(+) monocyte subset and increased inflammatory cytokine production by monocytes and T cells in the circulation of GD patients. We showed increased levels of osteoclast precursors in PBMC from patients and a higher expression of RANKL in the surface of T cells. PBMC from patients presented higher osteoclast differentiation compared to healthy controls when cultured in the presence of M-CSF alone or in combination with RANKL. In vitro treatment with Velaglucerase reduced osteoclast levels to control levels. On the other hand THP-1 derived osteoclast precursors cultured in the presence of conditioned media from PBMC of GD patients presented higher differentiation to active osteoclasts. This induction involved TNF-α and RANKL.
Collapse
Affiliation(s)
- J M Mucci
- IIFP, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata - CONICET, La Plata 1900, Argentina
| | - M F Cuello
- Servicio de Hematología, Hospital de Niños "Sor María Ludovica", La Plata, Argentina
| | | | - M Larroude
- Consultorio Larrea N° 1106 3°E, Buenos Aires, Argentina
| | - M V Delpino
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), Hospital de Clínicas "José de San Martín", Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - P A Rozenfeld
- IIFP, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata - CONICET, La Plata 1900, Argentina.
| |
Collapse
|
22
|
Mechanism of Action of IL-7 and Its Potential Applications and Limitations in Cancer Immunotherapy. Int J Mol Sci 2015; 16:10267-80. [PMID: 25955647 PMCID: PMC4463645 DOI: 10.3390/ijms160510267] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 04/16/2015] [Accepted: 04/29/2015] [Indexed: 01/10/2023] Open
Abstract
Interleukin-7 (IL-7) is a non-hematopoietic cell-derived cytokine with a central role in the adaptive immune system. It promotes lymphocyte development in the thymus and maintains survival of naive and memory T cell homeostasis in the periphery. Moreover, it is important for the organogenesis of lymph nodes (LN) and for the maintenance of activated T cells recruited into the secondary lymphoid organs (SLOs). The immune capacity of cancer patients is suppressed that is characterized by lower T cell counts, less effector immune cells infiltration, higher levels of exhausted effector cells and higher levels of immunosuppressive cytokines, such as transforming growth factor β (TGF-β). Recombinant human IL-7 (rhIL-7) is an ideal solution for the immune reconstitution of lymphopenia patients by promoting peripheral T cell expansion. Furthermore, it can antagonize the immunosuppressive network. In animal models, IL-7 has been proven to prolong the survival of tumor-bearing hosts. In this review, we will focus on the mechanism of action and applications of IL-7 in cancer immunotherapy and the potential restrictions for its usage.
Collapse
|
23
|
Mori G, D'Amelio P, Faccio R, Brunetti G. Bone-immune cell crosstalk: bone diseases. J Immunol Res 2015; 2015:108451. [PMID: 26000310 PMCID: PMC4427089 DOI: 10.1155/2015/108451] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 01/22/2015] [Accepted: 01/25/2015] [Indexed: 01/14/2023] Open
Abstract
Bone diseases are associated with great morbidity; thus, the understanding of the mechanisms leading to their development represents a great challenge to improve bone health. Recent reports suggest that a large number of molecules produced by immune cells affect bone cell activity. However, the mechanisms are incompletely understood. This review aims to shed new lights into the mechanisms of bone diseases involving immune cells. In particular, we focused our attention on the major pathogenic mechanism underlying periodontal disease, psoriatic arthritis, postmenopausal osteoporosis, glucocorticoid-induced osteoporosis, metastatic solid tumors, and multiple myeloma.
Collapse
Affiliation(s)
- Giorgio Mori
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy
| | - Patrizia D'Amelio
- Department of Medical Science, Section of Gerontology and Bone Metabolism Diseases, University of Torino, 10126 Torino, Italy
| | - Roberta Faccio
- Department of Orthopedics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Giacomina Brunetti
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, Section of Human Anatomy and Histology, University of Bari, 70124 Bari, Italy
| |
Collapse
|
24
|
Hardaway AL, Herroon MK, Rajagurubandara E, Podgorski I. Marrow adipocyte-derived CXCL1 and CXCL2 contribute to osteolysis in metastatic prostate cancer. Clin Exp Metastasis 2015; 32:353-68. [PMID: 25802102 DOI: 10.1007/s10585-015-9714-5] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 03/12/2015] [Indexed: 01/25/2023]
Abstract
Increased bone marrow adiposity is a common feature of advanced age, obesity and associated metabolic pathologies. Augmented numbers of marrow adipocytes positively correlate with dysregulated bone remodeling, also a well-established complication of metastatic disease. We have shown previously that marrow adiposity accelerates prostate tumor progression in the skeleton and promotes extensive destruction of the bone; however, the factors behind adipocyte-driven osteolysis in the skeletal tumor microenvironment are not currently known. In this study, utilizing in vivo diet-induced models of bone marrow adiposity, we reveal evidence for positive correlation between increased marrow fat content, bone degradation by ARCaP(M) and PC3 prostate tumors, and augmented levels of host-derived CXCL1 and CXCL2, ligands of CXCR2 receptor. We show by in vitro osteoclastogenesis assays that media conditioned by bone marrow adipocytes is a significant source of CXCL1 and CXCL2 proteins. We also demonstrate that both the adipocyte-conditioned media and the recombinant CXCL1 and CXCL2 ligands efficiently accelerate osteoclast maturation, a process that can be blocked by neutralizing antibodies to each of the chemokines. We further confirm the contribution of CXCR2 signaling axis to adiposity-driven osteoclastogenesis by blocking fat cell-induced osteoclast differentiation with CXCR2 antagonist or neutralizing antibodies. Together, our results link CXCL1 and CXCL2 chemokines with bone marrow adiposity and implicate CXCR2 signaling in promoting effects of marrow fat on progression of skeletal tumors in bone.
Collapse
Affiliation(s)
- Aimalie L Hardaway
- Department of Pharmacology, Wayne State University School of Medicine, 540 E. Canfield, Rm 6304, Detroit, MI, 48201, USA
| | | | | | | |
Collapse
|
25
|
Jung K, Lein M. Bone turnover markers in serum and urine as diagnostic, prognostic and monitoring biomarkers of bone metastasis. Biochim Biophys Acta Rev Cancer 2014; 1846:425-38. [PMID: 25220832 DOI: 10.1016/j.bbcan.2014.09.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 08/14/2014] [Accepted: 09/01/2014] [Indexed: 01/25/2023]
Abstract
Bone metastases are characterized by increased osteoblastic and/or osteolytic processes depending on the tumor type. The altogether destructive effect of metastasis formation promoted by increased metabolic activity raises the release of components from the osseous metabolism into the blood stream. These components are either enzymes directly involved in the alteration processes, metabolites/proteins that develop during this or bone matrix proteins released during this. These biomarkers are categorized in relation to their involvement in the bone formation or resorption as bone formation and resorption markers. Based on a PubMed literature search, a critical appraisal of the various biomarkers for diagnostic, prognostic, and monitoring purposes is given for patients with skeletal metastases caused by breast, prostate, lung, or renal cell carcinomas.
Collapse
Affiliation(s)
- Klaus Jung
- Department of Urology, University Hospital Charité, Berlin, Germany; Berlin Institute for Urologic Research, Berlin, Germany.
| | - Michael Lein
- Berlin Institute for Urologic Research, Berlin, Germany; Department of Urology, Sana Hospital Center, Offenbach, Germany
| |
Collapse
|
26
|
Emerging lung cancer therapeutic targets based on the pathogenesis of bone metastases. Int J Cell Biol 2014; 2014:236246. [PMID: 25197279 PMCID: PMC4147348 DOI: 10.1155/2014/236246] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 07/31/2014] [Accepted: 08/01/2014] [Indexed: 12/22/2022] Open
Abstract
Lung cancer is the second most common cancer and the leading cause of cancer related mortality in both men and women. Each year, more people die of lung cancer than of colon, breast, and prostate cancers combined. It is widely accepted that tumor metastasis is a formidable barrier to effective treatment of lung cancer. The bone is one of the frequent metastatic sites for lung cancer occurring in a large number of patients. Bone metastases can cause a wide range of symptoms that could impair quality of life of lung cancer patients and shorten their survival. We strongly believe that molecular targets (tumor-related and bone microenvironment based) that have been implicated in lung cancer bone metastases hold great promise in lung cancer therapeutics. Thus, this paper discusses some of the emerging molecular targets that have provided insights into the cascade of metastases in lung cancer with the focus on bone invasion. It is anticipated that the information gathered might be useful in future efforts of optimizing lung cancer treatment strategies.
Collapse
|
27
|
D'Amelio P, Roato I, Oderda M, Soria F, Zitella A, Ferracini R, Mengozzi G, Gontero P, Isaia GC. DKK-1 in prostate cancer diagnosis and follow up. BMC Clin Pathol 2014; 14:11. [PMID: 24655661 PMCID: PMC3976154 DOI: 10.1186/1472-6890-14-11] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 03/10/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Dickoppf-1 (DKK-1) is a negative regulator of bone formation with tumorigenic potential. The up-regulation of DKK-1 is an early event in prostate cancer (PCa) development, thus we investigated its role as a marker in the diagnosis and prognosis of PCa. METHODS We retrospectively enrolled 159 patients who underwent prostate biopsy, either for elevated PSA or suspect digital rectal examination, between 2003 and 2010. During the biopsy, one serum sample was collected from all patients; PSA and DKK-1 were measured by ELISA technique. Amongst the biopsy of 159 patients 75 were affected by PCa and 84 were not the mean period of follow-up for these patients was 5 years; a new biopsy was performed in case of PCa suspicion. RESULTS PSA performed better than DKK-1 in detecting PCa (0.63 vs 0.51 respectively). Differently from PSA DKK-1 was significantly higher in patients who developed PCa during follow-up than in cancer-free ones, thus DKK-1 performed better than PSA in detecting these patients (0.67 vs 0.55). DKK-1 was significantly lower in patients with bone metastases, whereas PSA was not significantly different in patients with different outcomes. CONCLUSIONS DKK-1 might be predictive for patients negative at first biopsy who will develop PCa and in the prognosis of bone metastases. It performed worse than PSA in the early diagnosis of Pca.
Collapse
Affiliation(s)
- Patrizia D'Amelio
- Gerontology Section, Department of Medical Sciences, University of Torino, Corso Bramante 88/90, 10126 Torino, Italy
| | - Ilaria Roato
- Center for Research in Experimental Medicine (CeRMS), Hospital City of Health and Science of Turin, Turin, Italy
| | - Marco Oderda
- Urology Section, Department of Surgical Science, Hospital City of Health and Science of Turin, University of Turin, Turin, Italy
| | - Francesco Soria
- Urology Section, Department of Surgical Science, Hospital City of Health and Science of Turin, University of Turin, Turin, Italy
| | - Andrea Zitella
- Urology Section, Department of Surgical Science, Hospital City of Health and Science of Turin, University of Turin, Turin, Italy
| | - Riccardo Ferracini
- Department of Orthopedics, Hospital City of Health and Science of Turin, Turin, Italy
| | - Giulio Mengozzi
- Baldi & Riberi Lab, Hospital City of Health and Science of Turin, Turin, Italy
| | - Paolo Gontero
- Urology Section, Department of Surgical Science, Hospital City of Health and Science of Turin, University of Turin, Turin, Italy
| | - Giovanni Carlo Isaia
- Gerontology Section, Department of Medical Sciences, University of Torino, Corso Bramante 88/90, 10126 Torino, Italy
| |
Collapse
|
28
|
Kovacic N, Croucher PI, McDonald MM. Signaling between tumor cells and the host bone marrow microenvironment. Calcif Tissue Int 2014; 94:125-39. [PMID: 24046000 DOI: 10.1007/s00223-013-9794-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 08/25/2013] [Indexed: 02/04/2023]
Abstract
Tumor cells with high skeletal homing affinity express numerous cell surface receptors that bind ligands produced in bone. Upon arrival, these cells survive in the host environment, encompassed in close proximity to bone marrow cells. Interactions between tumor cells and cells of the host microenvironment are essential to not only tumor cell survival but also their activation and proliferation into environment-modifying tumors. Through the production of RANKL, PTHrP, cytokines, and integrins, activated tumor cells stimulate osteoclastogenesis, enhance bone resorption, and subsequently release matrix-bound proteins that further promote tumor growth and bone resorption. In addition, alterations in the TGF-β/BMP and Wnt signaling pathways via tumor cell growth can either stimulate or suppress osteoblastic bone formation and function, leading to sclerotic or lytic bone disease, respectively. Hence, the presence of tumor cells in bone dysregulates bone remodeling, dramatically impairing skeletal integrity. Furthermore, through complex mechanisms, cells of the immune system interact with tumor cells to further impact bone remodeling. Lastly, with alterations in bone cell activity, the environment is permissive to promoting tumor growth further, suggesting an interdependence between tumor cells and bone cells in metastatic bone disease and multiple myeloma.
Collapse
Affiliation(s)
- Natasa Kovacic
- Bone Biology Group, Musculoskeletal Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia
| | | | | |
Collapse
|
29
|
The role of vascular endothelial growth factor in metastatic prostate cancer to the skeleton. Prostate Cancer 2013; 2013:418340. [PMID: 24396604 PMCID: PMC3874956 DOI: 10.1155/2013/418340] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 11/04/2013] [Accepted: 11/14/2013] [Indexed: 12/20/2022] Open
Abstract
Despite the clinical implication and high incidence of bone and spinal metastases, the molecular mechanisms behind prostate cancer metastasis to bone and spine are not well understood. In this review the molecular mechanisms that may contribute to the highly metastatic phenotype of prostate cancer are discussed. Proangiogenic factors such as vascular endothelial growth factor (VEGF) have been shown to not only aid in the metastatic capabilities of prostate cancer but also encourage the colonization and growth of prostate tumour cells in the skeleton. The importance of VEGF in the complex process of prostate cancer dissemination to the skeleton is discussed, including its role in the development of the bone premetastatic niche, metastatic tumour cell recognition of bone, and bone remodeling. The expression of VEGF has also been shown to be upregulated in prostate cancer and is associated with clinical stage, Gleason score, tumour stage, progression, metastasis, and survival. Due to the multifaceted effect VEGF has on tumour angiogenesis, tumour cell proliferation, and bone destruction, therapies targeting the VEGF pathways have shown promising clinical application and are being investigated in clinical trials.
Collapse
|
30
|
Begenik H, Kemik AS, Emre H, Dulger AC, Demirkiran D, Ebinc S, Kemik O. The association between serum Dickkopf-1 levels and esophageal squamous cell carcinoma. Hum Exp Toxicol 2013; 33:785-8. [PMID: 24220876 DOI: 10.1177/0960327113510537] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Serum Dickkopf-1(DKK-1) is elevated in many malignancies and is an important indicator of malignant potential. However, its significance in esophageal squamous cell cancer (ESCC) has not yet been clarified. We hypothesized a role for DKK-1 in patients with ESCC. The study consisted of 90 ESCC patients and 85 healthy controls. After diagnosis, the level of DKK-1 was measured in the serum samples by enzyme-linked immunosorbent assay and the levels of DKK-1 were much higher in the ESCC patients than in the healthy control group (p < 0.0001). For serum DKK-1, the sensitivity and specificity of the assay were 70 and 80%, respectively. The preoperative serum DKK-1 level was elevated in the ESCC patients. Although serum DKK-1 is not a specific biomarker for ESCC, it might be a useful marker for the diagnosis and treatment of ESCC.
Collapse
Affiliation(s)
- H Begenik
- Department of Internal Medicine, Medical Faculty, Yuzuncu Yil University, Van, Turkey
| | - A S Kemik
- Department of Biochemistry, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - H Emre
- Department of Internal Medicine, Medical Faculty, Yuzuncu Yil University, Van, Turkey
| | - A C Dulger
- Department of Gastroenterology, Medical Faculty, Yuzuncu Yil University, Van, Turkey
| | - D Demirkiran
- Department of Internal Medicine, Medical Faculty, Yuzuncu Yil University, Van, Turkey
| | - S Ebinc
- Department of Internal Medicine, Medical Faculty, Yuzuncu Yil University, Van, Turkey
| | - O Kemik
- Department of General Surgery, Medical Faculty, Yuzuncu Yil University, Van, Turkey
| |
Collapse
|
31
|
Inhibitory effect of metformin on bone metastasis of cancer via OPG/RANKL/RANK system. Med Hypotheses 2013; 81:805-6. [DOI: 10.1016/j.mehy.2013.08.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 08/14/2013] [Accepted: 08/23/2013] [Indexed: 02/05/2023]
|
32
|
|
33
|
Bi X, Sterling JA, Merkel AR, Perrien DS, Nyman JS, Mahadevan-Jansen A. Prostate cancer metastases alter bone mineral and matrix composition independent of effects on bone architecture in mice--a quantitative study using microCT and Raman spectroscopy. Bone 2013; 56:454-60. [PMID: 23867219 PMCID: PMC3799839 DOI: 10.1016/j.bone.2013.07.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Revised: 02/22/2013] [Accepted: 07/04/2013] [Indexed: 11/25/2022]
Abstract
Prostate cancer is the most common primary tumor and the second leading cause of cancer-related deaths in men in the United States. Prostate cancer bone metastases are characterized by abnormal bone remodeling processes and result in a variety of skeletal morbidities. Prevention of skeletal complications is a crucial element in prostate cancer management. This study investigated prostate cancer-induced alterations in the molecular composition and morphological structure of metastasis-bearing bones in a mouse model of prostate cancer using Raman spectroscopy and micro-computed tomography (microCT). LNCaP C4-2B prostate cancer cells were injected into the right tibiae of 5-week old male SCID mice. Upon sacrifice at 8weeks post tumor inoculation, two out of the ten tumor-bearing tibiae showed only osteoblastic lesions in the radiographs, 4 osteolytic lesions only and 4 mixed with osteoblastic and osteolytic lesions. Carbonate substitution was significantly increased while there was a marked reduction in the level of collagen mineralization, mineral crystallinity, and carbonate:matrix ratio in the cortex of the intact tumor-bearing tibiae compared to contralateral controls. MicroCT analysis revealed a significant reduction in bone volume/total volume, trabecular number and trabecular thickness, as well as significant increase in bone surface/volume ratio in tibiae with osteolytic lesions, suggesting active bone remodeling and bone loss. None of the changes in bone compositional properties were correlated with lesion area from radiographs or the changes in bone architecture from microCT. This study indicates that LNCaP C4-2B prostate cancer metastases alter bone tissue composition independent of changes in architecture, and altered bone quality may be an important contributor to fracture risk in these patients. Raman spectroscopy may provide a new avenue of investigation into interactions between tumor and bone microenvironment.
Collapse
Affiliation(s)
- Xiaohong Bi
- Department of Biomedical Engineering, Vanderbilt University, VU Station B#351631, 2301 Vanderbilt Place, Nashville, TN 37235, USA.
| | | | | | | | | | | |
Collapse
|
34
|
|
35
|
Larson S, Zhang X, Dumpit R, Coleman I, Lakely B, Roudier M, Higano C, True LD, Lange PH, Montgomery B, Corey E, Nelson PS, Vessella RL, Morrissey C. Characterization of osteoblastic and osteolytic proteins in prostate cancer bone metastases. Prostate 2013; 73:932-40. [PMID: 23334979 PMCID: PMC4214278 DOI: 10.1002/pros.22639] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 12/12/2012] [Indexed: 01/30/2023]
Abstract
BACKGROUND Approximately 90% of patients who die of Prostate Cancer (PCa) have bone metastases, which promote a spectrum of osteoblastic, osteolytic or mixed bone responses. Numerous secreted proteins have been reported to promote osteoblastic or osteolytic bone responses. We determined whether previously identified and/or novel proteins were associated with the osteoblastic or osteolytic response in clinical specimens of PCa bone metastases. METHODS Gene expression was analyzed on 14 PCa metastases from 11 patients by microarray profiling and qRT-PCR, and protein expression was analyzed on 33 PCa metastases from 30 patients by immunohistochemistry on highly osteoblastic and highly osteolytic bone specimens. RESULTS Transcript and protein levels of BMP-2, BMP-7, DKK-1, ET-1, and Sclerostin were not significantly different between osteoblastic and osteolytic metastases. However, levels of OPG, PGK1, and Substance P proteins were increased in osteoblastic samples. In addition, Emu1, MMP-12, and sFRP-1 were proteins identified with a novel role of being associated with either the osteoblastic or osteolytic bone response. CONCLUSIONS This is the first detailed analysis of bone remodeling proteins in human specimens of PCa bone metastases. Three proteins not previously shown to be involved may have a role in the PCa bone response. Furthermore, our data suggests that the relative expression of numerous, rather than a single, bone remodeling proteins determine the bone response in PCa bone metastases.
Collapse
Affiliation(s)
- Sandy Larson
- Department of Urology, University of Washington, Seattle, WA
| | - Xiaotun Zhang
- Department of Urology, University of Washington, Seattle, WA
| | - Ruth Dumpit
- Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Ilsa Coleman
- Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Bryce Lakely
- Department of Urology, University of Washington, Seattle, WA
| | - Martine Roudier
- Department of Pathology, University of Washington, Seattle, WA
| | - Celestia Higano
- Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | | | - Paul H. Lange
- Department of Urology, University of Washington, Seattle, WA
- Department of Veterans Affairs Medical Center, Seattle, WA
| | | | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA
| | - Peter S. Nelson
- Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Robert L. Vessella
- Department of Urology, University of Washington, Seattle, WA
- Department of Veterans Affairs Medical Center, Seattle, WA
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA
| |
Collapse
|
36
|
Interaction among cells of bone, immune system, and solid tumors leads to bone metastases. Clin Dev Immunol 2013; 2013:315024. [PMID: 23710201 PMCID: PMC3655477 DOI: 10.1155/2013/315024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 04/08/2013] [Indexed: 01/05/2023]
Abstract
Bone metastases are a dismal consequence for different types of solid tumors, such as breast, prostate, lung, and kidney cancer. The mechanisms regulating the interactions among bone, immune system, and tumor cells have been deeply investigated, and many studies are ongoing to define the specific role of the different cells in the bone metastatic process. The affinity of some tumors to growth in bone results from the special microenvironment provided by bone. Moreover, immune system and bone have a bidirectional relationship: bone cells express surface molecules ruling the expansion of hemopoietic stem cells from which all cells of the mammalian immune system derive, and various immunoregulatory cytokines influence the fate of bone cells. The last findings allow to extend the concept of vicious cycle and add T cells as mediators of the tumor growth in bone.
Collapse
|
37
|
Cody JJ, Rivera AA, Lyons GR, Yang SW, Wang M, Ashley JW, Meleth S, Feng X, Siegal GP, Douglas JT. Expression of osteoprotegerin from a replicating adenovirus inhibits the progression of prostate cancer bone metastases in a murine model. J Transl Med 2013; 93:268-78. [PMID: 23358109 PMCID: PMC3584184 DOI: 10.1038/labinvest.2012.179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Metastatic involvement of the skeleton is a frequent consequence of advanced prostate cancer. These skeletal metastases cause a number of debilitating complications and are refractory to current treatments. New therapeutic options are being explored, including conditionally replicating adenoviruses (CRAds). CRAds are engineered to selectively replicate in and destroy tumor cells and can be 'armed' with exogenous transgenes for enhanced potency. We hypothesized that a CRAd armed with osteoprotegerin (OPG), an inhibitor of osteoclastogenesis, would inhibit the progression of prostate cancer bone metastases by directly lysing tumor cells and by reducing osteoclast activity. Although prostate cancer bone metastases are predominantly osteoblastic in nature, increased osteoclast activity is critical for the growth of these lesions. Ad5-Δ24-sOPG-Fc-RGD is a CRAd that carries a fusion of the ligand-binding domains of OPG and the Fc region of human IgG1 in place of the viral E3B genes. To circumvent low tumor cell expression of the native adenoviral receptor, an arginine-glycine-aspartic acid (RGD) peptide insertion within the viral fiber knob allows infection of cells expressing α(v) integrins. A 24-base pair deletion (Δ24) within viral E1A limits replication to cells with aberrant retinoblastoma cell cycle regulator/tumor suppressor expression. We have confirmed that Ad5-Δ24-sOPG-Fc-RGD replicates within and destroys prostate cancer cells and, in both murine and human coculture models, that infection of prostate cancer cells inhibits osteoclastogenesis in vitro. In a murine model, progression of advanced prostate cancer bone metastases was inhibited by treatment with Ad5-Δ24-sOPG-Fc-RGD but not by an unarmed control CRAd.
Collapse
Affiliation(s)
- James J. Cody
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Angel A. Rivera
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology and Surgery, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gray R. Lyons
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sherry W. Yang
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ming Wang
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology and Surgery, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jason W. Ashley
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sreelatha Meleth
- Division of Preventive Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Xu Feng
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gene P. Siegal
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA,The Center for Metabolic Bone Disease Core Laboratory, The University of Alabama at Birmingham, Birmingham, AL, USA,The Gene Therapy Center, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Joanne T. Douglas
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology and Surgery, The University of Alabama at Birmingham, Birmingham, AL, USA,The Gene Therapy Center, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
38
|
Brunetti G, Faienza MF, Piacente L, Ventura A, Oranger A, Carbone C, Benedetto AD, Colaianni G, Gigante M, Mori G, Gesualdo L, Colucci S, Cavallo L, Grano M. High dickkopf-1 levels in sera and leukocytes from children with 21-hydroxylase deficiency on chronic glucocorticoid treatment. Am J Physiol Endocrinol Metab 2013; 304:E546-54. [PMID: 23299503 DOI: 10.1152/ajpendo.00535.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Children with 21-hydroxylase deficiency (21-OHD) need chronic glucocorticoid (cGC) therapy to replace congenital deficit of cortisol synthesis, and this therapy is the most frequent and severe form of drug-induced osteoporosis. In this study, we enrolled 18 patients (9 females) and 18 sex- and age-matched controls. We found in 21-OHD patients high serum and leukocyte levels of dickkopf-1 (DKK1), a secreted antagonist of the Wnt/β-catenin signaling pathway known to be a key regulator of bone mass. In particular, we demonstrated by flow cytometry, confocal microscopy, and real-time PCR that monocytes, T lymphocytes, and neutrophils from patients expressed high levels of DKK1, which may be related to the cGC therapy. In fact, we showed that dexamethasone treatment markedly induced the expression of DKK1 in a dose- and time-dependent manner in leukocytes. The serum from patients containing elevated levels of DKK1 can directly inhibit in vitro osteoblast differentiation and receptor activator of NF-κB ligand (RANKL) expression. We also found a correlation between both DKK1 and RANKL or COOH-terminal telopeptides of type I collagen (CTX) serum levels in 21-OHD patients on cGC treatment. Our data indicated that DKK1, produced by leukocytes, may contribute to the alteration of bone remodeling in 21-OHD patients on cGC treatment.
Collapse
Affiliation(s)
- Giacomina Brunetti
- Dept. of Basic Medical Sciences, Neuroscience, and Sense Organs, Section of Human Anatomy and Histology, Univ. of Bari, Piazza Giulio Cesare, 11 70124 Bari, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
D'Amico L, Satolli MA, Mecca C, Castiglione A, Ceccarelli M, D'Amelio P, Garino M, De Giuli M, Sandrucci S, Ferracini R, Roato I. Bone metastases in gastric cancer follow a RANKL-independent mechanism. Oncol Rep 2013; 29:1453-8. [PMID: 23404437 DOI: 10.3892/or.2013.2280] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 12/27/2012] [Indexed: 01/06/2023] Open
Abstract
Gastric cancer is one of the most common and lethal malignancies worldwide. Bone metastases in gastric cancer are less common than in other solid tumors, but when they occur the prognosis is generally poor. Increased osteoclastogenesis and osteoclast activity are common features in bone metastases caused by different osteotropic cancer. We investigated osteoclastogenesis and its mechanisms in gastric cancer by enrolling 31 newly diagnosed gastric cancer patients and 45 healthy controls. We studied in vitro osteoclastogenesis in the peripheral blood mononuclear cell cultures of patients and controls, showing spontaneous osteoclastogenesis for half of the patients. This osteoclastogenesis was RANKL- and TNF-α-independent. We analyzed primary tumor and bone metastatic tissues of gastric cancer for the expression of genes involved in osteoclastogenesis. The expression of transforming growth factor-β (TGF-β), osteoprotegerin (OPG), IL-7 and dickkopf-1 (DKK-1) was higher in primary tumors than in bone metastases. RANKL was not detectable in primary tumor or in bone metastatic tissue. The serum RANKL level was significantly higher in healthy controls than in patients, and it was not related to osteoclastogenesis, thereby suggesting that RANKL is not involved in the bone metastatic mechanisms in gastric cancer. We hypothesized a role of RANKL in angiogenesis, thus we compared the serum levels of RANKL to those of VEGF, since VEGF is directly related to angiogenesis. Different from RANKL, the VEGF serum levels were higher in gastric patients than in controls, suggesting a block of the angiogenesis inhibition due to RANKL. RANKL and VEGF serum levels were not predictive of overall survival in our cohort of gastric patients.
Collapse
Affiliation(s)
- Lucia D'Amico
- CeRMS (Center for Experimental Research and Medical Studies), Department of Medical Oncology, San Giovanni Battista Hospital, and Department of Surgical and Medical Discplines, University of Turin, Turin, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Hadji P, Aapro M, Costa L, Gnant M. Antiresorptive treatment options and bone health in cancer patients—safety profiles and clinical considerations. Cancer Treat Rev 2012; 38:815-24. [DOI: 10.1016/j.ctrv.2012.03.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 03/02/2012] [Accepted: 03/07/2012] [Indexed: 11/29/2022]
|
41
|
Revannasiddaiah S, Rastogi M, Thakur P, Gupta MK, Sood A, Sharma C. MRI for the detection of prostate cancer origin vertebral metastases in the preosteoblastic phase. BMJ Case Rep 2012; 2012:bcr2012006265. [PMID: 22778472 PMCID: PMC4543210 DOI: 10.1136/bcr-2012-006265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
This report describes the case of a gentleman aged 59 years presenting with low-back pain, who had underwent radical prostatectomy for prostate cancer 8 years ago. On evaluation, a slightly elevated serum alkaline-phosphatase level prompted a search for bone metastases. Although x-ray radiography and a bone scan were apparently normal, an MRI scan revealed the presence of metastatic marrow infiltration in the lumbar vertebrae. The patient subsequently was initiated on therapy with androgen-deprivation therapy and bisphosphonates, and currently enjoys symptom-free and progression-free survival. The images in this paper intend to impress upon the limitations of bone scan and x-ray radiography with regard to the detection of vertebral marrow infiltration in the absence of cortical bone invasion. In addition, a brief review of the pathophysiology of vertebral metastases arising from prostate cancer is included.
Collapse
Affiliation(s)
- Swaroop Revannasiddaiah
- Department of Radiotherapy & Oncology, Regional Cancer Centre, Indira Gandhi Medical College, Shimla, Himachal Pradesh, India.
| | | | | | | | | | | |
Collapse
|
42
|
Owen HC, Vanhees I, Solie L, Roberts SJ, Wauters A, Luyten FP, Van Cromphaut S, Van den Berghe G. Critical illness-related bone loss is associated with osteoclastic and angiogenic abnormalities. J Bone Miner Res 2012; 27:1541-52. [PMID: 22461003 DOI: 10.1002/jbmr.1612] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Critically ill patients are at increased risk of fractures during rehabilitation, and can experience impaired healing of traumatic and surgical bone fractures. In addition, markers of bone resorption are markedly increased in critically ill patients, while markers of bone formation are decreased. In the current study, we have directly investigated the effect of critical illness on bone metabolism and repair. In a human in vitro model of critical illness, Fluorescence-activated cell sorting (FACS) analysis revealed an increase in circulating CD14+/CD11b+ osteoclast precursors in critically ill patient peripheral blood compared to healthy controls. In addition, the formation of osteoclasts was increased in patient peripheral blood mononuclear cell (PBMC) cultures compared to healthy controls, both in the presence and absence of osteoclastogenic factors receptor activator of NF-κB ligand (RANKL) and macrophage colony-stimulating factor (M-CSF). Culturing PBMCs with 10% critically ill patient serum further increased osteoclast formation and activity in patient PBMCs only, and neutralization studies revealed that immunoglobulin G (IgG) antibody signaling through the immunoreceptor Fc receptor common γ chain III (FcRγIII) played an important role. When analyzing bone formation, no differences in osteogenic differentiation were observed using human periosteal-derived cells (hPDCs) treated with patient serum in vitro, but a decrease in the expression of vascular endothelial growth factor receptor 1 (VEGF-R1) suggested impaired vascularization. This was confirmed using serum-treated hPDCs implanted onto calcium phosphate scaffolds in a murine in vivo model of bone formation, where decreased vascularization and increased osteoclast activity led to a decrease in bone formation in scaffolds with patient serum-treated hPDCs. Together, these findings may help to define novel therapeutic targets to prevent bone loss and optimize fracture healing in critically ill patients.
Collapse
Affiliation(s)
- Helen C Owen
- Department and Laboratory of Intensive Care Medicine, Katholieke Universiteit Leuven, Leuven, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Drudge-Coates L, Turner B. Bone micro-environment in prostate cancer: impact, treatment and implications for nursing practice. INTERNATIONAL JOURNAL OF UROLOGICAL NURSING 2012. [DOI: 10.1111/j.1749-771x.2012.01148.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
44
|
Feller L, Kramer B, Lemmer J. Pathobiology of cancer metastasis: a short account. Cancer Cell Int 2012; 12:24. [PMID: 22676510 PMCID: PMC3407798 DOI: 10.1186/1475-2867-12-24] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 06/07/2012] [Indexed: 01/18/2023] Open
Abstract
Cancer-initiating cells display aberrant functional and phenotypic characteristics of normal stem cells from which they evolved by accumulation of multiple cytogenetic and/or epigenetic alterations. Signal transduction pathways which are essential for normal stem cell function are abnormally expressed by cancer cells, with a cancer cell phenotype playing an essential role in cancerization and metastasis.Local tumour progression, metastasis and metastatic tumour growth are mediated by direct cell-to-cell and paracrine reciprocal interactions between cancer cells and various stromal cells including fibroblasts, macrophages, bone marrow derived stem cells and progenitor cells. These interactions mediate breakdown of basement membrane barriers and angiogenesis both locally at the invasive front of the primary tumour and at the distant metastatic site; attract primary tumour cells to the candidate metastatic site; and promote proliferation, survival and growth of primary tumour cells and of metastatic cells at their distant site.It is the purpose of this article to highlight the analogies between some of the genetic programs of normal stem cells, and of cancer cells participating in the process of metastasis.
Collapse
Affiliation(s)
- Liviu Feller
- Department of Periodontology and Oral Medicine, School of Oral Health Sciences, Faculty of Health Sciences, University of Limpopo, Medunsa Campus, Medunsa, 0204, South Africa.
| | | | | |
Collapse
|
45
|
Abstract
Bone marrow macrophages (BMMs) share common progenitors with osteoclasts and are critical components of bone-tumor microenvironment; however, their function in prostate tumor growth in the skeleton has not been explored. BMMs are the major source of inflammatory factors and proteases, including cysteine protease cathepsin K (CTSK). In this study, utilizing mice deficient in CTSK, we demonstrate the critical involvement of this potent collagenase in tumor progression in bone. We present the evidence that tumor growth and progression in the bone are impaired in the absence of CTSK. Most importantly, we show for the first time that BMM-supplied CTSK may be involved in CCL2- and COX-2-driven pathways that contribute to tumor progression in bone. Together, our data unravel novel roles for CTSK in macrophage-regulated processes, and provide evidence for close interplay between inflammatory, osteolytic and tumor cell-driven events in the bone-tumor microenvironment.
Collapse
|
46
|
Li Y, Kong D, Ahmad A, Bao B, Sarkar FH. Targeting bone remodeling by isoflavone and 3,3'-diindolylmethane in the context of prostate cancer bone metastasis. PLoS One 2012; 7:e33011. [PMID: 22412975 PMCID: PMC3296768 DOI: 10.1371/journal.pone.0033011] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 02/02/2012] [Indexed: 12/20/2022] Open
Abstract
Prostate cancer (PCa) bone metastases have long been believed to be osteoblastic because of bone remodeling leading to the formation of new bone. However, recent studies have shown increased osteolytic activity in the beginning stages of PCa bone metastases, suggesting that targeting both osteolytic and osteoblastic mediators would likely inhibit bone remodeling and PCa bone metastasis. In this study, we found that PCa cells could stimulate differentiation of osteoclasts and osteoblasts through the up-regulation of RANKL, RUNX2 and osteopontin, promoting bone remodeling. Interestingly, we found that formulated isoflavone and 3,3′-diindolylmethane (BR-DIM) were able to inhibit the differentiation of osteoclasts and osteoblasts through the inhibition of cell signal transduction in RANKL, osteoblastic, and PCa cell signaling. Moreover, we found that isoflavone and BR-DIM down-regulated the expression of miR-92a, which is known to be associated with RANKL signaling, EMT and cancer progression. By pathway and network analysis, we also observed the regulatory effects of isoflavone and BR-DIM on multiple signaling pathways such as AR/PSA, NKX3-1/Akt/p27, MITF, etc. Therefore, isoflavone and BR-DIM with their multi-targeted effects could be useful for the prevention of PCa progression, especially by attenuating bone metastasis mechanisms.
Collapse
Affiliation(s)
- Yiwei Li
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Dejuan Kong
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Aamir Ahmad
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Bin Bao
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Fazlul H. Sarkar
- Department of Pathology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- * E-mail:
| |
Collapse
|
47
|
He W, Cronstein BN. Adenosine A1 receptor regulates osteoclast formation by altering TRAF6/TAK1 signaling. Purinergic Signal 2012; 8:327-37. [PMID: 22311477 DOI: 10.1007/s11302-012-9292-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 01/19/2012] [Indexed: 11/27/2022] Open
Abstract
Adenosine is an endogenous nucleoside that modulates many physiological processes through four receptor subtypes (A(1), A(2a), A(2b), A(3)). Previous work from our laboratory has uncovered a critical role for adenosine A(1) receptor (A(1) R) in osteoclastogenesis both in vivo and in vitro. Our current work focuses on understanding the details of how A(1) R modulates the receptor activator of NF-κB ligand (RANKL)-induced signaling in osteoclastogenesis. Osteoclasts were generated from mouse bone marrow precursors in the presence of RANKL and macrophage-colony stimulating factor. A pharmacological antagonist of A(1) R (DPCPX) inhibited RANKL-induced osteoclast differentiation, including osteoclast-specific genes (Acp5, MMP9, β(3) Integrin, α(v) Integrin, and CTSK) and osteoclast-specific transcription factors such as c-fos and nuclear factor of activated T cells cytoplasmic 1 (NFATc1) expression in a dose-dependent manner. DPCPX also inhibited RANKL-induced activation of NF-κB and JNK/c-Jun but had little effect on other mitogen-activated protein kinases (p38 and Erk). Finally, immunoprecipitation analysis showed that blockade of A(1)R resulted in disruption of the association of tumor necrosis factor receptor-associated factor 6 (TRAF6) and transforming growth factor-β-activated kinase 1 (TAK1), a signaling event that is important for activation of NF-κB and JNK, suggesting the participation of adenosine/A(1)R in early signaling of RANKL. Collectively, these data demonstrated an important role of adenosine, through A(1)R in RANKL-induced osteoclastogenesis.
Collapse
Affiliation(s)
- W He
- New York University School of Medicine, New York, NY 10016, USA
| | | |
Collapse
|
48
|
Park SI, Soki FN, McCauley LK. Roles of bone marrow cells in skeletal metastases: no longer bystanders. CANCER MICROENVIRONMENT : OFFICIAL JOURNAL OF THE INTERNATIONAL CANCER MICROENVIRONMENT SOCIETY 2011; 4:237-46. [PMID: 21809058 PMCID: PMC3234319 DOI: 10.1007/s12307-011-0081-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Accepted: 07/20/2011] [Indexed: 01/09/2023]
Abstract
Bone serves one of the most congenial metastatic microenvironments for multiple types of solid tumors, but its role in this process remains under-explored. Among many cell populations constituting the bone and bone marrow microenvironment, osteoblasts (originated from mesenchymal stem cells) and osteoclasts (originated from hematopoietic stem cells) have been the main research focus for pro-tumorigenic roles. Recently, increasing evidence further elucidates that hematopoietic lineage cells as well as stromal cells in the bone marrow mediate distinct but critical functions in tumor growth, metastasis, angiogenesis and apoptosis in the bone microenvironment. This review article summarizes the key evidence describing differential roles of bone marrow cells, including hematopoietic stem cells (HSCs), megakaryocytes, macrophages and myeloid-derived suppressor cells in the development of metastatic bone lesions. HSCs promote tumor growth by switching on angiogenesis, but at the same time compete with metastatic tumor cells for occupancy of osteoblastic niche. Megakaryocytes negatively regulate the extravasating tumor cells by inducing apoptosis and suppressing proliferation. Macrophages and myeloid cells have pro-tumorigenic roles in general, suggesting a similar effect in the bone marrow. Hematopoietic and stromal cell populations in the bone marrow, previously considered as simple by-standers in the context of tumor metastasis, have distinct and active roles in promoting or suppressing tumor growth and metastasis in bone. Further investigation on the extended roles of bone marrow cells will help formulate better approaches to treatment through improved understanding of the metastatic bone microenvironment.
Collapse
Affiliation(s)
- Serk In Park
- Department of Periodontics and Oral Medicine, The University of Michigan School of Dentistry, 1011 N. University Avenue, Ann Arbor, MI 48109 USA
| | - Fabiana N. Soki
- Department of Periodontics and Oral Medicine, The University of Michigan School of Dentistry, 1011 N. University Avenue, Ann Arbor, MI 48109 USA
| | - Laurie K. McCauley
- Department of Periodontics and Oral Medicine, The University of Michigan School of Dentistry, 1011 N. University Avenue, Ann Arbor, MI 48109 USA
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, MI USA
| |
Collapse
|
49
|
D'Amelio P, Roato I, D'Amico L, Veneziano L, Suman E, Sassi F, Bisignano G, Ferracini R, Gargiulo G, Castoldi F, Pescarmona GP, Isaia GC. Bone and bone marrow pro-osteoclastogenic cytokines are up-regulated in osteoporosis fragility fractures. Osteoporos Int 2011; 22:2869-77. [PMID: 21116815 DOI: 10.1007/s00198-010-1496-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Accepted: 10/29/2010] [Indexed: 12/21/2022]
Abstract
UNLABELLED This study evaluates cytokines production in bone and bone marrow of patients with an osteoporotic fracture or with osteoarthritis by real time PCR, Western blot and immunohistochemistry. We demonstrate that the cytokine pattern is shifted towards osteoclast activation and osteoblast inhibition in patients with osteoporotic fractures. INTRODUCTION Fragility fractures are the resultant of low bone mass and poor bone architecture typical of osteoporosis. Cytokines involved in the control of bone cell maturation and function are produced by both bone itself and bone marrow cells, but the roles of these two sources in its control and the amounts they produce are not clear. This study compares their production in patients with an osteoporotic fracture and those with osteoarthritis. METHODS We evaluated 52 femoral heads from women subjected to hip-joint replacement surgery for femoral neck fractures due to low-energy trauma (37), or for osteoarthritis (15). Total RNA was extracted from both bone and bone marrow, and quantitative PCR was used to identify the receptor activator of nuclear factor kB Ligand (RANKL), osteoprotegerin (OPG), macrophage colony stimulating factor (M-CSF), transforming growth factor β (TGFβ), Dickoppf-1 (DKK-1) and sclerostin (SOST) expression. Immunohistochemistry and Western blot were performed in order to quantify and localize in bone and bone marrow the cytokines. RESULTS We found an increase of RANKL/OPG ratio, M-CSF, SOST and DKK-1 in fractured patients, whereas TGFβ was increased in osteoarthritic bone. Bone marrow produced greater amounts of RANKL, M-CSF and TGFβ compared to bone, whereas the production of DKK-1 and SOST was higher in bone. CONCLUSIONS We show that bone marrow cells produced the greater amount of pro-osteoclastogenic cytokines, whereas bone cells produced higher amount of osteoblast inhibitors in patients with fragility fracture, thus the cytokine pattern is shifted towards osteoclast activation and osteoblast inhibition in these patients.
Collapse
Affiliation(s)
- P D'Amelio
- Gerontology Section, Department of Surgical and Medical Disciplines, University of Torino, Corso Bramante 88/90, Torino, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Gadolinium inhibits prostate cancer PC3 cell migration and suppresses osteoclast differentiationin vitro. Cell Biol Int 2011; 35:1159-67. [DOI: 10.1042/cbi20100870] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|