1
|
Mesny E, Martz N, Stacoffe N, Clarençon F, Louis M, Mansouri N, Sirveaux F, Thureau S, Faivre JC. State-of-the-art of multidisciplinary approach of bone metastasis-directed therapy: review and challenging questions for preparation of a GEMO practice guidelines. Cancer Metastasis Rev 2025; 44:45. [PMID: 40220136 PMCID: PMC11993453 DOI: 10.1007/s10555-025-10262-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
Bone is a common secondary site of dissemination during the course of cancer. Bone metastases (BM) can be associated with skeletal-related events (SRE) such as disabling pain, hypercalcemia, and bone instability that leads to pathological fractures or spinal cord compression. SRE contribute to high morbidity as well as, mortality, and have a negative economic impact. Modern management of BM integrates focal treatments (such as radiotherapy, surgery, and interventional radiology), orthoses, and antiresorptive and systemic oncological treatment. The choice of a metastasis-directed therapy depends on the objective of the treatment, the patient characteristics, and the complete assessment of the bone lesion (pain, neurological risk, and instability). In the narrative review present herein, we aim to provide an updated summary of the literature, with description of the advantages and disadvantages of current and emerging strategies in the multimodal treatment of BM and, based on these data, an updated algorithm for the management of BM.
Collapse
Affiliation(s)
- Emmanuel Mesny
- Radiation Oncology Department, Hospices Civils de Lyon, CHLS, Lyon, France.
| | - Nicolas Martz
- Radiation Oncology Department, Institut de Cancérologie de Lorraine-Alexis-Vautrin, Vandœuvre-Lès-Nancy, France
| | - Nicolas Stacoffe
- Radiology Department, Hospices Civils de Lyon, CHLS, Lyon, France
| | - Frédéric Clarençon
- Department of Interventional Neuroradiology, AP-HP La Pitié-Salpêtrière, Paris, France
| | | | | | | | - Sébastien Thureau
- Radiation Oncology Department and Litis Quantif, EA, 4108 Unity, Centre Henri Becquerel, Rouen, France
| | - Jean-Christophe Faivre
- Radiation Oncology Department, Institut de Cancérologie de Lorraine-Alexis-Vautrin, Vandœuvre-Lès-Nancy, France
| |
Collapse
|
2
|
Aljagthmi AA, Abdel-Aziz AK. Hematopoietic stem cells: Understanding the mechanisms to unleash the therapeutic potential of hematopoietic stem cell transplantation. Stem Cell Res Ther 2025; 16:60. [PMID: 39924510 PMCID: PMC11809095 DOI: 10.1186/s13287-024-04126-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/21/2024] [Indexed: 02/11/2025] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a promising approach in regenerative medicine and serves as a standard treatment for different malignant and non-malignant conditions. Despite its widespread applications, HSCT is associated with various complications that compromise patients' lives and pose considerable risks of morbidity and mortality. Understanding the molecular physiology of HSCs is fundamental to ultimately enhance the mobilization, engraftment and differentiation of HSCs, thus unleashing the full therapeutic potential of HSCT in the treated patients. This review outlines the current understanding of HSC biology and its relevance to the clinical challenges associated with HSCT. Furthermore, we critically discuss the pros and cons of the preclinical murine models exploited in the HSCT field. Understanding the molecular physiology of HSCs will ultimately unleash the full therapeutic potential of HSCT. HSCs derived from induced pluripotent stem cells (iPSCs) might present an attractive tool which could be exploited preclinically and clinically. Nonetheless, further studies are warranted to systematically evaluate their potential in terms of improving the therapeutic outcome and minimizing the adverse effects of HSCT.
Collapse
Affiliation(s)
- Amjad Ahmed Aljagthmi
- Research center, King Faisal Specialist Hospital and Research Centre, Jeddah, 21499, Kingdom of Saudi Arabia.
| | - Amal Kamal Abdel-Aziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| |
Collapse
|
3
|
Zhu N, Ni H, Guo S, Shen YQ, Chen Q. Bone complications of cancer treatment. Cancer Treat Rev 2024; 130:102828. [PMID: 39270364 DOI: 10.1016/j.ctrv.2024.102828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024]
Abstract
With the advancements in conventional treatment modalities such as radiation, chemotherapy, and surgery, as well as the emergence of immunotherapy, the overall cure rate for solid tumor malignancies has experienced a significant increase. However, it is unfortunate that exposure to cancer treatments can have detrimental effects on the function of osteoblasts and osteoclasts, disturbing bone metabolic homeostasis in patients, as well as causing damage to bone marrow cells and other bone tissues. Consequently, certain tumor treatment options may pose a risk for subsequent bone diseases. Common bone disorders associated with cancer treatment include osteonecrosis, bone loss, and secondary bone tumors. (1)Cancer treatment-related osteonecrosis is primarily linked to the use of radiation therapy and certain chemicals, such as bisphosphonates, denosumab, antiangiogenic agents, and immunomodulators. It has been observed that high-dose radiation therapy is more likely to result in osteonecrosis. (2)Chemicals and hormones, particularly sex hormones, glucocorticoids, and thyroid hormones or thyrotropic hormones, are among the factors that can contribute to cancer treatment-related bone loss. (3)Secondary bone tumors differ from metastases originating from primary tumors, and radiotherapy plays a significant role in their development, while chemotherapy may also exert some influence. Radiogenic secondary bone tumors are predominantly malignant, with osteosarcoma being the most common type. Chemotherapy may be a risk factor for the relatively rare occurrence of secondary Ewing sarcoma of the bone. These treatment-related bone disorders have a considerable adverse impact on the prognosis of cancer patients. Hence, it is imperative to prioritize the bone health of patients undergoing cancer treatment and give it further attention.
Collapse
Affiliation(s)
- Nanxi Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Hao Ni
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shengzhao Guo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ying-Qiang Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
4
|
Landspersky T, Stein M, Saçma M, Geuder J, Braitsch K, Rivière J, Hettler F, Romero Marquez S, Vilne B, Hameister E, Richter D, Schönhals E, Tuckermann J, Verbeek M, Herhaus P, Hecker JS, Bassermann F, Götze KS, Enard W, Geiger H, Oostendorp RAJ, Schreck C. Targeting CDC42 reduces skeletal degeneration after hematopoietic stem cell transplantation. Blood Adv 2024; 8:5400-5414. [PMID: 39159429 PMCID: PMC11526086 DOI: 10.1182/bloodadvances.2024012879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/13/2024] [Accepted: 06/16/2024] [Indexed: 08/21/2024] Open
Abstract
ABSTRACT Osteopenia and osteoporosis are common long-term complications of the cytotoxic conditioning regimen for hematopoietic stem cell transplantation (HSCT). We examined mesenchymal stem and progenitor cells (MSPCs), which include skeletal progenitors, from mice undergoing HSCT. Such MSPCs showed reduced fibroblastic colony-forming units frequency, increased DNA damage, and enhanced occurrence of cellular senescence, whereas there was a reduced bone volume in animals that underwent HSCT. This reduced MSPC function correlated with elevated activation of the small Rho guanosine triphosphate hydrolase CDC42, disorganized F-actin distribution, mitochondrial abnormalities, and impaired mitophagy in MSPCs. Changes and defects similar to those in mice were also observed in MSPCs from humans undergoing HSCT. A pharmacological treatment that attenuated the elevated activation of CDC42 restored F-actin fiber alignment, mitochondrial function, and mitophagy in MSPCs in vitro. Finally, targeting CDC42 activity in vivo in animals undergoing transplants improved MSPC quality to increase both bone volume and trabecular bone thickness. Our study shows that attenuation of CDC42 activity is sufficient to attenuate reduced function of MSPCs in a BM transplant setting.
Collapse
Affiliation(s)
- Theresa Landspersky
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Merle Stein
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Mehmet Saçma
- Institute of Molecular Medicine, Stem Cells, and Aging, Ulm University, Ulm, Germany
| | - Johanna Geuder
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians University, Munich, Germany
| | - Krischan Braitsch
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Jennifer Rivière
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Franziska Hettler
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Sandra Romero Marquez
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Baiba Vilne
- Bioinformatics Laboratory, Rīga Stradiņš University, Riga, Lettland
| | - Erik Hameister
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Daniel Richter
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Emely Schönhals
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | - Mareike Verbeek
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Peter Herhaus
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Judith S. Hecker
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Florian Bassermann
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Katharina S. Götze
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians University, Munich, Germany
| | - Hartmut Geiger
- Institute of Molecular Medicine, Stem Cells, and Aging, Ulm University, Ulm, Germany
| | - Robert A. J. Oostendorp
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| | - Christina Schreck
- School of Medicine, Department of internal Medicine III, Technical University of Munich, Munich, Germany
| |
Collapse
|
5
|
Ebeling PR, Nguyen HH, Aleksova J, Vincent AJ, Wong P, Milat F. Secondary Osteoporosis. Endocr Rev 2022; 43:240-313. [PMID: 34476488 DOI: 10.1210/endrev/bnab028] [Citation(s) in RCA: 139] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Indexed: 02/07/2023]
Abstract
Osteoporosis is a global public health problem, with fractures contributing to significant morbidity and mortality. Although postmenopausal osteoporosis is most common, up to 30% of postmenopausal women, > 50% of premenopausal women, and between 50% and 80% of men have secondary osteoporosis. Exclusion of secondary causes is important, as treatment of such patients often commences by treating the underlying condition. These are varied but often neglected, ranging from endocrine to chronic inflammatory and genetic conditions. General screening is recommended for all patients with osteoporosis, with advanced investigations reserved for premenopausal women and men aged < 50 years, for older patients in whom classical risk factors for osteoporosis are absent, and for all patients with the lowest bone mass (Z-score ≤ -2). The response of secondary osteoporosis to conventional anti-osteoporosis therapy may be inadequate if the underlying condition is unrecognized and untreated. Bone densitometry, using dual-energy x-ray absorptiometry, may underestimate fracture risk in some chronic diseases, including glucocorticoid-induced osteoporosis, type 2 diabetes, and obesity, and may overestimate fracture risk in others (eg, Turner syndrome). FRAX and trabecular bone score may provide additional information regarding fracture risk in secondary osteoporosis, but their use is limited to adults aged ≥ 40 years and ≥ 50 years, respectively. In addition, FRAX requires adjustment in some chronic conditions, such as glucocorticoid use, type 2 diabetes, and HIV. In most conditions, evidence for antiresorptive or anabolic therapy is limited to increases in bone mass. Current osteoporosis management guidelines also neglect secondary osteoporosis and these existing evidence gaps are discussed.
Collapse
Affiliation(s)
- Peter R Ebeling
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia.,Department of Endocrinology, Monash Health, Clayton, Victoria 3168, Australia
| | - Hanh H Nguyen
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia.,Department of Endocrinology, Monash Health, Clayton, Victoria 3168, Australia.,Department of Endocrinology and Diabetes, Western Health, Victoria 3011, Australia
| | - Jasna Aleksova
- Department of Endocrinology, Monash Health, Clayton, Victoria 3168, Australia.,Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Amanda J Vincent
- Department of Endocrinology, Monash Health, Clayton, Victoria 3168, Australia.,Monash Centre for Health Research and Implementation, School of Public Health and Preventative Medicine, Monash University, Clayton, Victoria 3168, Australia
| | - Phillip Wong
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia.,Department of Endocrinology, Monash Health, Clayton, Victoria 3168, Australia.,Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Frances Milat
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia.,Department of Endocrinology, Monash Health, Clayton, Victoria 3168, Australia.,Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| |
Collapse
|
6
|
Nicol JL, Woodrow C, Cunningham BJ, Mollee P, Weber N, Smith MD, Nicol AJ, Gordon LG, Hill MM, Skinner TL. An Individualized Exercise Intervention for People with Multiple Myeloma—Study Protocol of a Randomized Waitlist-Controlled Trial. Curr Oncol 2022; 29:901-923. [PMID: 35200576 PMCID: PMC8870457 DOI: 10.3390/curroncol29020077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/28/2022] [Accepted: 01/31/2022] [Indexed: 11/16/2022] Open
Abstract
People with multiple myeloma (MM) are second only to people with lung cancer for the poorest reported health-related quality of life (HRQoL) of all cancer types. Whether exercise can improve HRQoL in MM, where bone pain and lesions are common, requires investigation. This trial aims to evaluate the efficacy of an exercise intervention compared with control on HRQoL in people with MM. Following baseline testing, people with MM (n = 60) will be randomized to an exercise (EX) or waitlist control (WT) group. EX will complete 12-weeks of supervised (24 sessions) and unsupervised (12 sessions) individualized, modular multimodal exercise training. From weeks 12–52, EX continue unsupervised training thrice weekly, with one optional supervised group-based session weekly from weeks 12–24. The WT will be asked to maintain their current activity levels for the first 12-weeks, before completing the same protocol as EX for the following 52 weeks. Primary (patient-reported HRQoL) and secondary (bone health and pain, fatigue, cardiorespiratory fitness, muscle strength, body composition, disease response, and blood biomarkers) outcomes will be assessed at baseline, 12-, 24- and 52-weeks. Adverse events, attendance, and adherence will be recorded and cost-effectiveness analysis performed. The findings will inform whether exercise should be included as part of standard myeloma care to improve the health of this unique population.
Collapse
Affiliation(s)
- Jennifer L. Nicol
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane 4072, Australia; (B.J.C.); (A.J.N.); (T.L.S.)
- QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia; (L.G.G.); (M.M.H.)
- Correspondence:
| | - Carmel Woodrow
- Haematology, Division of Cancer, Princess Alexandra Hospital, Brisbane 4102, Australia; (C.W.); (P.M.)
| | - Brent J. Cunningham
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane 4072, Australia; (B.J.C.); (A.J.N.); (T.L.S.)
| | - Peter Mollee
- Haematology, Division of Cancer, Princess Alexandra Hospital, Brisbane 4102, Australia; (C.W.); (P.M.)
- Faculty of Medicine, The University of Queensland, Brisbane 4006, Australia
| | - Nicholas Weber
- Haematology, Cancer Care Services, Royal Brisbane and Women’s Hospital, Brisbane 4006, Australia;
| | - Michelle D. Smith
- School of Health and Rehabilitation Sciences, The University of Queensland, Brisbane 4072, Australia;
| | - Andrew J. Nicol
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane 4072, Australia; (B.J.C.); (A.J.N.); (T.L.S.)
- Brisbane Clinic for Lymphoma, Myeloma and Leukaemia, Greenslopes Private Hospital, Brisbane 4120, Australia
| | - Louisa G. Gordon
- QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia; (L.G.G.); (M.M.H.)
- Faculty of Medicine, The University of Queensland, Brisbane 4006, Australia
- Palliative Care Outcomes Centre, School of Nursing and Cancer, Queensland University of Technology, Brisbane 4059, Australia
| | - Michelle M. Hill
- QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia; (L.G.G.); (M.M.H.)
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane 4006, Australia
| | - Tina L. Skinner
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane 4072, Australia; (B.J.C.); (A.J.N.); (T.L.S.)
| |
Collapse
|
7
|
Soto CA, Lo Celso C, Purton LE, Frisch BJ. From the niche to malignant hematopoiesis and back: reciprocal interactions between leukemia and the bone marrow microenvironment. JBMR Plus 2021; 5:e10516. [PMID: 34693187 PMCID: PMC8520063 DOI: 10.1002/jbm4.10516] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/03/2021] [Accepted: 05/13/2021] [Indexed: 12/19/2022] Open
Abstract
The bone marrow microenvironment (BMME) regulates hematopoiesis through a complex network of cellular and molecular components. Hematologic malignancies reside within, and extensively interact with, the same BMME. These interactions consequently alter both malignant and benign hematopoiesis in multiple ways, and can encompass initiation of malignancy, support of malignant progression, resistance to chemotherapy, and loss of normal hematopoiesis. Herein, we will review supporting studies for interactions of the BMME with hematologic malignancies and discuss challenges still facing this exciting field of research. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Celia A. Soto
- Department of PathologyUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | - Cristina Lo Celso
- Department of Life SciencesImperial College LondonLondonUK
- Sir Francis Crick InstituteLondonUK
| | - Louise E. Purton
- St Vincent's Institute of Medical ResearchFitzroyVictoriaAustralia
- Department of Medicine at St. Vincent's HospitalThe University of MelbourneMelbourneVictoriaAustralia
| | - Benjamin J. Frisch
- Department of PathologyUniversity of Rochester Medical CenterRochesterNew YorkUSA
- Wilmot Cancer InstituteUniversity of Rochester School of Medicine and DentistryRochesterNew YorkUSA
- Center for Musculoskeletal ResearchUniversity of Rochester School of Medicine and DentistryRochesterNew YorkUSA
| |
Collapse
|
8
|
Hong M, Chen D, Hong Z, Tang K, Yao Y, Chen L, Ye T, Qian J, Du Y, Sun R. Ex vivo and in vivo chemoprotective activity and potential mechanism of Martynoside against 5-fluorouracil-induced bone marrow cytotoxicity. Biomed Pharmacother 2021; 138:111501. [PMID: 33765584 DOI: 10.1016/j.biopha.2021.111501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/03/2021] [Accepted: 03/09/2021] [Indexed: 12/12/2022] Open
Abstract
Martynoside (MAR) is a bioactive glycoside of Rehmannia glutinosa, a traditional Chinese herb frequently prescribed for treating chemotherapy-induced pancytopenia. Despite its clinical usage in China for thousands of years, the mechanism of MAR's hematopoietic activity and its impact on chemotherapy-induced antitumor activity are still unclear. Here, we showed that MAR protected ex vivo bone marrow cells from 5-fluorouracil (5-FU)-induced cell death and inflammation response by down-regulating the TNF signaling pathway, in which II1b was the most regulatory gene. Besides, using mouse models with melanoma and colon cancer, we further demonstrated that MAR had protective effects against 5-FU-induced myelosuppression in mice without compromising its antitumor activity. Our results showed that MAR increased the number of bone marrow nucleated cells (BMNCs) and the percentage of leukocyte and granulocytic populations in 5-FU-induced myelosuppressive mice, accompanied by an increase in numbers of circulating white blood cells and platelets. The transcriptome profile of BMNCs further showed that the mode of action of MAR might be associated with the increased survival of BMNCs and the improvement of the bone marrow microenvironment. In summary, we revealed the potential molecular mechanism of MAR to counteract 5-FU-induced bone marrow cytotoxicity both ex vivo and in vivo, and highlighted its potential clinical usage in cancer patients experiencing chemotherapy-induced multi-lineage myelosuppression.
Collapse
Affiliation(s)
- Mengying Hong
- Cancer Institute, The Second Affiliated Hospital, ZJU-UCLA Joint Center for Medical Education and Research, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Dongdong Chen
- Cancer Institute, The Second Affiliated Hospital, ZJU-UCLA Joint Center for Medical Education and Research, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Zhuping Hong
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Kejun Tang
- Cancer Institute, The Second Affiliated Hospital, ZJU-UCLA Joint Center for Medical Education and Research, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Yuanyuan Yao
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Liubo Chen
- Cancer Institute, The Second Affiliated Hospital, ZJU-UCLA Joint Center for Medical Education and Research, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Tingting Ye
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Jing Qian
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, China
| | - Yushen Du
- Cancer Institute, The Second Affiliated Hospital, ZJU-UCLA Joint Center for Medical Education and Research, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA.
| | - Ren Sun
- Cancer Institute, The Second Affiliated Hospital, ZJU-UCLA Joint Center for Medical Education and Research, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
9
|
Whyne CM, Ferguson D, Clement A, Rangrez M, Hardisty M. Biomechanical Properties of Metastatically Involved Osteolytic Bone. Curr Osteoporos Rep 2020; 18:705-715. [PMID: 33074529 DOI: 10.1007/s11914-020-00633-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/29/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE OF REVIEW Skeletal metastasis involves the uncoupling of physiologic bone remodeling resulting in abnormal bone turnover and radical changes in bony architecture, density, and quality. Bone strength assessment and fracture risk prediction are critical in clinical treatment decision-making. This review focuses on bone tissue and structural mechanisms altered by osteolytic metastasis and the resulting changes to its material and mechanical behavior. RECENT FINDINGS Both organic and mineral phases of bone tissue are altered by osteolytic metastatic disease, with diminished bone quality evident at multiple length-scales. The mechanical performance of bone with osteolytic lesions is influenced by a combination of tissue-level and structural changes. This review considers the effects of osteolytic metastasis on bone biomechanics demonstrating its negative impact at tissue and structural levels. Future studies need to assess the cumulative impact of cancer treatments on metastatically involved bone quality, and its utility in directing multimodal treatment planning.
Collapse
Affiliation(s)
- Cari M Whyne
- Orthopaedic Biomechanics Lab, Sunnybrook Research Institute, University of Toronto, Toronto, Canada.
- Department of Surgery, University of Toronto, Toronto, Canada.
- Biomedical Engineering, University of Toronto, Toronto, Canada.
| | - Dallis Ferguson
- Orthopaedic Biomechanics Lab, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
- Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Allison Clement
- Orthopaedic Biomechanics Lab, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Mohammedayaz Rangrez
- Orthopaedic Biomechanics Lab, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Michael Hardisty
- Orthopaedic Biomechanics Lab, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
- Department of Surgery, University of Toronto, Toronto, Canada
| |
Collapse
|
10
|
Fraint E, Ulloa BA, Feliz Norberto M, Potts KS, Bowman TV. Advances in preclinical hematopoietic stem cell models and possible implications for improving therapeutic transplantation. Stem Cells Transl Med 2020; 10:337-345. [PMID: 33058566 PMCID: PMC7900582 DOI: 10.1002/sctm.20-0294] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/04/2020] [Accepted: 09/20/2020] [Indexed: 12/11/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a treatment for many malignant, congenital, and acquired hematologic diseases. Some outstanding challenges in the HSCT field include the paucity of immunologically‐matched donors, our inability to effectively expand hematopoeitic stem cells (HSCs) ex vivo, and the high infection risk during engraftment. Scientists are striving to develop protocols to generate, expand, and maintain HSCs ex vivo, however these are not yet ready for clinical application. Given these problems, advancing our understanding of HSC specification, regulation, and differentiation in preclinical models is essential to improve the therapeutic utility of HSCT. In this review, we link biomedical researchers and transplantation clinicians by discussing the potential therapeutic implications of recent fundamental HSC research in model organisms. We consider deficiencies in current HSCT practice, such as problems achieving adequate cell dose for successful and rapid engraftment, immense inflammatory cascade activation after myeloablation, and graft‐vs‐host disease. Furthermore, we discuss recent advances in the field of HSC biology and transplantation made in preclinical models of zebrafish, mouse, and nonhuman primates that could inform emerging practice for clinical application.
Collapse
Affiliation(s)
- Ellen Fraint
- Department of Pediatrics, Children's Hospital at Montefiore, Bronx, New York, USA
| | - Bianca A Ulloa
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, USA.,Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - María Feliz Norberto
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, USA.,Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kathryn S Potts
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, USA.,Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Teresa V Bowman
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, USA.,Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Medicine (Oncology), Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, USA
| |
Collapse
|
11
|
Cheung E, Borno HT. The limitations of today’s clinical guidance: Atypical femoral fracture and long-term bone-modifying agents in the oncology setting. J Oncol Pharm Pract 2020; 26:1180-1189. [DOI: 10.1177/1078155220907965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
While bone-modifying agents such as bisphosphonates and denosumab are crucial to preventing skeletal-related events in patients with bone metastases, the optimal duration remains undefined. Extended duration may be associated with adverse effects such as osteonecrosis of the jaw and atypical femoral fracture. Although uncommon, atypical femoral fracture represents a serious consequence of prolonged bone-modifying agent use and are characterized by a prodrome and distinct radiographic findings. The oncology setting encompasses a unique set of atypical femoral fracture risk factors and considerations, with hormonal therapy in early stage disease, bone metastases in the advanced setting, and new targeted agents that may affect bone homeostasis. As outcomes in cancer treatment continue to improve, the questions of risks versus benefits of long-term bone-modifying agents and how to mitigate atypical femoral fracture risk become increasingly pertinent.
Collapse
Affiliation(s)
- Edna Cheung
- Department of Clinical Pharmacy and School of Pharmacy, University of California San Francisco, San Francisco, USA
| | - Hala T Borno
- Division of Hematology/Oncology, School of Medicine, University of California San Francisco, San Francisco, USA
| |
Collapse
|
12
|
Zhang R, Li J, Li G, Jin F, Wang Z, Yue R, Wang Y, Wang X, Sun Y. LncRNA Nron regulates osteoclastogenesis during orthodontic bone resorption. Int J Oral Sci 2020; 12:14. [PMID: 32385254 PMCID: PMC7210890 DOI: 10.1038/s41368-020-0077-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 02/19/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022] Open
Abstract
Activation of osteoclasts during orthodontic tooth treatment is a prerequisite for alveolar bone resorption and tooth movement. However, the key regulatory molecules involved in osteoclastogenesis during this process remain unclear. Long noncoding RNAs (lncRNAs) are a newly identified class of functional RNAs that regulate cellular processes, such as gene expression and translation regulation. Recently, lncRNAs have been reported to be involved in osteogenesis and bone formation. However, as the most abundant noncoding RNAs in vivo, the potential regulatory role of lncRNAs in osteoclast formation and bone resorption urgently needs to be clarified. We recently found that the lncRNA Nron (long noncoding RNA repressor of the nuclear factor of activated T cells) is highly expressed in osteoclast precursors. Nron is downregulated during osteoclastogenesis and bone ageing. To further determine whether Nron regulates osteoclast activity during orthodontic treatment, osteoclastic Nron transgenic (Nron cTG) and osteoclastic knockout (Nron CKO) mouse models were generated. When Nron was overexpressed, the orthodontic tooth movement rate was reduced. In addition, the number of osteoclasts decreased, and the activity of osteoclasts was inhibited. Mechanistically, Nron controlled the maturation of osteoclasts by regulating NFATc1 nuclear translocation. In contrast, by deleting Nron specifically in osteoclasts, tooth movement speed increased in Nron CKO mice. These results indicate that lncRNAs could be potential targets to regulate osteoclastogenesis and orthodontic tooth movement speed in the clinic in the future.
Collapse
Affiliation(s)
- Ruilin Zhang
- Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Junhui Li
- Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Gongchen Li
- Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Fujun Jin
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Zuolin Wang
- Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Rui Yue
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yibin Wang
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiaogang Wang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Yao Sun
- Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China.
| |
Collapse
|
13
|
Cui J, Li X, Wang S, Su Y, Chen X, Cao L, Zhi X, Qiu Z, Wang Y, Jiang H, Huang B, Ji F, Su J. Triptolide prevents bone loss via suppressing osteoclastogenesis through inhibiting PI3K-AKT-NFATc1 pathway. J Cell Mol Med 2020; 24:6149-6161. [PMID: 32347017 PMCID: PMC7294126 DOI: 10.1111/jcmm.15229] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/14/2020] [Accepted: 02/06/2020] [Indexed: 12/22/2022] Open
Abstract
Bone loss (osteopenia) is a common complication in human solid tumour. In addition, after surgical treatment of gynaecological tumour, osteoporosis often occurs due to the withdrawal of oestrogen. The major characteristic of osteoporosis is the low bone mass with micro-architectural deteriorated bone tissue. And the main cause is the overactivation of osteoclastogenesis, which is one of the most important therapeutic targets. Inflammation could induce the interaction of RANKL/RANK, which is the promoter of osteoclastogenesis. Triptolide is derived from the traditional Chinese herb lei gong teng, presented multiple biological effects, including anti-cancer, anti-inflammation and immunosuppression. We hypothesized that triptolide could inhibits osteoclastogenesis by suppressing inflammation activation. In this study, we confirmed that triptolide could suppress RANKL-induced osteoclastogenesis in bone marrow mononuclear cells (BMMCs) and RAW264.7 cells and inhibited the osteoclast bone resorption functions. PI3K-AKT-NFATc1 pathway is one of the most important downstream pathways of RANKL-induced osteogenesis. The experiments in vitro indicated that triptolide suppresses the activation of PI3K-AKT-NFATc1 pathway and the target point located at the upstream of AKT because both NFATc1 overexpression and AKT phosphorylation could ameliorate the triptolide suppression effects. The expression of MDM2 was elevated, which demonstrated the MDM-p53-induced cell death might contribute to the osteoclastogenesis suppression. Ovariectomy-induced bone loss and inflammation activation were also found to be ameliorated in the experiments in vivo. In summary, the new effect of anti-cancer drug triptolide was demonstrated to be anti-osteoclastogenesis, and we demonstrated triptolide might be a promising therapy for bone loss caused by tumour.
Collapse
Affiliation(s)
- Jin Cui
- Department of Orthopedics, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China.,China-South Korea Bioengineering Center, Shanghai, China
| | - Xiaoqun Li
- Graduate Management Unit, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | | | - Yiming Su
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Xiao Chen
- Department of Orthopedics, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China.,China-South Korea Bioengineering Center, Shanghai, China
| | - Liehu Cao
- Department of Orthopedics, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xin Zhi
- Graduate Management Unit, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zili Qiu
- Jinling High School, Nanjing, China
| | - Yao Wang
- Department of Orthopedics, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hao Jiang
- Department of Orthopedics, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Biaotong Huang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Fang Ji
- Department of Orthopedics, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jiacan Su
- Department of Orthopedics, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China.,China-South Korea Bioengineering Center, Shanghai, China
| |
Collapse
|
14
|
Straszkowski L, Jovic T, Castillo-Tandazo W, Ritchie DS, Purton LE. Effects of chemotherapy agents used to treat pediatric acute lymphoblastic leukemia patients on bone parameters and longitudinal growth of juvenile mice. Exp Hematol 2020; 82:1-7. [PMID: 32006607 DOI: 10.1016/j.exphem.2020.01.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/28/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood cancer. Therapies for pediatric ALL have improved such that more than 80% of patients survive to 5 years post-therapy, and most survive to adulthood. These ALL patients experience long-term side effects that permanently affect their quality of life, with bone loss and reduced longitudinal growth being the most common skeletal complications. To determine the effects of the chemotherapeutic agents used in ALL induction therapy on bone density and longitudinal growth in mice, we treated juvenile mice with doxorubicin, dexamethasone, vincristine, l-asparaginase, or combination therapy. At adulthood, mice were culled and bones collected and scanned by micro-computed tomography (micro-CT). Mice that received doxorubicin and combination therapy exhibited reduced longitudinal growth and significant reductions in trabecular bone volume, trabecular thickness, and trabecular number, with increased trabecular separation. Mean cortical thickness, cortical area, marrow area, endocortical perimeter, and polar moment of inertia were significantly reduced by doxorubicin and combination therapy. Vincristine treatment significantly decreased trabecular bone volume, trabecular number, and increased trabecular separation but had no effects on cortical bone. Dexamethasone treatment increased trabecular bone separation, cortical marrow area, and cortical bone periosteal perimeter. Mice treated with l-asparaginase did not have any bone phenotypes. In conclusion, these data indicate that the majority of the chemotherapy agents used in induction therapy for pediatric ALL have long-term effects on bone in mice. A single dose of doxorubicin in juvenile mice was sufficient to cause the majority of the bone phenotypes, with combination therapy intensifying these effects.
Collapse
Affiliation(s)
| | - Tanja Jovic
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Wilson Castillo-Tandazo
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia; Department of Medicine, University of Melbourne, Parkville, VIC, Australia
| | - David S Ritchie
- Department of Medicine, University of Melbourne, Parkville, VIC, Australia; Peter MacCallum Cancer Centre, Parkville, VIC, Australia; Victorian Comprehensive Cancer Centre, Parkville, VIC, Australia; Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Louise E Purton
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia; Department of Medicine, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
15
|
Walz S, Maas M, Stenzl A, Todenhöfer T. Bone Health Issues in Patients with Prostate Cancer: An Evidence-Based Review. World J Mens Health 2019; 38:151-163. [PMID: 31081297 PMCID: PMC7076314 DOI: 10.5534/wjmh.190044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 03/23/2019] [Indexed: 01/13/2023] Open
Abstract
Bone health in prostate cancer patients represents a prerequisite for acceptable quality of life and optimal outcome of this disease. The major threat for bone health in prostate cancer displays cancer treatment induced bone loss as well as the development of bone metastases. In recent years, several new pharmaceuticals targeting bone metabolism such as denosumab or androgen pathway targeting drugs (abiraterone acetate and enzalutamide) have been approved for the treatment of progressive disease aiming to interrupt the vicious circle of bone metastasis and aberrant bone resorption. This development raised the awareness of the pivotal role of bone health in prostate cancer and introduced (symptomatic) skeletal related events as an important end point in recent clinical trials. Bone targeted drugs have become standard of care in patients with metastatic castration resistant prostate cancer, their role in metastatic hormone sensitive prostate cancer has been discussed controversely. In oligometastatic prostate cancer patients several promising approaches in metastasis directed therapy, including conventional surgery, stereotactic ablative radiation and image-guided single-fraction robotic stereotactic radiosurgery (CyberKnife®) were launched but are not in routine clinical use until now caused by sparse clinical evidence.
Collapse
Affiliation(s)
- Simon Walz
- Department of Urology, Eberhard Karls University, Tuebingen, Germany
| | - Moritz Maas
- Department of Urology, Eberhard Karls University, Tuebingen, Germany
| | - Arnulf Stenzl
- Department of Urology, Eberhard Karls University, Tuebingen, Germany
| | - Tilman Todenhöfer
- Department of Urology, Eberhard Karls University, Tuebingen, Germany.
| |
Collapse
|
16
|
Tjin G, Flores-Figueroa E, Duarte D, Straszkowski L, Scott M, Khorshed RA, Purton LE, Lo Celso C. Imaging methods used to study mouse and human HSC niches: Current and emerging technologies. Bone 2019; 119:19-35. [PMID: 29704697 DOI: 10.1016/j.bone.2018.04.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/23/2018] [Accepted: 04/23/2018] [Indexed: 12/18/2022]
Abstract
Bone marrow contains numerous different cell types arising from hematopoietic stem cells (HSCs) and non-hematopoietic mesenchymal/skeletal stem cells, in addition to other cell types such as endothelial cells- these non-hematopoietic cells are commonly referred to as stromal cells or microenvironment cells. HSC function is intimately linked to complex signals integrated by their niches, formed by combinations of hematopoietic and stromal cells. Studies of hematopoietic cells have been significantly advanced by flow cytometry methods, enabling the quantitation of each cell type in normal and perturbed situations, in addition to the isolation of these cells for molecular and functional studies. Less is known, however, about the specific niches for distinct developing hematopoietic lineages, or the changes occurring in the niche size and function in these distinct anatomical sites in the bone marrow under stress situations and ageing. Significant advances in imaging technology during the last decade have permitted studies of HSC niches in mice. Additional imaging technologies are emerging that will facilitate the study of human HSC niches in trephine BM biopsies. Here we provide an overview of imaging technologies used to study HSC niches, in addition to highlighting emerging technology that will help us to more precisely identify and characterize HSC niches in normal and diseased states.
Collapse
Affiliation(s)
- Gavin Tjin
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Eugenia Flores-Figueroa
- Oncology Research Unit, Oncology Hospital, National Medical Center Century XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Delfim Duarte
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, London, UK; The Sir Francis Crick Institute, London, UK
| | - Lenny Straszkowski
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Mark Scott
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, London, UK; Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Reema A Khorshed
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, London, UK
| | - Louise E Purton
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; The University of Melbourne, Department of Medicine at St Vincent's Hospital, Fitzroy, Victoria, Australia.
| | - Cristina Lo Celso
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, London, UK; The Sir Francis Crick Institute, London, UK.
| |
Collapse
|
17
|
Essex AL, Pin F, Huot JR, Bonewald LF, Plotkin LI, Bonetto A. Bisphosphonate Treatment Ameliorates Chemotherapy-Induced Bone and Muscle Abnormalities in Young Mice. Front Endocrinol (Lausanne) 2019; 10:809. [PMID: 31803146 PMCID: PMC6877551 DOI: 10.3389/fendo.2019.00809] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 11/04/2019] [Indexed: 12/14/2022] Open
Abstract
Chemotherapy is frequently accompanied by several side effects, including nausea, diarrhea, anorexia and fatigue. Evidence from ours and other groups suggests that chemotherapy can also play a major role in causing not only cachexia, but also bone loss. This complicates prognosis and survival among cancer patients, affects quality of life, and can increase morbidity and mortality rates. Recent findings suggest that soluble factors released from resorbing bone directly contribute to loss of muscle mass and function secondary to metastatic cancer. However, it remains unknown whether similar mechanisms also take place following treatments with anticancer drugs. In this study, we found that young male CD2F1 mice (8-week old) treated with the chemotherapeutic agent cisplatin (2.5 mg/kg) presented marked loss of muscle and bone mass. Myotubes exposed to bone conditioned medium from cisplatin-treated mice showed severe atrophy (-33%) suggesting a bone to muscle crosstalk. To test this hypothesis, mice were administered cisplatin in combination with an antiresorptive drug to determine if preservation of bone mass has an effect on muscle mass and strength following chemotherapy treatment. Mice received cisplatin alone or combined with zoledronic acid (ZA; 5 μg/kg), a bisphosphonate routinely used for the treatment of osteoporosis. We found that cisplatin resulted in progressive loss of body weight (-25%), in line with reduced fat (-58%) and lean (-17%) mass. As expected, microCT bone histomorphometry analysis revealed significant reduction in bone mass following administration of chemotherapy, in line with reduced trabecular bone volume (BV/TV) and number (Tb.N), as well as increased trabecular separation (Tb.Sp) in the distal femur. Conversely, trabecular bone was protected when cisplatin was administered in combination with ZA. Interestingly, while the animals exposed to chemotherapy presented significant muscle wasting (~-20% vs. vehicle-treated mice), the administration of ZA in combination with cisplatin resulted in preservation of muscle mass (+12%) and strength (+42%). Altogether, these observations support our hypothesis of bone factors targeting muscle and suggest that pharmacological preservation of bone mass can benefit muscle mass and function following chemotherapy.
Collapse
Affiliation(s)
- Alyson L. Essex
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Fabrizio Pin
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Joshua R. Huot
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Lynda F. Bonewald
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
- Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, United States
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- IUPUI Center for Cachexia Research, Innovation and Therapy, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Lilian I. Plotkin
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Andrea Bonetto
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
- Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, United States
- IUPUI Center for Cachexia Research, Innovation and Therapy, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Otolaryngology – Head & Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- *Correspondence: Andrea Bonetto
| |
Collapse
|
18
|
Kendler DL, Body JJ, Brandi ML, Broady R, Cannata-Andia J, Cannata-Ortiz MJ, El Maghraoui A, Guglielmi G, Hadji P, Pierroz DD, de Villiers TJ, Rizzoli R, Ebeling PR. Bone management in hematologic stem cell transplant recipients. Osteoporos Int 2018; 29:2597-2610. [PMID: 30178158 DOI: 10.1007/s00198-018-4669-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 08/13/2018] [Indexed: 12/13/2022]
Abstract
Autologous and allogeneic hematopoietic stem cell transplantation (HSCT) is the treatment of choice for patients with some malignant and non-malignant hematological diseases. Advances in transplantation techniques and supportive care measures have substantially increased the number of long-term HSCT survivors. This has led to an increasing patient population suffering from the late effects of HSCT, of which, bone loss and its consequent fragility fractures lead to substantial morbidity. Altered bone health, with consequent fragility fractures, and chronic graft-versus-host disease (GVHD) are factors affecting long-term quality of life after HSCT. Hypogonadism, HSCT preparative regimens, nutritional factors, and glucocorticoids all contribute to accelerated bone loss and increased fracture risk. Management strategies should include bone mineral density examination, evaluation of clinical risk factors, and general dietary and physical activity measures. Evidence has accumulated permitting recommendations for more attentiveness to evaluation and monitoring of bone health, with appropriate application of osteoporosis pharmacotherapies to patients at increased risk of bone loss and fracture.
Collapse
Affiliation(s)
- D L Kendler
- Department of Medicine, Division of Endocrinology, University of British Columbia, 150 - 943 W. Broadway, Vancouver, V5Z 4E1, Canada.
| | - J J Body
- CHU Brugmann, Université Libre de Bruxelles, Brussels, Belgium
| | - M L Brandi
- Mineral and Bone Metabolic Unit, Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - R Broady
- Department of Medicine, Division of Hematology, University of British Columbia, Vancouver, Canada
| | - J Cannata-Andia
- Servicio de Metabolismo Óseo y Mineral, Hospital Universitario Central de Asturias, Universidad de Oviedo, Oviedo, Spain
| | - M J Cannata-Ortiz
- Haematology Department, IIS Princesa, Hospital de la Princesa, Madrid, Spain
| | - A El Maghraoui
- Rheumatology Department, Military Hospital Mohammed V, Mohammed V-Souissi University, Rabat, Morocco
| | - G Guglielmi
- Department of Radiology, University of Foggia, Foggia, Italy
| | - P Hadji
- Department of Bone Oncology, Endocrinology and Reproductive Medicine, Nord West Hospital, Frankfurt, Germany
| | - D D Pierroz
- International Osteoporosis Foundation (IOF), Nyon, Switzerland
| | - T J de Villiers
- Department of Gynaecology, Faculty of Health Sciences, Stellenbosch University, Stellenbosch, South Africa
- Mediclinic Panorama, Cape Town, South Africa
| | - R Rizzoli
- Division of Bone Diseases, Faculty of Medicine, Geneva University Hospital, Geneva, Switzerland
| | - P R Ebeling
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Australia
| | | |
Collapse
|
19
|
Green AC, Rudolph-Stringer V, Straszkowski L, Tjin G, Crimeen-Irwin B, Walia M, Martin TJ, Sims NA, Purton LE. Retinoic Acid Receptor γ Activity in Mesenchymal Stem Cells Regulates Endochondral Bone, Angiogenesis, and B Lymphopoiesis. J Bone Miner Res 2018; 33:2202-2213. [PMID: 30040873 DOI: 10.1002/jbmr.3558] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 07/02/2018] [Accepted: 07/14/2018] [Indexed: 12/17/2022]
Abstract
Retinoic acid receptor (RAR) signaling regulates bone structure and hematopoiesis through intrinsic and extrinsic mechanisms. This study aimed to establish how early in the osteoblast lineage loss of RARγ (Rarg) disrupts the bone marrow microenvironment. Bone structure was analyzed by micro-computed tomography (μCT) in Rarg-/- mice and mice with Rarg conditional deletion in Osterix-Cre-targeted osteoblast progenitors or Prrx1-Cre-targeted mesenchymal stem cells. Rarg-/- tibias exhibited less trabecular and cortical bone and impaired longitudinal and radial growth. The trabecular bone and longitudinal, but not radial, growth defects were recapitulated in Prrx1:RargΔ/Δ mice but not Osx1:RargΔ/Δ mice. Although both male and female Prrx1:RargΔ/Δ mice had low trabecular bone mass, males exhibited increased numbers of trabecular osteoclasts and Prrx1:RargΔ/Δ females had impaired mineral deposition. Both male and female Prrx1:RargΔ/Δ growth plates were narrower than controls and their epiphyses contained hypertrophic chondrocyte islands. Flow cytometry revealed that male Prrx1:RargΔ/Δ bone marrow exhibited elevated pro-B and pre-B lymphocyte numbers, accompanied by increased Cxcl12 expression in bone marrow cells. Prrx1:RargΔ/Δ bone marrow also had elevated megakaryocyte-derived Vegfa expression accompanied by smaller sinusoidal vessels. Thus, RARγ expression by Prrx1-Cre-targeted cells directly regulates endochondral bone formation and indirectly regulates tibial vascularization. Furthermore, RARγ expression by Prrx1-Cre-targeted cells extrinsically regulates osteoclastogenesis and B lymphopoiesis in male mice. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Alanna C Green
- St Vincent's Institute, Fitzroy, VIC, Australia
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| | - Victoria Rudolph-Stringer
- St Vincent's Institute, Fitzroy, VIC, Australia
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| | | | - Gavin Tjin
- St Vincent's Institute, Fitzroy, VIC, Australia
| | | | - Mannu Walia
- St Vincent's Institute, Fitzroy, VIC, Australia
| | - T John Martin
- St Vincent's Institute, Fitzroy, VIC, Australia
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| | - Natalie A Sims
- St Vincent's Institute, Fitzroy, VIC, Australia
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| | - Louise E Purton
- St Vincent's Institute, Fitzroy, VIC, Australia
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| |
Collapse
|
20
|
Xu JJ, Smeets MF, Tan SY, Wall M, Purton LE, Walkley CR. Modeling human RNA spliceosome mutations in the mouse: not all mice were created equal. Exp Hematol 2018; 70:10-23. [PMID: 30408513 DOI: 10.1016/j.exphem.2018.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/26/2018] [Accepted: 11/01/2018] [Indexed: 01/23/2023]
Abstract
Myelodysplastic syndromes (MDS) and related myelodysplastic/myeloproliferative neoplasms (MDS/MPNs) are clonal stem cell disorders, primarily affecting patients over 65 years of age. Mapping of the MDS and MDS/MPN genome identified recurrent heterozygous mutations in the RNA splicing machinery, with the SF3B1, SRSF2, and U2AF1 genes being frequently mutated. To better understand how spliceosomal mutations contribute to MDS pathogenesis in vivo, numerous groups have sought to establish conditional murine models of SF3B1, SRSF2, and U2AF1 mutations. The high degree of conservation of hematopoiesis between mice and human and the well-established phenotyping and genetic modification approaches make murine models an effective tool with which to study how a gene mutation contributes to disease pathogenesis. The murine models of spliceosomal mutations described to date recapitulate human MDS or MDS/MPN to varying extents. Reasons for the differences in phenotypes reported between alleles of the same mutation are varied, but the nature of the genetic modification itself and subsequent analysis methods are important to consider. In this review, we summarize recently reported murine models of SF3B1, SRSF2, and U2AF1 mutations, with a particular focus on the genetically engineered modifications underlying the models and the experimental approaches applied.
Collapse
Affiliation(s)
- Jane Jialu Xu
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia
| | - Monique F Smeets
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia
| | - Shuh Ying Tan
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia; Department of Hematology, St. Vincent's Hospital, Fitzroy, Victoria 3065, Australia
| | - Meaghan Wall
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia; Victorian Cancer Cytogenetics Service, St. Vincent's Hospital, Fitzroy, Victoria 3065, Australia
| | - Louise E Purton
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia
| | - Carl R Walkley
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria 3000, Australia.
| |
Collapse
|
21
|
Abstract
BACKGROUND Effects of high-dose radiation using protons and photons on bone are relatively unexplored, but high rates of insufficiency fractures are reported, and the causes of this are incompletely understood. Imaging studies with pre- and postradiation scans can help one understand the effect of radiation on bone. QUESTIONS/PURPOSES The purpose of this study was to assess the effects of high-dose radiation on the trabecular density of bone in the sacrum using CT-derived Hounsfield units (HU). METHODS Between 2009 and 2015, we treated 57 patients (older then 18 years) with sacral chordoma. Fourteen (25%) of them were treated with radiation only. The general indication for this approach is inoperability resulting from tumor size. Forty-two (74%) patients were treated with transverse sacral resections and high-dose radiotherapy (using either protons or photons or a combination) before surgery and after surgery. During this time period, our indication for this approach generally was symptomatic sacral chordoma in which resection would prevent further growth and reasonable sacrifice of nerve roots was possible. Of those patients, 21 (50%) had CT scans both before and after radiation treatment. We used HU as a surrogate for bone density. CT uses HU to derive information on tissue and bone quantity. A recent study presented reference HU values for normal (mean 133 ± 38 HU), osteoporotic (101 ± 25 HU), and osteopenic bone (79 ± 32 HU). To adjust for scanning protocol-induced changes in HU, we calculated the ratio between bone inside and outside the radiation field rather than using absolute values. To assess the effect of radiation, we tested whether there was a difference in ratio (sacrum/L1) before and after radiation. A control measurement was performed (L2/L1) and also tested for a difference before and after radiation. Statistical analyses were performed using the paired t-test. RESULTS The effects of radiation appeared confined to the intended field, because the bone density outside the treated field was not observed to decrease. The ratio of HU (a surrogate for bone density) in L2 relative to L1 did not change after radiotherapy (preradiation mean: 0.979 ± 0.009, postradiation mean: 0.980 ± 0.009, mean difference outside the radiation field: -0.001, 95% confidence interval [CI], -0.009 to 0.007, p = 0.799). The ratio of HU within the radiation field relative to L1 decreased after radiotherapy (preradiation mean: 0.895 ± 0.050, postradiation mean: 0.658 ± 0.050, mean difference inside the radiation field: 0.237, 95% CI, 0.187-0.287, p < 0.001), suggesting the bone density stayed the same outside the radiation field but decreased inside the radiation field. CONCLUSIONS Trabecular bone density decreased after high-dose radiation therapy in a small group of patients with sacral chordoma. High-dose radiation is increasingly gaining acceptance for treating sacral malignancies; further long-term prospective studies using calibrated CT scanners and preferably bone biopsies are needed. LEVEL OF EVIDENCE Level IV, therapeutic study.
Collapse
|
22
|
Zhao D, Wang C, Zhao Y, Shu B, Jia Y, Liu S, Wang H, Chang J, Dai W, Lu S, Shi Q, Yang Y, Zhang Y, Wang Y. Cyclophosphamide causes osteoporosis in C57BL/6 male mice: suppressive effects of cyclophosphamide on osteoblastogenesis and osteoclastogenesis. Oncotarget 2017; 8:98163-98183. [PMID: 29228681 PMCID: PMC5716721 DOI: 10.18632/oncotarget.21000] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/23/2017] [Indexed: 12/19/2022] Open
Abstract
The clinical evidence indicated that cyclophosphamide (CPD), one of the chemotherapy drugs, caused severe deteriorations in bones of cancer patients. However, the exact mechanisms by which CPD exerts effects on bone remodeling is not yet fully elucidated. Therefore, this study was performed to investigate the role and potential mechanism of CPD in osteoblastogenesis and osteoclastogenesis. Here it was found that CPD treatment (100mg/kg/day) for 7 days led to osteoporosis phenotype in male mice. CPD inhibited osteoblastogenesis as shown by decreasing the number and differentiation of bone mesenchymal stem cells (MSCs) and reducing the formation and activity of osteoblasts. Moreover, CPD suppressed the osteoclastogenesis mediated by receptor activator for nuclear factor-κ B ligand (RANKL) as shown by reducing the maturation and activity of osteoclasts. At the molecular level, CPD exerted inhibitory effect on the expression of components (Cyclin D1, β-catenin, Wnt 1, Wnt10b) of Wnt/β-catenin signaling pathway in MSCs and osteoblasts-specific factors (alkaline phosphatase, Runx2, and osteocalcin). CPD also down-regulated the expression of the components (tumor necrosis factor receptor-associated factor 6, nuclear factor of activated T-cells cytoplasm 1, c-Fos and NF-κB) of RANKL signaling pathway and the factors (matrix metalloproteinase 9, cathepsin K, tartrate-resistant acid phosphates and carbonic anhydrase II) for osteoclastic activity. Taken together, this study demonstrated that the short-term treatment of CPD induced osteoporosis in mice and the underlying mechanism might be attributed to its marked suppression on osteoblastogenesis and osteoclastogenesis, especially the effect of CPD on bone formation might play a dominant role in its detrimental effects on bone remodeling.
Collapse
Affiliation(s)
- Dongfeng Zhao
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China.,Spine Disease Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Chenglong Wang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China.,Spine Disease Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R China.,Central Laboratory of Research, Longhua Hospital, Shanghai, P.R. China
| | - Yongjian Zhao
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China.,Spine Disease Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Bing Shu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China.,Spine Disease Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Youji Jia
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China.,Spine Disease Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R China
| | - Shufen Liu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China.,Spine Disease Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Hongshen Wang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China.,Spine Disease Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Junli Chang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China.,Spine Disease Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Weiwei Dai
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China.,Central Laboratory of Research, Longhua Hospital, Shanghai, P.R. China
| | - Sheng Lu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China.,Spine Disease Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R China
| | - Qi Shi
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China.,Spine Disease Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Yanping Yang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China.,Spine Disease Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Yan Zhang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China.,Spine Disease Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Yongjun Wang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China.,Spine Disease Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China.,School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| |
Collapse
|
23
|
Koh AJ, Sinder BP, Entezami P, Nilsson L, McCauley LK. The skeletal impact of the chemotherapeutic agent etoposide. Osteoporos Int 2017; 28:2321-2333. [PMID: 28429052 PMCID: PMC5527337 DOI: 10.1007/s00198-017-4032-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 03/29/2017] [Indexed: 12/13/2022]
Abstract
Effects of the chemotherapeutic agent etoposide on the skeleton were determined in mice. Numbers of bone marrow cells were reduced and myeloid cells were increased. Bone volume was significantly decreased with signs of inhibition of bone formation. Etoposide after pre-treatment with zoledronic acid still reduced bone but overall bone volume was higher than with etoposide alone. INTRODUCTION Chemotherapeutics target rapidly dividing tumor cells yet also impact hematopoietic and immune cells in an off target manner. A wide array of therapies have negative side effects on the skeleton rendering patients osteopenic and prone to fracture. This study focused on the pro-apoptotic chemotherapeutic agent etoposide and its short- and long-term treatment effects in the bone marrow and skeleton. METHODS Six- to 16-week-old mice were treated with etoposide (20-25 mg/kg) or vehicle control in short-term (daily for 5-9 days) or long-term (3×/week for 17 days or 6 weeks) regimens. Bone marrow cell populations and their phagocytic/efferocytic functions were analyzed by flow cytometry. Blood cell populations were assessed by CBC analysis. Bone volume and area compartments and osteoclast numbers were measured by microCT, histomorphometry, and TRAP staining. Biomarkers of bone formation (P1NP) and resorption (TRAcP5b) were assayed from serum. Gene expression in bone marrow was assessed using qPCR. RESULTS Flow cytometric analysis of the bone marrow revealed short-term etoposide reduced overall cell numbers and B220+ cells, with increased marrow apoptotic (AnnexinV+PI-) cells, mesenchymal stem-like cells, and CD68+, CD45+, and CD11b+ monocyte/myeloid cells (as a percent of the total marrow). After 6 weeks, the CD68+, Gr1+, CD11b+, and CD45+ cell populations were still relatively increased in etoposide-treated bone marrow. Skeletal phenotyping revealed etoposide decreased bone volume, trabecular thickness, and cortical bone volume. Gene expression in the marrow for the leptin receptor and CXCL12 were reduced with short-term etoposide, and an increased ratio of RANKL/OPG mRNA was observed. In whole bone, Runx2 and osteocalcin gene expressions were reduced, and in serum, P1NP was significantly reduced with etoposide. Treatment with the antiresorptive agent zoledronic acid prior to etoposide increased bone volume and improved the etoposide-induced decrease in skeletal parameters. CONCLUSIONS These data suggest that etoposide induces apoptosis in the bone marrow and significantly reduces parameters of bone formation with rapid reduction in bone volume. Pre-treatment with an antiresorptive agent results in a preservation of bone mass. Preventive approaches to preserving the skeleton should be considered in human clinical studies.
Collapse
Affiliation(s)
- A J Koh
- Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, 48109-1078, USA
| | - B P Sinder
- Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, 48109-1078, USA
| | - P Entezami
- Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, 48109-1078, USA
| | - L Nilsson
- Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, 48109-1078, USA
| | - L K McCauley
- Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, 48109-1078, USA.
- Department of Pathology, School of Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
- School of Dentistry, University of Michigan, 1011 N. University Ave., Ann Arbor, MI, 48109-1078, USA.
| |
Collapse
|
24
|
Chen X, Zhi X, Pan P, Cui J, Cao L, Weng W, Zhou Q, Wang L, Zhai X, Zhao Q, Hu H, Huang B, Su J. Matrine prevents bone loss in ovariectomized mice by inhibiting RANKL-induced osteoclastogenesis. FASEB J 2017; 31:4855-4865. [PMID: 28739641 PMCID: PMC5636701 DOI: 10.1096/fj.201700316r] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 07/05/2017] [Indexed: 01/30/2023]
Abstract
Osteoporosis is a metabolic bone disease characterized by decreased bone density and strength due to excessive loss of bone protein and mineral content. The imbalance between osteogenesis by osteoblasts and osteoclastogenesis by osteoclasts contributes to the pathogenesis of postmenopausal osteoporosis. Estrogen withdrawal leads to increased levels of proinflammatory cytokines. Overactivated osteoclasts by inflammation play a vital role in the imbalance. Matrine is an alkaloid found in plants from the Sophora genus with various pharmacological effects, including anti-inflammatory activity. Here we demonstrate that matrine significantly prevented ovariectomy-induced bone loss and inhibited osteoclastogenesis in vivo with decreased serum levels of TRAcp5b, TNF-α, and IL-6. In vitro matrine significantly inhibited osteoclast differentiation induced by receptor activator for NF-κB ligand (RANKL) and M-CSF in bone marrow monocytes and RAW264.7 cells as demonstrated by tartrate-resistant acid phosphatase (TRAP) staining and actin-ring formation as well as bone resorption through pit formation assays. For molecular mechanisms, matrine abrogated RANKL-induced activation of NF-κB, AKT, and MAPK pathways and suppressed osteoclastogenesis-related marker expression, including matrix metalloproteinase 9, NFATc1, TRAP, C-Src, and cathepsin K. Our study demonstrates that matrine inhibits osteoclastogenesis through modulation of multiple pathways and that matrine is a promising agent in the treatment of osteoclast-related diseases such as osteoporosis.-Chen, X., Zhi, X., Pan, P., Cui, J., Cao, L., Weng, W., Zhou, Q., Wang, L., Zhai, X. Zhao, Q., Hu, H., Huang, B., Su, J. Matrine prevents bone loss in ovariectomized mice by inhibiting RANKL-induced osteoclastogenesis.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Orthopedics Trauma and Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China.,China-South Korea Bioengineering Center, Shanghai, China
| | - Xin Zhi
- Graduate Management Unit, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China; and
| | - Panpan Pan
- Department of Orthopedics Trauma and Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China.,China-South Korea Bioengineering Center, Shanghai, China
| | - Jin Cui
- Graduate Management Unit, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China; and
| | - Liehu Cao
- Department of Orthopedics Trauma and Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China.,China-South Korea Bioengineering Center, Shanghai, China
| | - Weizong Weng
- Department of Orthopedics Trauma and Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China.,China-South Korea Bioengineering Center, Shanghai, China
| | - Qirong Zhou
- Department of Orthopedics Trauma and Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China.,China-South Korea Bioengineering Center, Shanghai, China
| | - Lin Wang
- Department of Orthopedics Trauma and Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China.,China-South Korea Bioengineering Center, Shanghai, China
| | - Xiao Zhai
- Department of Orthopedics Trauma and Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Qingiie Zhao
- China-South Korea Bioengineering Center, Shanghai, China.,School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Honggang Hu
- China-South Korea Bioengineering Center, Shanghai, China.,School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Biaotong Huang
- China-South Korea Bioengineering Center, Shanghai, China
| | - Jiacan Su
- Department of Orthopedics Trauma and Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China; .,China-South Korea Bioengineering Center, Shanghai, China
| |
Collapse
|
25
|
Grigg A, Butcher B, Khodr B, Bajel A, Hertzberg M, Patil S, D'Souza AB, Ganly P, Ebeling P, Wong E. An individualised risk-adapted protocol of pre- and post transplant zoledronic acid reduces bone loss after allogeneic stem cell transplantation: results of a phase II prospective trial. Bone Marrow Transplant 2017. [DOI: 10.1038/bmt.2017.108] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
26
|
Retinoic acid receptor signalling directly regulates osteoblast and adipocyte differentiation from mesenchymal progenitor cells. Exp Cell Res 2016; 350:284-297. [PMID: 27964926 DOI: 10.1016/j.yexcr.2016.12.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 12/05/2016] [Accepted: 12/07/2016] [Indexed: 11/20/2022]
Abstract
Low and high serum retinol levels are associated with increased fracture risk and poor bone health. We recently showed retinoic acid receptors (RARs) are negative regulators of osteoclastogenesis. Here we show RARs are also negative regulators of osteoblast and adipocyte differentiation. The pan-RAR agonist, all-trans retinoic acid (ATRA), directly inhibited differentiation and mineralisation of early osteoprogenitors and impaired the differentiation of more mature osteoblast populations. In contrast, the pan-RAR antagonist, IRX4310, accelerated differentiation of early osteoprogenitors. These effects predominantly occurred via RARγ and were further enhanced by an RARα agonist or antagonist, respectively. RAR agonists similarly impaired adipogenesis in osteogenic cultures. RAR agonist treatment resulted in significant upregulation of the Wnt antagonist, Sfrp4. This accompanied reduced nuclear and cytosolic β-catenin protein and reduced expression of the Wnt target gene Axin2, suggesting impaired Wnt/β-catenin signalling. To determine the effect of RAR inhibition in post-natal mice, IRX4310 was administered to male mice for 10 days and bones were assessed by µCT. No change to trabecular bone volume was observed, however, radial bone growth was impaired. These studies show RARs directly influence osteoblast and adipocyte formation from mesenchymal cells, and inhibition of RAR signalling in vivo impairs radial bone growth in post-natal mice.
Collapse
|
27
|
Ishida T, Suzuki S, Lai CY, Yamazaki S, Kakuta S, Iwakura Y, Nojima M, Takeuchi Y, Higashihara M, Nakauchi H, Otsu M. Pre-Transplantation Blockade of TNF-α-Mediated Oxygen Species Accumulation Protects Hematopoietic Stem Cells. Stem Cells 2016; 35:989-1002. [PMID: 27753160 DOI: 10.1002/stem.2524] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 09/07/2016] [Accepted: 10/04/2016] [Indexed: 12/31/2022]
Abstract
Hematopoietic stem cell (HSC) transplantation (HSCT) for malignancy requires toxic pre-conditioning to maximize anti-tumor effects and donor-HSC engraftment. While this induces bone marrow (BM)-localized inflammation, how this BM environmental change affects transplanted HSCs in vivo remains largely unknown. We here report that, depending on interval between irradiation and HSCT, residence within lethally irradiated recipient BM compromises donor-HSC reconstitution ability. Both in vivo and in vitro we demonstrate that, among inflammatory cytokines, TNF-α plays a role in HSC damage: TNF-α stimulation leads to accumulation of reactive oxygen species (ROS) in highly purified hematopoietic stem/progenitor cells (HSCs/HSPCs). Transplantation of flow-cytometry-sorted murine HSCs reveals damaging effects of accumulated ROS on HSCs. Short-term incubation either with an specific inhibitor of tumor necrosis factor receptor 1 signaling or an antioxidant N-acetyl-L-cysteine (NAC) prevents TNF-α-mediated ROS accumulation in HSCs. Importantly, pre-transplantation exposure to NAC successfully demonstrats protective effects in inflammatory BM on graft-HSCs, exhibiting better reconstitution capability than that of nonprotected control grafts. We thus suggest that in vivo protection of graft-HSCs from BM inflammation is a feasible and attractive approach, which may lead to improved hematopoietic reconstitution kinetics in transplantation with myeloablative conditioning that inevitably causes inflammation in recipient BM. Stem Cells 2017;35:989-1002.
Collapse
Affiliation(s)
- Takashi Ishida
- Department of Hematology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan.,Division of Stem Cell Processing/Stem Cell Bank, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Sachie Suzuki
- Department of Hematology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Chen-Yi Lai
- Division of Stem Cell Processing/Stem Cell Bank, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Satoshi Yamazaki
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Shigeru Kakuta
- Department of Biomedical Science, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Yoichiro Iwakura
- Center for Experimental Animal Models, Institute for Medical Sciences, Tokyo University of Science, Tokyo, Japan
| | - Masanori Nojima
- Division of Advanced Medicine Promotion, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yasuo Takeuchi
- Department of Nephrology in Internal Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Masaaki Higashihara
- Department of Hematology, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Makoto Otsu
- Division of Stem Cell Processing/Stem Cell Bank, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| |
Collapse
|
28
|
Multi-color immune-phenotyping of CD34 subsets reveals unexpected differences between various stem cell sources. Bone Marrow Transplant 2016; 51:1093-100. [PMID: 27042837 DOI: 10.1038/bmt.2016.88] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/19/2016] [Accepted: 02/25/2016] [Indexed: 12/23/2022]
Abstract
Flow cytometric routine CD34 analysis enumerates hematopoietic stem and progenitor cells irrespective of their subpopulations although this might predict engraftment dynamics and immune reconstitution. We established a multi-color CD34 assay containing CD133, CD45RA, CD10, CD38 and CD33. We examined PBSC, donor bone marrow (BMd) and BM of patients 1 year after allografting (BM1y) regarding their CD34 subset composition, which differed significantly amongst those materials: the early CD45RA(-)CD133(+)CD38(low) subpopulations were significantly more frequent in PBSC than in BMd, and very low in BM1y. Vice versa, clearly more committed CD34 stages prevailed in BM, particularly in BM1y where the proportion of multi-lymphoid and CD38(++) B-lymphoid precursors was highest (mean 59%). CD33 was expressed at different intensity on CD45RA(±)CD133(±) subsets allowing discrimination of earlier from more committed myeloid precursors. Compared with conventional CD34(+) cell enumeration, the presented multi-color phenotyping is a qualitative approach defining different CD34 subtypes in any CD34 source. Its potential impact to predict engraftment kinetics and immune reconstitution has to be evaluated in future studies.
Collapse
|
29
|
Pawlowska M, Yang Q, Hamata B, Kendler DL, Broady R. Early changes in bone mineral density and trabecular bone score following allogeneic stem cell transplant. Bone Marrow Transplant 2016; 51:738-40. [PMID: 26752146 DOI: 10.1038/bmt.2015.329] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- M Pawlowska
- Department of Endocrinology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Q Yang
- Prohealth Clinical Research, Vancouver, British Columbia, Canada
| | - B Hamata
- Department of Family Medicine, University of British Columbia, Prince George, British Columbia, Canada
| | - D L Kendler
- Department of Endocrinology, University of British Columbia, Vancouver, British Columbia, Canada
| | - R Broady
- Department of Hematology, University of British Columbia, Leukemia BMT Program of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
30
|
Sinder BP, Pettit AR, McCauley LK. Macrophages: Their Emerging Roles in Bone. J Bone Miner Res 2015; 30:2140-9. [PMID: 26531055 PMCID: PMC4876707 DOI: 10.1002/jbmr.2735] [Citation(s) in RCA: 214] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 10/30/2015] [Accepted: 11/03/2015] [Indexed: 12/14/2022]
Abstract
Macrophages are present in nearly all tissues and are critical for development, homeostasis, and regeneration. Resident tissue macrophages of bone, termed osteal macrophages, are recently classified myeloid cells that are distinct from osteoclasts. Osteal macrophages are located immediately adjacent to osteoblasts, regulate bone formation, and play diverse roles in skeletal homeostasis. Genetic or pharmacological modulation of macrophages in vivo results in significant bone phenotypes, and these phenotypes depend on which macrophage subsets are altered. Macrophages are also key mediators of osseous wound healing and fracture repair, with distinct roles at various stages of the repair process. A central function of macrophages is their phagocytic ability. Each day, billions of cells die in the body and efferocytosis (phagocytosis of apoptotic cells) is a critical process in both clearing dead cells and recruitment of replacement progenitor cells to maintain homeostasis. Recent data suggest a role for efferocytosis in bone biology and these new mechanisms are outlined. Finally, although macrophages have an established role in primary tumors, emerging evidence suggests that macrophages in bone support cancers which preferentially metastasize to the skeleton. Collectively, this developing area of osteoimmunology raises new questions and promises to provide novel insights into pathophysiologic conditions as well as therapeutic and regenerative approaches vital for skeletal health.
Collapse
Affiliation(s)
- Benjamin P Sinder
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Allison R Pettit
- Blood and Bone Diseases Program, Mater Research Institute–The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Laurie K McCauley
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Medical School, Ann Arbor, MI, USA
| |
Collapse
|
31
|
Flores-Figueroa E, Essers M, Bowman TV. Hematopoiesis "awakens": Evolving technologies, the force behind them. Exp Hematol 2015; 44:101-5. [PMID: 26546749 DOI: 10.1016/j.exphem.2015.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 10/27/2015] [Indexed: 11/24/2022]
Abstract
Amid the beauty of the Kyoto countryside, leaders in the field of hematology met at the 44th annual International Society for Experimental Hematology (ISEH) meeting in late September 2015. Led by ISEH President Paul Frenette and President-Elect David Traver, the meeting covered many aspects of hematopoiesis with a focus on technology. At the meeting, it became clear that the future of hematology is being shaped by innovations in single-cell "omics" and imaging approaches that will provide answers to age-old questions on cellular identity. In this meeting review, we highlight the advances presented in understanding the hematopoietic stem cell (HSC) niche, heterogeneity, stress response, epigenetics, and how these processes change from birth to old age.
Collapse
Affiliation(s)
- Eugenia Flores-Figueroa
- Oncology Research Unit, Oncology Hospital, National Medical Center, IMSS, Mexico City, Mexico
| | - Marieke Essers
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM GmbH), Heidelberg, Germany; Division of Stem Cells and Cancer, Hematopoietic Stem Cells and Stress Group, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Teresa V Bowman
- Departments of Developmental and Molecular Biology and Medicine (Oncology), Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY.
| |
Collapse
|
32
|
Ciliary neurotrophic factor has intrinsic and extrinsic roles in regulating B cell differentiation and bone structure. Sci Rep 2015; 5:15529. [PMID: 26487326 PMCID: PMC4614391 DOI: 10.1038/srep15529] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 09/22/2015] [Indexed: 02/07/2023] Open
Abstract
The gp130 receptor and its binding partners play a central role in cytokine signalling. Ciliary neurotrophic factor (CNTF) is one of the cytokines that signals through the gp130 receptor complex. CNTF has previously been shown to be a negative regulator of trabecular bone remodelling and important for motor neuron development. Since haematopoietic cell maintenance and differentiation is dependent on the bone marrow (BM) microenvironment, where cells of the osteoblastic lineage are important regulators, we hypothesised that CNTF may also have important roles in regulating haematopoiesis. Analysis of haematopoietic parameters in male and female Cntf−/− mice at 12 and 24 weeks of age revealed altered B lymphopoiesis. Strikingly, the B lymphocyte phenotype differed based on sex, age and also the BM microenvironment in which the B cells develop. When BM cells from wildtype mice were transplanted into Cntf−/− mice, there were minimal effects on B lymphopoiesis or bone parameters. However, when Cntf−/− BM cells were transplanted into a wildtype BM microenvironment, there were changes in both haematopoiesis and bone parameters. Our data reveal that haematopoietic cell-derived CNTF has roles in regulating BM B cell lymphopoiesis and both trabecular and cortical bone, the latter in a sex-dependent manner.
Collapse
|
33
|
Abstract
It is estimated that bone loss occurs in 70 % of all patients dying from cancer, causing a significant disease burden in cancer patients. Bone loss is caused by cancer itself and its metastases, but also by cancer therapies. Of the cancer therapy-induced bone loss, hormone therapies are best known for their bone damaging abilities. However, chemo- and radiotherapy may result in bone loss too. In this review, direct and indirect effects of various chemotherapies (such as methotrexate, imatinib, and taxanes) that cause bone loss are discussed. Furthermore, we discuss bone loss caused by radiotherapy and radionuclides, of which the latter may be reduced with the introduction of the alpha-emitter Radium-223. Finally, agents preventing chemotherapy- or radiotherapy-induced bone loss, in particular denosumab and bisphosphonates, are being reviewed for their efficacy in preventing chemotherapy- and irradiation-induced bone loss in cancer patients.
Collapse
Affiliation(s)
- Michel D Wissing
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands,
| |
Collapse
|