1
|
Chawla P, Gau D, Chen F, Welling N, Boone D, Taboas J, Lee AV, Galson DL, Roy P. Breast cancer cells promote osteoclast differentiation in an MRTF-dependent paracrine manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.06.570453. [PMID: 38106226 PMCID: PMC10723471 DOI: 10.1101/2023.12.06.570453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Bone is a frequent site for breast cancer metastasis. Conditioning of the local tumor microenvironment (TME) through crosstalk between tumor cells and bone resident cells in the metastatic niche is a major driving force for bone colonization of breast cancer cells. The vast majority of breast cancer-associated metastasis is osteolytic in nature, and RANKL-induced differentiation of bone marrow-derived macrophages to osteoclasts (OCLs) is a key requirement for osteolytic metastatic growth of cancer cells. In this study, we demonstrate that breast cancer cell-secreted factors stimulate RANKL-induced OCL differentiation of BMDMs requiring the function of Myocardin-related transcription factor (MRTF) in tumor cells. This is partly attributed to the critical role of MRTF in maintaining the basal cellular expression of connective tissue growth factor (CTGF), a pro-osteoclastogenic matricellular factor known to promote bone metastasis in human breast cancer. Supporting these in vitro findings, bioinformatics analyses of multiple human breast cancer transcriptome datasets reveal a strong positive correlation between CTGF expression and MRTF gene signature further establishing the relevance of our findings in a human disease context. By Luminex analyses, we show that MRTF depletion in breast cancer cells has a broad impact on OCL-regulatory cell-secreted factors that extends beyond CTGF. These findings, taken together with demonstration of MRTF-dependence for bone colonization breast cancer cells in vivo, suggest that MRTF inhibition could be an effective strategy to diminish OCL formation and skeletal involvement in breast cancer. In summary, this study highlights a novel tumor-extrinsic function of MRTF relevant to breast cancer metastasis.
Collapse
|
2
|
Wang CH, Huang YN, Liao WL, Hsieh AR, Lin WD, Liu KW, Lu WL, Huang CC, Chien YH, Lee NC, Su PH, Tsai FJ. GBA1 as a risk gene for osteoporosis in the specific populations and its role in the development of Gaucher disease. Orphanet J Rare Dis 2024; 19:144. [PMID: 38575988 PMCID: PMC10993575 DOI: 10.1186/s13023-024-03132-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/03/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Osteoporosis and its primary complication, fragility fractures, contribute to substantial global morbidity and mortality. Gaucher disease (GD) is caused by glucocerebrosidase (GBA1) deficiency, leading to skeletal complications. This study aimed to investigate the impact of the GBA1 gene on osteoporosis progression in GD patients and the specific populations. METHODS We selected 8115 patients with osteoporosis (T-score ≤ - 2.5) and 55,942 healthy individuals (T-score > - 1) from a clinical database (N = 95,223). Monocytes from GD patients were evaluated in relation to endoplasmic reticulum (ER) stress, inflammasome activation, and osteoclastogenesis. An in vitro model of GD patient's cells treated with adeno-associated virus 9 (AAV9)-GBA1 to assess GBA1 enzyme activity, chitotriosidase activity, ER stress, and osteoclast differentiation. Longitudinal dual-energy X-ray absorptiometry (DXA) data tracking bone density in patients with Gaucher disease (GD) undergoing enzyme replacement therapy (ERT) over an extended period. RESULTS The GBA1 gene variant rs11264345 was significantly associated [P < 0.002, Odds Ratio (OR) = 1.06] with an increased risk of bone disease. Upregulation of Calnexin, NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) and Apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain (ASC) was positively associated with osteoclastogenesis in patients with GD. In vitro AAV9-GBA1 treatment of GD patient cells led to enhanced GBA1 enzyme activity, reduced chitotriosidase activity, diminished ER stress, and decreased osteoclast differentiation. Long-term bone density data suggests that initiating ERT earlier in GD leads to greater improvements in bone density. CONCLUSIONS Elevated ER stress and inflammasome activation are indicative of osteoporosis development, suggesting the need for clinical monitoring of patients with GD. Furthermore, disease-associated variant in the GBA1 gene may constitute a risk factor predisposing specific populations to osteoporosis.
Collapse
Affiliation(s)
- Chung-Hsing Wang
- Division of Genetics and Metabolism, Children's Hospital of China Medical University, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
- Department of Pediatrics, Chung Shan Medical University Hospital, No. 110, Sec. 1, Jianguo N. Rd., South Dist., Taichung, 402306, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Nan Huang
- Department of Pediatrics, Chung Shan Medical University Hospital, No. 110, Sec. 1, Jianguo N. Rd., South Dist., Taichung, 402306, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Life Sciences, National Chung-Hsing University, Taichung, Taiwan
| | - Wen-Ling Liao
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Research, Center for Personalized Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Ai-Ru Hsieh
- Department of Statistics, Tamkang University, New Taipei City, Taiwan
| | - Wei-De Lin
- Department of Medical Research, Genetic Center, China Medical University Hospital, No. 2 Yuh-Der Road, Taichung, 404, Taiwan
- School of Post Baccalaureate Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Kai-Wen Liu
- Division of Genetics and Metabolism, Children's Hospital of China Medical University, Taichung, Taiwan
- Department of Life Sciences, National Chung-Hsing University, Taichung, Taiwan
| | - Wen-Li Lu
- Division of Genetics and Metabolism, Children's Hospital of China Medical University, Taichung, Taiwan
| | - Chieh-Chen Huang
- Department of Life Sciences, National Chung-Hsing University, Taichung, Taiwan
| | - Yin-Hsiu Chien
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Ni-Chung Lee
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Pen-Hua Su
- Department of Pediatrics, Chung Shan Medical University Hospital, No. 110, Sec. 1, Jianguo N. Rd., South Dist., Taichung, 402306, Taiwan.
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan.
| | - Fuu-Jen Tsai
- Division of Genetics and Metabolism, Children's Hospital of China Medical University, Taichung, Taiwan.
- Department of Medical Research, Genetic Center, China Medical University Hospital, No. 2 Yuh-Der Road, Taichung, 404, Taiwan.
| |
Collapse
|
3
|
Suzuki T, Ishii S, Katayama Y. Regulation of granulocyte colony-stimulating factor-induced hematopoietic stem cell mobilization by the sympathetic nervous system. Curr Opin Hematol 2023; 30:124-129. [PMID: 37052297 DOI: 10.1097/moh.0000000000000764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
PURPOSE OF REVIEW Granulocyte colony-stimulating factor (G-CSF) is now a standard agent to mobilize hematopoietic stem cells (HSCs) from the bone marrow to circulation. This review introduced mechanistic insights from the aspect of the sympathetic nervous system (SNS). RECENT FINDINGS Mobilization efficiency is determined by the balance between promotion and suppression pathways critically regulated by the SNS. G-CSF-induced high catecholaminergic tone promotes mobilization by (1) the strong suppression of osteolineage cells as a hematopoietic microenvironment and (2) fibroblast growth factor 23 production from erythroblasts, which inhibits CXCR4 function in HSCs. Simultaneously, SNS signals inhibit mobilization by (1) prostaglandin E2 production from mature neutrophils to induce osteopontin in osteoblasts to anchor HSCs and (2) angiopoietin-like protein 4 production from immature neutrophils via peroxisome proliferator-activated receptor δ to inhibit BM vascular permeability. SUMMARY We now know not only the regulatory mechanisms of G-CSF-induced mobilization but also the leads about unfavorable clinical phenomena, such as low-grade fever, bone pain, and poor mobilizers. Recent understanding of the mechanism will assist clinicians in the treatment for mobilization and researchers in the studies of the hidden potential of BM.
Collapse
|
4
|
Araki D, Chen V, Redekar N, Salisbury-Ruf C, Luo Y, Liu P, Li Y, Smith RH, Dagur P, Combs C, Larochelle A. Post-Transplant Administration of G-CSF Impedes Engraftment of Gene Edited Human Hematopoietic Stem Cells by Exacerbating the p53-Mediated DNA Damage Response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.29.547089. [PMID: 37425704 PMCID: PMC10327043 DOI: 10.1101/2023.06.29.547089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Granulocyte colony stimulating factor (G-CSF) is commonly used as adjunct treatment to hasten recovery from neutropenia following chemotherapy and autologous transplantation of hematopoietic stem and progenitor cells (HSPCs) for malignant disorders. However, the utility of G-CSF administration after ex vivo gene therapy procedures targeting human HSPCs has not been thoroughly evaluated. Here, we provide evidence that post-transplant administration of G-CSF impedes engraftment of CRISPR-Cas9 gene edited human HSPCs in xenograft models. G-CSF acts by exacerbating the p53-mediated DNA damage response triggered by Cas9- mediated DNA double-stranded breaks. Transient p53 inhibition in culture attenuates the negative impact of G-CSF on gene edited HSPC function. In contrast, post-transplant administration of G-CSF does not impair the repopulating properties of unmanipulated human HSPCs or HSPCs genetically engineered by transduction with lentiviral vectors. The potential for post-transplant G-CSF administration to aggravate HSPC toxicity associated with CRISPR-Cas9 gene editing should be considered in the design of ex vivo autologous HSPC gene editing clinical trials.
Collapse
|
5
|
Crees ZD, Rettig MP, DiPersio JF. Innovations in hematopoietic stem-cell mobilization: a review of the novel CXCR4 inhibitor motixafortide. Ther Adv Hematol 2023; 14:20406207231174304. [PMID: 37250913 PMCID: PMC10214082 DOI: 10.1177/20406207231174304] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 04/20/2023] [Indexed: 05/31/2023] Open
Abstract
Hematopoietic stem-cell transplantation (HCT) and stem-cell-based gene therapies rely on the ability to collect sufficient CD34+ hematopoietic stem and progenitor cells (HSPCs), typically via peripheral blood mobilization. Commonly used HSPC mobilization regimens include single-agent granulocyte colony-stimulating factor (G-CSF), plerixafor, chemotherapy, or a combination of these agents. These regimens, however, frequently require multiple days of injections and leukapheresis procedures to collect adequate HSPCs for HCT (minimum = >2 × 106 CD34+ cells/kg; optimal = 5-6 × 106 CD34+ cells/kg). In addition, these regimens frequently yield suboptimal CD34+ HSPC numbers for HSPC-based gene-edited therapies, given the significantly higher HSPC number needed for successful gene-editing and manufacturing. Meanwhile, G-CSF is associated with common adverse events such as bone pain as well as an increased risk of rare but potentially life-threatening splenic rupture. Moreover, G-CSF is unsafe in patients with sickle-cell disease, a key patient population that may benefit from autologous HSPC-based gene-edited therapies, where it has been associated with unacceptable rates of serious vaso-occlusive and thrombotic events. Motixafortide is a novel CXCR4 inhibitor with extended in vivo activity (>48 h) that has been shown in preclinical and clinical trials to rapidly mobilize robust numbers of HSPCs in preparation for HCT, while preferentially mobilizing increased numbers of more primitive HSPCs by immunophenotyping and single-cell RNA expression profiling. In this review, we present a history of stem-cell mobilization and update of recent innovations in novel mobilization strategies with a specific focus on the development of motixafortide, a long-acting CXCR4 inhibitor, as a novel HSPC mobilizing agent.
Collapse
Affiliation(s)
- Zachary D. Crees
- Division of Oncology, School of Medicine,
Washington University in St. Louis, 660 S. Euclid Avenue, Campus Box 8007,
St. Louis, MO 63131, USA
| | - Michael P. Rettig
- Division of Oncology, School of Medicine,
Washington University in St. Louis, St. Louis, MO, USA
| | - John F. DiPersio
- Division of Oncology, School of Medicine,
Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
6
|
Sánchez‐Lanzas R, Kalampalika F, Ganuza M. Diversity in the bone marrow niche: Classic and novel strategies to uncover niche composition. Br J Haematol 2022; 199:647-664. [PMID: 35837798 PMCID: PMC9796334 DOI: 10.1111/bjh.18355] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/20/2022] [Accepted: 06/30/2022] [Indexed: 01/01/2023]
Abstract
Our view on the role and composition of the bone marrow (BM) has dramatically changed over time from a simple nutrient for the bone to a highly complex multicellular tissue that sustains haematopoiesis. Among these cells, multipotent haematopoietic stem cells (HSCs), which are predominantly quiescent, possess unique self-renewal capacity and multilineage differentiation potential and replenish all blood lineages to maintain lifelong haematopoiesis. Adult HSCs reside in specialised BM niches, which support their functions. Much effort has been put into deciphering HSC niches due to their potential clinical relevance. Multiple cell types have been implicated as HSC-niche components including sinusoidal endothelium, perivascular stromal cells, macrophages, megakaryocytes, osteoblasts and sympathetic nerves. In this review we provide a historical perspective on how technical advances, from genetic mouse models to imaging and high-throughput sequencing techniques, are unveiling the plethora of molecular cues and cellular components that shape the niche and regulate HSC functions.
Collapse
Affiliation(s)
- Raúl Sánchez‐Lanzas
- Centre for Haemato‐Oncology, Barts Cancer InstituteQueen Mary University of LondonLondonUK
| | - Foteini Kalampalika
- Centre for Haemato‐Oncology, Barts Cancer InstituteQueen Mary University of LondonLondonUK
| | - Miguel Ganuza
- Centre for Haemato‐Oncology, Barts Cancer InstituteQueen Mary University of LondonLondonUK
| |
Collapse
|
7
|
Li S, Yao JC, Oetjen KA, Krambs JR, Xia J, Zhang J, Schmidt AP, Helton NM, Fulton RS, Heath SE, Turnbull IR, Mbalaviele G, Ley TJ, Walter MJ, Link DC. IL-1β expression in bone marrow dendritic cells is induced by TLR2 agonists and regulates HSC function. Blood 2022; 140:1607-1620. [PMID: 35675516 PMCID: PMC9707400 DOI: 10.1182/blood.2022016084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/14/2022] [Indexed: 12/14/2022] Open
Abstract
Hematopoietic stem/progenitor cells (HSPCs) reside in localized microenvironments, or niches, in the bone marrow that provide key signals regulating their activity. A fundamental property of hematopoiesis is the ability to respond to environmental cues such as inflammation. How these cues are transmitted to HSPCs within hematopoietic niches is not well established. Here, we show that perivascular bone marrow dendritic cells (DCs) express a high basal level of Toll-like receptor-1 (TLR1) and TLR2. Systemic treatment with a TLR1/2 agonist induces HSPC expansion and mobilization. It also induces marked alterations in the bone marrow microenvironment, including a decrease in osteoblast activity and sinusoidal endothelial cell numbers. TLR1/2 agonist treatment of mice in which Myd88 is deleted specifically in DCs using Zbtb46-Cre show that the TLR1/2-induced expansion of multipotent HPSCs, but not HSPC mobilization or alterations in the bone marrow microenvironment, is dependent on TLR1/2 signaling in DCs. Interleukin-1β (IL-1β) is constitutively expressed in both murine and human DCs and is further induced after TLR1/2 stimulation. Systemic TLR1/2 agonist treatment of Il1r1-/- mice show that TLR1/2-induced HSPC expansion is dependent on IL-1β signaling. Single-cell RNA-sequencing of low-risk myelodysplastic syndrome bone marrow revealed that IL1B and TLR1 expression is increased in DCs. Collectively, these data suggest a model in which TLR1/2 stimulation of DCs induces secretion of IL-1β and other inflammatory cytokines into the perivascular niche, which in turn, regulates multipotent HSPCs. Increased DC TLR1/2 signaling may contribute to altered HSPC function in myelodysplastic syndrome by increasing local IL-1β expression.
Collapse
Affiliation(s)
- Sidan Li
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
- Hematology Oncology Center, Beijing Children’s Hospital, National Center for Children’s Health, Capital Medial University, Beijing, China
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Juo-Chin Yao
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Karolyn A. Oetjen
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Joseph R. Krambs
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Jun Xia
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Jingzhu Zhang
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Amy P. Schmidt
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Nichole M. Helton
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Robert S. Fulton
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO
| | - Sharon E. Heath
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Isaiah R. Turnbull
- Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | - Gabriel Mbalaviele
- Division of Bone and Mineral Disease, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Timothy J. Ley
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Matthew J. Walter
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Daniel C. Link
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
8
|
Huang J, Freyhult E, Buckland R, Josefsson A, Damber JE, Welén K. Osteoclasts directly influence castration-resistant prostate cancer cells. Clin Exp Metastasis 2022; 39:801-814. [PMID: 35971022 PMCID: PMC9474581 DOI: 10.1007/s10585-022-10179-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/28/2022] [Indexed: 11/30/2022]
Abstract
Metastasis to bone is the leading cause of death from prostate cancer. Interaction between tumor cells and bone cells can promote progression and influence tumor phenotype. It is known that prostate cancer cells support osteoclast differentiation, and degradation of bone matrix by osteoclasts releases growth factors stimulating tumor cell proliferation and invasion. In the present study osteolytic (PC-3) and osteoblastic (LNCaP-19) castration-resistant prostate cancer (CRPC) cells were co-cultured with mature osteoclasts or their precursor cells (RAW 264.7) to characterize direct effects of mature osteoclasts on CRPC cells. Osteoclasts increased proliferation and decrease apoptosis of CRPC cells as assessed with flow cytometry. RNA sequencing revealed that osteolytic CRPC cells were more responsive to osteoclast stimulation regarding gene expression, but the overall induced expression patterns were similar between the prostate cancer cell lines. Genes related to DNA repair were upregulated by osteoclasts, while genes related to endoplasmic reticulum stress-induced apoptosis and cholesterol synthesis were downregulated. The results of this study shows that osteoclasts directly influence CRPC cells, increasing proliferation, decreasing apoptosis, and affecting gene expression pathways that can affect sensitivity to DNA damage and endoplasmic reticulum function. This suggests targeting of osteoclasts to be a possible way to affect efficacy of other drugs by combination regimens in treating prostate cancer metastases.
Collapse
Affiliation(s)
- Junchi Huang
- Department of Urology, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Eva Freyhult
- Department of Cell and Molecular Biology, Science for Life Laboratory, National Bioinformatics Infrastructure Sweden, Uppsala University, 75124, Uppsala, Sweden
| | - Robert Buckland
- Department of Surgical and Perioperative Sciences, Umeå University, Urology & Andrology, Umeå, Sweden
| | - Andreas Josefsson
- Department of Urology, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Surgical and Perioperative Sciences, Umeå University, Urology & Andrology, Umeå, Sweden
- Wallenberg Center for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
| | - Jan-Erik Damber
- Department of Urology, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karin Welén
- Department of Urology, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
9
|
Krüger TB, Syversen U, Herlofson BB, Lian AM, Reseland JE. Targeting a therapeutically relevant concentration of alendronate for in vitro studies on osteoblasts. Acta Odontol Scand 2022; 80:619-625. [PMID: 35605138 DOI: 10.1080/00016357.2022.2072522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
OBJECTIVE Bisphosphonates like alendronate mainly exert their effects on osteoclasts. However, osteoblasts are also affected, but exposed to a much lower concentration in vivo than the osteoclasts. Given that the effects are dose-dependent, the intention of the study was to identify a therapeutically relevant concentration of alendronate for in vitro studies on osteoblasts. MATERIALS AND METHODS Primary human osteoblasts were incubated with alendronate (5, 20 and 100 µM) for 1, 3, 7 and 14 days. Proliferation and viability were assessed, and the effects on cellular growth and function were evaluated by multianalyte profiling of selected proteins in cell culture media using the Luminex 200TM. RESULTS The viability was not affected by any of the dosages. Exposure to 5 µM alendronate had a neutral effect on osteoblast proliferation, and on secretion of osteogenic and inflammatory markers, while enhancing synthesis of a marker of angiogenesis. 20 µM alendronate induced a decline in proliferation and affected angiogenic and osteogenic biomarkers adversely. 100 µM alendronate reduced proliferation dramatically, and this dosage was excluded from further experiments. CONCLUSION A concentration of 5 µM alendronate exerted effects on human osteoblasts that may translate to those observed in vivo and could therefore be relevant for in vitro studies.
Collapse
Affiliation(s)
- Tormod B. Krüger
- Department of Oral Surgery and Oral Medicine, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Unni Syversen
- Department of Biomaterials, Faculty of Dentistry, University of Oslo, Oslo, Norway
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU—Norwegian University of Science and Technology, Trondheim, Norway
- Department of Endocrinology, Clinic of medicine, St. Olavs University Hospital, Trondheim, Norway
| | - Bente B. Herlofson
- Department of Oral Surgery and Oral Medicine, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Aina M. Lian
- Oral Research Laboratory, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Janne E. Reseland
- Department of Biomaterials, Faculty of Dentistry, University of Oslo, Oslo, Norway
| |
Collapse
|
10
|
Dikilitas A, Karaaslan F, Aydin EÖ, Yigit U, Ertugrul AS. Granulocyte-macrophage colony-stimulating factor (GM-CSF) in subjects with different stages of periodontitis according to the new classification. J Appl Oral Sci 2022; 30:e20210423. [PMID: 35262594 PMCID: PMC8908860 DOI: 10.1590/1678-7757-2021-0423] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 01/05/2022] [Indexed: 12/29/2022] Open
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a multifunctional cytokine that regulates inflammatory responses in various autoimmune and inflammatory disorders. OBJECTIVE The purpose of this study was to analyze the gingival crevicular fluid (GCF) for GM-CSF, interleukin-1 beta (IL-1β), and macrophage inflammatory protein-1 alpha (MIP-1α) levels in patients with stage I, stage II, stage III, and stage IV periodontitis (SI-P, SII-P, SIII-P, and SIV-P). METHODOLOGY A total of 126 individuals were recruited for this study, including 21 periodontal healthy (PH), 21 gingivitis (G), 21 SI-P, 21 SII-P, 21 SIII-P, and 21 SIV-P patients. Plaque index (PI), gingival index (GI), presence of bleeding on probing (BOP), probing depth (PD), and attachment loss (AL) were used during the clinical periodontal assessment. GCF samples were obtained and analyzed by an enzyme-linked immunosorbent assay (ELISA). RESULTS GCF GM-CSF, MIP-1α, and IL-1β were significantly higher in SII-P and SIII-P groups than in PH, G, and SI-P groups (p<0.05). There was no significant difference among the PH, G, and SI-P groups in IL-1β, GM-CSF, and MIP-1α levels (p>0.05). CONCLUSIONS These results show that GM-CSF expression was increased in SII-P, SIII-P, and SIV-P. Furthermore, GM-CSF levels may have some potential to discriminate between early and advanced stages of periodontitis.
Collapse
Affiliation(s)
- Ahu Dikilitas
- Usak UniversityFaculty of DentistryDepartment of PeriodontologyUsakTurkeyUsak University, Faculty of Dentistry, Department of Periodontology, Usak, Turkey.
| | - Fatih Karaaslan
- Usak UniversityFaculty of DentistryDepartment of PeriodontologyUsakTurkeyUsak University, Faculty of Dentistry, Department of Periodontology, Usak, Turkey.
| | - Esra Özge Aydin
- Usak UniversityFaculty of DentistryDepartment of PeriodontologyUsakTurkeyUsak University, Faculty of Dentistry, Department of Periodontology, Usak, Turkey.
| | - Umut Yigit
- Usak UniversityFaculty of DentistryDepartment of PeriodontologyUsakTurkeyUsak University, Faculty of Dentistry, Department of Periodontology, Usak, Turkey.
| | - Abdullah Seckin Ertugrul
- IZMIR Katip Celebi UniversityFaculty of DentistryDepartment of PeriodontologyİzmirTurkeyIZMIR Katip Celebi University, Faculty of Dentistry, Department of Periodontology, İzmir, Turkey.
| |
Collapse
|
11
|
Porfyriou E, Letsa S, Kosmas C. Hematopoietic stem cell mobilization strategies to support high-dose chemotherapy: A focus on relapsed/refractory germ cell tumors. World J Clin Oncol 2021; 12:746-766. [PMID: 34631440 PMCID: PMC8479351 DOI: 10.5306/wjco.v12.i9.746] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/19/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
High-dose chemotherapy (HDCT) with autologous hematopoietic stem cell transplantation has been explored and has played an important role in the management of patients with high-risk germ cell tumors (GCTs) who failed to be cured by conventional chemotherapy. Hematopoietic stem cells (HSCs) collected from the peripheral blood, after appropriate pharmacologic mobilization, have largely replaced bone marrow as the principal source of HSCs in transplants. As it is currently common practice to perform tandem or multiple sequential cycles of HDCT, it is anticipated that collection of large numbers of HSCs from the peripheral blood is a prerequisite for the success of the procedure. Moreover, the CD34+ cell dose/kg of body weight infused after HDCT has proven to be a major determinant of hematopoietic engraftment, with patients who receive > 2 × 106 CD34+ cells/kg having consistent, rapid, and sustained hematopoietic recovery. However, many patients with relapsed/refractory GCTs have been exposed to multiple cycles of myelosuppressive chemotherapy, which compromises the efficacy of HSC mobilization with granulocyte colony-stimulating factor with or without chemotherapy. Therefore, alternative strategies that use novel agents in combination with traditional mobilizing regimens are required. Herein, after an overview of the mechanisms of HSCs mobilization, we review the existing literature regarding studies reporting various HSC mobilization approaches in patients with relapsed/refractory GCTs, and finally report newer experimental mobilization strategies employing novel agents that have been applied in other hematologic or solid malignancies.
Collapse
Affiliation(s)
- Eleni Porfyriou
- Department of Medical Oncology and Hematopoietic Cell Transplant Unit, “Metaxa” Cancer Hospital, Piraeus 18537, Greece
| | - Sylvia Letsa
- Department of Medical Oncology and Hematopoietic Cell Transplant Unit, “Metaxa” Cancer Hospital, Piraeus 18537, Greece
| | - Christos Kosmas
- Department of Medical Oncology and Hematopoietic Cell Transplant Unit, “Metaxa” Cancer Hospital, Piraeus 18537, Greece
| |
Collapse
|
12
|
Kitamura N, Sento S, Sasabe E, Kiyasu K, Nakaji K, Daibata M, Yamamoto T. Vertebral fracture and splenomegaly in a head and neck cancer producing granulocyte colony-stimulating factor: A case report of systemic complications associated with a cytokine-producing solid tumor. Mol Clin Oncol 2021; 15:202. [PMID: 34462658 PMCID: PMC8375029 DOI: 10.3892/mco.2021.2364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/15/2021] [Indexed: 12/01/2022] Open
Abstract
Granulocyte colony-stimulating factor (G-CSF)-producing tumors are rare and are associated with a poor prognosis when they occur in the lungs and the head and neck region. Positron emission tomography/computed tomography has been reported to show systemic specific accumulation of fluorodeoxyglucose in these cases, but the systemic complications associated with the cytokines produced are not well known. We herein present the case of a G-CSF-producing maxillary sinus squamous cell carcinoma in a 73-year-old Japanese woman with a vertebral fracture and splenomegaly. These findings are known severe adverse events of high-dose recombinant human G-CSF treatment. The aim of the present study was to further discuss the hypothesis that cytokines produced by solid tumors may induce spinal vertebral fracture and splenomegaly.
Collapse
Affiliation(s)
- Naoya Kitamura
- Department of Oral and Maxillofacial Surgery, Kochi Medical School, Kochi University, Kochi 783-8505, Japan
| | - Shinya Sento
- Department of Oral and Maxillofacial Surgery, Kochi Medical School, Kochi University, Kochi 783-8505, Japan
| | - Eri Sasabe
- Department of Oral and Maxillofacial Surgery, Kochi Medical School, Kochi University, Kochi 783-8505, Japan
| | - Katsuhito Kiyasu
- Department of Orthopedic Surgery, Kochi Medical School, Kochi University, Kochi 783-8505, Japan
| | - Kosuke Nakaji
- Department of Radiology, Kochi Medical School, Kochi University, Kochi 783-8505, Japan
| | - Masanori Daibata
- Department of Microbiology and Infection, Kochi Medical School, Kochi University, Nankoku, Kochi 783-8505, Japan
| | - Tetsuya Yamamoto
- Department of Oral and Maxillofacial Surgery, Kochi Medical School, Kochi University, Kochi 783-8505, Japan
| |
Collapse
|
13
|
Lévesque JP, Summers KM, Millard SM, Bisht K, Winkler IG, Pettit AR. Role of macrophages and phagocytes in orchestrating normal and pathologic hematopoietic niches. Exp Hematol 2021; 100:12-31.e1. [PMID: 34298116 DOI: 10.1016/j.exphem.2021.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 12/13/2022]
Abstract
The bone marrow (BM) contains a mosaic of niches specialized in supporting different maturity stages of hematopoietic stem and progenitor cells such as hematopoietic stem cells and myeloid, lymphoid, and erythroid progenitors. Recent advances in BM imaging and conditional gene knockout mice have revealed that niches are a complex network of cells of mesenchymal, endothelial, neuronal, and hematopoietic origins, together with local physicochemical parameters. Within these complex structures, phagocytes, such as neutrophils, macrophages, and dendritic cells, all of which are of hematopoietic origin, have been found to be important in regulating several niches in the BM, including hematopoietic stem cell niches, erythropoietic niches, and niches involved in endosteal bone formation. There is also increasing evidence that these macrophages have an important role in adapting hematopoiesis, erythropoiesis, and bone formation in response to inflammatory stressors and play a key part in maintaining the integrity and function of these. Likewise, there is also accumulating evidence that subsets of monocytes, macrophages, and other phagocytes contribute to the progression and response to treatment of several lymphoid malignancies such as multiple myeloma, Hodgkin lymphoma, and non-Hodgkin lymphoma, as well as lymphoblastic leukemia, and may also play a role in myelodysplastic syndrome and myeloproliferative neoplasms associated with Noonan syndrome and aplastic anemia. In this review, the potential functions of macrophages and other phagocytes in normal and pathologic niches are discussed, as are the challenges in studying BM and other tissue-resident macrophages at the molecular level.
Collapse
Affiliation(s)
- Jean-Pierre Lévesque
- Mater Research Institute, University of Queensland, Woolloongabba, QLD, Australia.
| | - Kim M Summers
- Mater Research Institute, University of Queensland, Woolloongabba, QLD, Australia
| | - Susan M Millard
- Mater Research Institute, University of Queensland, Woolloongabba, QLD, Australia
| | - Kavita Bisht
- Mater Research Institute, University of Queensland, Woolloongabba, QLD, Australia
| | - Ingrid G Winkler
- Mater Research Institute, University of Queensland, Woolloongabba, QLD, Australia
| | - Allison R Pettit
- Mater Research Institute, University of Queensland, Woolloongabba, QLD, Australia
| |
Collapse
|
14
|
Yu H, Zhang T, Lu H, Ma Q, Zhao D, Sun J, Wang Z. Granulocyte colony-stimulating factor (G-CSF) mediates bone resorption in periodontitis. BMC Oral Health 2021; 21:299. [PMID: 34118920 PMCID: PMC8196459 DOI: 10.1186/s12903-021-01658-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 05/30/2021] [Indexed: 01/13/2023] Open
Abstract
Background Granulocyte colony-stimulating factor (G-CSF) is an important immune factor that mediates bone metabolism by regulating the functions of osteoclasts and osteoblasts. Bone loss is a serious and progressive result of periodontitis. However, the mechanisms underlying the effects of G-CSF on periodontal inflammation have yet not been completely elucidated. Here, we examined whether an anti-G-CSF antibody could inhibit bone resorption in a model of experimental periodontitis and investigated the local expression of G-CSF in periodontal tissues. Methods Experimental periodontitis was induced in mice using ligatures. The levels of G-CSF in serum and bone marrow were measured; immunofluorescence was then performed to analyze the localization and expression of G-CSF in periodontal tissues. Mice with periodontitis were administered anti-G-CSF antibody by tail vein injection to assess the inhibition of bone resorption. Three-dimensional reconstruction was performed to measure bone destruction‐related parameters via micro-computed tomography analysis. Immunofluorescence staining was used to investigate the presence of osteocalcin-positive osteoblasts; tartrate-resistant acid phosphatase (TRAP) staining was used to observe osteoclast activity in alveolar bone. Results The level of G-CSF in serum was significantly elevated in mice with periodontitis. Immunofluorescence analyses showed that G-CSF was mostly expressed in the cell membrane of gingival epithelial cells; this expression was enhanced in the periodontitis group. Additionally, systemic administration of anti-G-CSF antibody significantly inhibited alveolar bone resorption, as evidenced by improvements in bone volume/total volume, bone surface area/bone volume, trabecular thickness, trabecular spacing, and trabecular pattern factor values. Immunofluorescence analysis revealed an enhanced number of osteocalcin-positive osteoblasts, while TRAP staining revealed reduction of osteoclast activity. Conclusions G-CSF expression levels were significantly up-regulated in the serum and gingival epithelial cells. Together, anti-G-CSF antibody administration could alleviates alveolar bone resorption, suggesting that G-CSF may be one of the essential immune factors that mediate the bone loss in periodontitis.
Collapse
Affiliation(s)
- Hui Yu
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, 8th Gongti South Road, Beijing, China.,Department of Stomatology, Affiliated Zhongshan Hospital of Dalian University, 6th Jiefang Street, Dalian, Liaoning, China
| | - Tianyi Zhang
- Shanxi Medical University, 382th WuyiRoad, Xinghualing Distrct, Taiyuan, Shanxi, China
| | - Haibin Lu
- Department of Stomatology, Affiliated Zhongshan Hospital of Dalian University, 6th Jiefang Street, Dalian, Liaoning, China
| | - Qi Ma
- Department of Pathology, Affiliated Zhongshan Hospital of Dalian University, 6th Jiefang Street, Dalian, Liaoning, China
| | - Dong Zhao
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, 8th Gongti South Road, Beijing, China
| | - Jiang Sun
- Department of Periodontology, Dalian Stomatological Hospital, 935th Changjiang Road, Dalian, Liaoning, China.
| | - Zuomin Wang
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, 8th Gongti South Road, Beijing, China.
| |
Collapse
|
15
|
Krüger TB, Herlofson BB, Lian AM, Syversen U, Reseland JE. Alendronate and omeprazole in combination reduce angiogenic and growth signals from osteoblasts. Bone Rep 2021; 14:100750. [PMID: 33553512 PMCID: PMC7856318 DOI: 10.1016/j.bonr.2021.100750] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 01/06/2021] [Accepted: 01/21/2021] [Indexed: 11/24/2022] Open
Abstract
Objective Due to gastrointestinal side effects of oral bisphosphonates (BPs), proton pump inhibitors (PPIs) are often prescribed. PPIs may enhance the risk of osteonecrosis of the jaw, a rare side effect of BPs. Therefore, the objective of this study was to evaluate the effects of the oral BP alendronate (ALN) and the PPI omeprazole (OME) alone and in combination on primary human osteoblasts and gingival fibroblasts in vitro. Methods Human gingival fibroblasts and normal human osteoblasts were incubated with either 5 μM of ALN or 1 μM of OME, or ALN + OME for 1, 3, 7 or 14 days. Effect on viability was evaluated by the lactate dehydrogenase activity in the medium and on proliferation by quantifying 3H-thymidin incorporation. Multianalyte profiling of proteins in cell culture media was performed using the Luminex 200TM system to assess the effect on selected bone markers and cytokines. Results The proliferation of osteoblasts and fibroblasts was reduced upon exposure to ALN + OME. ALN induced an early, temporary rise in markers of inflammation, and OME and ALN + OME promoted a transient decline. An initial increase in IL-13 occurred after exposure to all three options, whereas ALN + OME promoted IL-8 release after 7 days. OME and ALN + OME promoted a transient reduction in vascular endothelial growth factor (VEGF) from osteoblasts, whereas ALN and ALN + OME induced a late rise in VEGF from fibroblasts. Osteoprotegerin release was enhanced by ALN and suppressed by OME and ALN + OME. Conclusions ALN + OME seemed to exaggerate the negative effects of each drug alone on human osteoblasts and gingival fibroblasts. The anti-proliferative effects, modulation of inflammation and impairment of angiogenesis, may induce unfavorable conditions in periodontal tissue facilitating development of osteonecrosis. Alendronate and omeprazole reduce proliferation in osteoblasts and fibroblasts. Unchanged viability after exposure to either drug or the combination Omeprazole, alone and combined with alendronate, cause impairment of angiogenesis. Alendronate promotes an initial, transient increase in pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Tormod B Krüger
- Department of Oral Surgery and Oral Medicine, Faculty of Dentistry, University of Oslo, Norway
| | - Bente B Herlofson
- Department of Oral Surgery and Oral Medicine, Faculty of Dentistry, University of Oslo, Norway
| | - Aina M Lian
- Clinical Oral Research Laboratory, Faculty of Dentistry, University of Oslo, Norway
| | - Unni Syversen
- Clinical Oral Research Laboratory, Faculty of Dentistry, University of Oslo, Norway.,Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology, 7491 Trondheim, Norway.,Department of Endocrinology, Clinic of Medicine, St. Olavs University Hospital, 7491 Trondheim, Norway
| | - Janne E Reseland
- Clinical Oral Research Laboratory, Faculty of Dentistry, University of Oslo, Norway
| |
Collapse
|
16
|
Najafi S, Ghanavat M, Shahrabi S, Gatavizadeh Z, Saki N. The effect of inflammatory factors and their inhibitors on the hematopoietic stem cells fate. Cell Biol Int 2021; 45:900-912. [PMID: 33386770 DOI: 10.1002/cbin.11545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 12/02/2020] [Accepted: 12/25/2020] [Indexed: 11/12/2022]
Abstract
Inflammatory cytokines exert different effects on hematopoietic stem cells (HSCs), lead to the development of various cell lineages in bone marrow (BM) and are thus a differentiation axis for HSCs. The content used in this article has been obtained by searching PubMed database and Google Scholar search engine of English-language articles (1995-2020) using "Hematopoietic stem cell," "Inflammatory cytokine," "Homeostasis," and "Myelopoiesis." Inflammatory cytokines are involved in the differentiation and proliferation of hematopoietic progenitors to compensate for cellular death due to inflammation. Since each of these cytokines differentiates HSCs into a specific cell line, the difference in the effect of these cytokines on the fate of HSC progenitors can be predicted. Inhibitors of these cytokines can also control the inflammatory process as well as the cells involved in leukemic conditions. In general, inflammatory signaling can specify the dominant cell line in BM to counteract inflammation and leukemic condition via stimulating or inhibiting hematopoietic progenitors. Therefore, detection of the effects of inflammatory cytokines on the differentiation of HSCs can be an appropriate approach to check inflammatory and leukemic conditions and the suppression of these cytokines by their inhibitors allows for control of homeostasis in stressful conditions.
Collapse
Affiliation(s)
- Sahar Najafi
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Majid Ghanavat
- Child Growth and Development Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saied Shahrabi
- Department of Biochemistry and Hematology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
17
|
Tseng HW, Kulina I, Salga M, Fleming W, Vaquette C, Genêt F, Levesque JP, Alexander KA. Neurogenic Heterotopic Ossifications Develop Independently of Granulocyte Colony-Stimulating Factor and Neutrophils. J Bone Miner Res 2020; 35:2242-2251. [PMID: 32568412 DOI: 10.1002/jbmr.4118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/05/2020] [Accepted: 06/17/2020] [Indexed: 12/25/2022]
Abstract
Neurogenic heterotopic ossifications (NHOs) are incapacitating heterotopic bones in periarticular muscles that frequently develop following traumatic brain or spinal cord injuries (SCI). Using our unique model of SCI-induced NHO, we have previously established that mononucleated phagocytes infiltrating injured muscles are required to trigger NHO via the persistent release of the pro-inflammatory cytokine oncostatin M (OSM). Because neutrophils are also a major source of OSM, we investigated whether neutrophils also play a role in NHO development after SCI. We now show that surgery transiently increased granulocyte colony-stimulating factor (G-CSF) levels in blood of operated mice, and that G-CSF receptor mRNA is expressed in the hamstrings of mice developing NHO. However, mice defective for the G-CSF receptor gene Csf3r, which are neutropenic, have unaltered NHO development after SCI compared to C57BL/6 control mice. Because the administration of recombinant human G-CSF (rhG-CSF) has been trialed after SCI to increase neuroprotection and neuronal regeneration and has been shown to suppress osteoblast function at the endosteum of skeletal bones in human and mice, we investigated the impact of a 7-day rhG-CSF treatment on NHO development. rhG-CSF treatment significantly increased neutrophils in the blood, bone marrow, and injured muscles. However, there was no change in NHO development compared to saline-treated controls. Overall, our results establish that unlike monocytes/macrophages, neutrophils are dispensable for NHO development following SCI, and rhG-CSF treatment post-SCI does not impact NHO development. Therefore, G-CSF treatment to promote neuroregeneration is unlikely to adversely promote or affect NHO development in SCI patients. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Hsu-Wen Tseng
- Mater Research Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Irina Kulina
- Mater Research Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Marjorie Salga
- Mater Research Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia.,Department of Physical Medicine and Rehabilitation, Raymond Poincaré Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Garches, France
| | - Whitney Fleming
- Mater Research Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Cedryck Vaquette
- School of Dentistry, The University of Queensland, Herston, QLD, Australia.,Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia
| | - François Genêt
- Department of Physical Medicine and Rehabilitation, Raymond Poincaré Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Garches, France.,Evolution of Neuromuscular Diseases: Innovative Concepts and Practice (END:ICAP) U1179 Institut Natational de la Santé et de la Recherche Médicale, Unité de Formation et de Recherche Simone Veil-Santé, University of Versailles Saint Quentin en Yvelines, Montigny-le-Bretonneux, France
| | - Jean-Pierre Levesque
- Mater Research Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Kylie A Alexander
- Mater Research Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| |
Collapse
|
18
|
Cymer M, Brzezniakiewicz-Janus K, Bujko K, Thapa A, Ratajczak J, Anusz K, Tracz M, Jackowska-Tracz A, Ratajczak MZ, Adamiak M. Pannexin-1 channel "fuels" by releasing ATP from bone marrow cells a state of sterile inflammation required for optimal mobilization and homing of hematopoietic stem cells. Purinergic Signal 2020; 16:313-325. [PMID: 32533388 PMCID: PMC7524928 DOI: 10.1007/s11302-020-09706-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 05/21/2020] [Indexed: 12/19/2022] Open
Abstract
An efficient harvest of hematopoietic stem/progenitor cells (HSPCs) after pharmacological mobilization from the bone marrow (BM) into peripheral blood (PB) and subsequent proper homing and engraftment of these cells are crucial for clinical outcomes from hematopoietic transplants. Since extracellular adenosine triphosphate (eATP) plays an important role in both processes as an activator of sterile inflammation in the bone marrow microenvironment, we focused on the role of Pannexin-1 channel in the secretion of ATP to trigger both egress of HSPCs out of BM into PB as well as in reverse process that is their homing to BM niches after transplantation into myeloablated recipient. We employed a specific blocking peptide against Pannexin-1 channel and noticed decreased mobilization efficiency of HSPCs as well as other types of BM-residing stem cells including mesenchymal stroma cells (MSCs), endothelial progenitors (EPCs), and very small embryonic-like stem cells (VSELs). To explain better a role of Pannexin-1, we report that eATP activated Nlrp3 inflammasome in Gr-1+ and CD11b+ cells enriched for granulocytes and monocytes. This led to release of danger-associated molecular pattern molecules (DAMPs) and mitochondrial DNA (miDNA) that activate complement cascade (ComC) required for optimal egress of HSPCs from BM. On the other hand, Pannexin-1 channel blockage in transplant recipient mice leads to a defect in homing and engraftment of HSPCs. Based on this, Pannexin-1 channel as a source of eATP plays an important role in HSPCs trafficking.
Collapse
Affiliation(s)
- Monika Cymer
- Center for Preclinical Studies and Technology, Department of Regenerative Medicine, Medical University of Warsaw, ul. Żwirki i Wigury 61, 02-091, Warsaw, Poland
| | | | - Kamila Bujko
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Arjun Thapa
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Janina Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Krzysztof Anusz
- Institute of Veterinary Medicine, Department of Food Hygiene and Public Health Protection, Warsaw University of Life Sciences (WULS-SGGW), Warsaw, Poland
| | - Michał Tracz
- Institute of Veterinary Medicine, Department of Food Hygiene and Public Health Protection, Warsaw University of Life Sciences (WULS-SGGW), Warsaw, Poland
| | - Agnieszka Jackowska-Tracz
- Institute of Veterinary Medicine, Department of Food Hygiene and Public Health Protection, Warsaw University of Life Sciences (WULS-SGGW), Warsaw, Poland
| | - Mariusz Z Ratajczak
- Center for Preclinical Studies and Technology, Department of Regenerative Medicine, Medical University of Warsaw, ul. Żwirki i Wigury 61, 02-091, Warsaw, Poland
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Mateusz Adamiak
- Center for Preclinical Studies and Technology, Department of Regenerative Medicine, Medical University of Warsaw, ul. Żwirki i Wigury 61, 02-091, Warsaw, Poland.
| |
Collapse
|
19
|
Krambs JR, Abou Ezzi G, Yao JC, Link DC. Canonical signaling by TGF family members in mesenchymal stromal cells is dispensable for hematopoietic niche maintenance under basal and stress conditions. PLoS One 2020; 15:e0233751. [PMID: 32470079 PMCID: PMC7259882 DOI: 10.1371/journal.pone.0233751] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/12/2020] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stromal cells are an important component of the bone marrow hematopoietic niche. Prior studies showed that signaling from members of the transforming growth factor (TGF) superfamily in mesenchymal stromal cells is required for normal niche development. Here, we assessed the impact of TGF family signaling on niche maintenance and stress responses by deleting Smad4 in mesenchymal stromal cells at birth, thereby abrogating canonical TGF signaling. No alteration in the number or spatial organization of CXCL12-abundant reticular (CAR) cells, osteoblasts, or adipocytes was observed in Osx-Cre, Smad4fl/fl mice, and expression of key niche factors was normal. Basal hematopoiesis and stress erythropoiesis responses to acute hemolytic anemia were normal. TGF-β potently inhibits stromal CXCL12 expression in vitro; however, G-CSF induced decreases in bone marrow CXCL12 expression and subsequent hematopoietic stem/progenitor cell mobilization were normal in Osx-Cre, Tgfbr2fl/fl mice, in which all TGF-β signaling in mesenchymal stromal is lost. Finally, although a prior study showed that TGF-β enhances recovery from myeloablative therapy, hematopoietic recovery following single or multiple doses of 5-flurauracil were normal in Osx-Cre, Tgfbr2fl/fl mice. Collectively, these data suggest that TGF family member signaling in mesenchymal stromal cells is dispensable for hematopoietic niche maintenance under basal and stress conditions.
Collapse
Affiliation(s)
- Joseph Ryan Krambs
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Grazia Abou Ezzi
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Juo-Chin Yao
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Daniel C. Link
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States of America
- * E-mail:
| |
Collapse
|
20
|
Jahandideh B, Derakhshani M, Abbaszadeh H, Akbar Movassaghpour A, Mehdizadeh A, Talebi M, Yousefi M. The pro-Inflammatory cytokines effects on mobilization, self-renewal and differentiation of hematopoietic stem cells. Hum Immunol 2020; 81:206-217. [DOI: 10.1016/j.humimm.2020.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/29/2019] [Accepted: 01/13/2020] [Indexed: 02/08/2023]
|
21
|
Stackowicz J, Jönsson F, Reber LL. Mouse Models and Tools for the in vivo Study of Neutrophils. Front Immunol 2020; 10:3130. [PMID: 32038641 PMCID: PMC6985372 DOI: 10.3389/fimmu.2019.03130] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/23/2019] [Indexed: 12/21/2022] Open
Abstract
Neutrophils are the most abundant leukocytes in human blood and critical actors of the immune system. Many neutrophil functions and facets of their activity in vivo were revealed by studying genetically modified mice or by tracking fluorescent neutrophils in animals using imaging approaches. Assessing the roles of neutrophils can be challenging, especially when exact molecular pathways are questioned or disease states are interrogated that alter normal neutrophil homeostasis. This review discusses the main in vivo models for the study of neutrophils, their advantages and limitations. The side-by-side comparison underlines the necessity to carefully choose the right model(s) to answer a given scientific question, and exhibit caveats that need to be taken into account when designing experimental procedures. Collectively, this review suggests that at least two models should be employed to legitimately conclude on neutrophil functions.
Collapse
Affiliation(s)
- Julien Stackowicz
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France.,Sorbonne Université, Collège Doctoral, Paris, France
| | - Friederike Jönsson
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France
| | - Laurent L Reber
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France.,Center for Pathophysiology Toulouse-Purpan (CPTP), UMR 1043, University of Toulouse, INSERM, CNRS, Toulouse, France
| |
Collapse
|
22
|
G-CSF Inhibits Growths of Osteoblasts and Osteocytes by Upregulating Nitric Oxide Production in Neutrophils. J Craniofac Surg 2020; 30:e776-e780. [PMID: 31689739 DOI: 10.1097/scs.0000000000005769] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Granulocyte colony-stimulating factor (G-CSF) is the critical regulator of the proliferation, differentiation, and survival of granulocytes. Recently, it has been shown that G-CSF can adversely affect bone health in both animal models and patients. Here, the authors aimed to investigate whether G-CSF could inhibit the growth of osteoblasts and osteocytes by regulating nitric oxide. METHODS The C57BL/6 mice were divided into the control group, G-CSF treatment group and recovery group (G-CSF+L-NAME). The morphology of femurs was assessed by histology and immunohistochemistry. The expression of apoptosis-related molecules in femurs was detected by immunohistochemistry and quantitative RT-PCR, respectively. To examine if neutrophil-secreted factors can induce apoptosis in osteoblasts, Gr1-positive (Gr1+) neutrophils from the bone marrow of wild-type mice were sorted and co-cultured with MC3T3 pre-osteoblasts for 2 days. RESULTS The number of osteoblasts and newly embedding osteocytes significantly decreased and markers related to osteoblasts and osteocytes were downregulated in the G-CSF treatment compared to the control group. Moreover, G-CSF treatment did not change proliferation markers but induced apoptosis in osteoblast-lineage cells. The combined treatment of mice with G-CSF and a nitric oxide inhibitor partially restored the number of osteoblasts and osteocyte parameters. CONCLUSIONS The G-CSF can inhibit osteoblasts and osteocytes by upregulating nitric oxide.
Collapse
|
23
|
Abstract
Cytokines and hematopoietic growth factors have traditionally been thought of as regulators of the development and function of immune and blood cells. However, an ever-expanding number of these factors have been discovered to have major effects on bone cells and the development of the skeleton in health and disease (Table 1). In addition, several cytokines have been directly linked to the development of osteoporosis in both animal models and in patients. In order to understand the mechanisms regulating bone cells and how this may be dysregulated in disease states, it is necessary to appreciate the diverse effects that cytokines and inflammation have on osteoblasts, osteoclasts, and bone mass. This chapter provides a broad overview of this topic with extensive references so that, if desired, readers can access specific references to delve into individual topics in greater detail.
Collapse
Affiliation(s)
- Joseph Lorenzo
- Departments of Medicine and Orthopaedic Surgery, UConn Health, Farmington, CT, USA.
| |
Collapse
|
24
|
Zhang Z, Yuan W, Deng J, Wang D, Zhang T, Peng L, Tian H, Wang Z, Ma J. Granulocyte colony stimulating factor (G-CSF) regulates neutrophils infiltration and periodontal tissue destruction in an experimental periodontitis. Mol Immunol 2019; 117:110-121. [PMID: 31765840 DOI: 10.1016/j.molimm.2019.11.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/29/2019] [Accepted: 11/10/2019] [Indexed: 12/15/2022]
Abstract
Although granulocyte colony-stimulating factor(G-CSF) has pathogenic roles in several immune inflammatory diseases, its role in periodontitis has not been investigated. Here we detected local expression of G-CSF using public datasets in the Gene Expression Omnibus (GEO) database, and immune cell infiltration into gingival tissue was estimated based on single-sample gene set enrichment analysis (ssGSEA). G-CSF expression and neutrophil infiltration were also confirmed by human gingival biopsies analysis. Moreover, anti-G-CSF neutralizing antibody was locally administrated to investigate the effects of G-CSF neutralization on neutrophils infiltration and periodontal tissue destruction in periodontitis mice model. Two public datasets (GSE10334 and GSE16134), which included 424 patients with periodontitis and 133 health controls, were used in the analysis. Markedly increased immune cell infiltration and G-CSF expression in gingival tissues were found in the periodontitis group as compared to the control group. The higher expression of G-CSF was correlated with higher infiltration of immune cells, especially with neutrophil infiltration. Analysis of gingival biopsies further confirmed high neutrophil infiltration and G-CSF expression. In addition, anti-G-CSF antibody-treated mice with periodontitis showed significantly reduced alveolar bone resorption and neutrophil infiltration when compared with periodontitis mice treated with isotype control antibody. Also, anti-G-CSF antibody treatment significantly reduced mRNA expression of CXC chemokines (CXCL1, CXCL2 and CXCL3), interleukin 1β (IL-1β), IL-6, matrix metalloproteinases 9, receptor activator of nuclear factor κB ligand/osteoprotegerin (RANKL/OPG) ratio and osteoclasts number in periodontal tissues. In summary, neutrophil infiltration and G-CSF expression levels were significantly increased in inflamed gingival tissues. G-CSF neutralization in periodontal inflammation could alleviate neutrophil infiltration and periodontal tissue destruction in experimental periodontitis.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, 8th Gongti South Road, Beijing, 100020, China; Department of Periodontology, Tianjin Stomatological Hospital, Hospital of Stomatology, Nankai University, 75th Dagu North Road, Tianjin, 300000, China
| | - Wei Yuan
- State Key Laboratory of Molecular Oncology, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 17(th) Panjiayuan Nanli, Beijing, 100021, China
| | - Junjie Deng
- State Key Laboratory of Molecular Oncology, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 17(th) Panjiayuan Nanli, Beijing, 100021, China
| | - Danyang Wang
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, 8th Gongti South Road, Beijing, 100020, China
| | - Tianyi Zhang
- School of Stomatology, Shanxi Medical University, 56th Xinjian South Road, Taiyuan, 030001, China
| | - Li Peng
- Department of Stomatology, The Third People's Hospital of Datong City, 1th Wenchang Road, Datong, 037008, China
| | - Huan Tian
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, 8th Gongti South Road, Beijing, 100020, China
| | - Zuomin Wang
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, 8th Gongti South Road, Beijing, 100020, China.
| | - Jie Ma
- Department of Biotherapy, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences & Peking Union Medical College, 1th Dongdan Dahua Road, Beijing, 100730, China.
| |
Collapse
|
25
|
G-CSF partially mediates bone loss induced by Staphylococcus aureus infection in mice. Clin Sci (Lond) 2019; 133:1297-1308. [PMID: 31175224 DOI: 10.1042/cs20181001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 05/28/2019] [Accepted: 06/07/2019] [Indexed: 12/13/2022]
Abstract
Bone loss in Staphylococcus aureus (S. aureus) osteomyelitis poses a serious challenge to orthopedic treatment. The present study aimed to elucidate how S. aureus infection in bone might induce bone loss. The C57BL/6 mice were injected with S. aureus (106 CFU/ml, 100 μl) or with the same amount of vehicle (control) via the tail vein. Microcomputed tomography (microCT) analysis showed bone loss progressing from week 1 to week 5 after infection, accompanied by a decreased number of osteocalcin-positive stained osteoblasts and the suppressed mRNA expression of Runx2 and osteocalcin. Transcriptome profiles of GSE30119 were downloaded and analyzed to determine the differences in expression of inflammatory factors between patients with S. aureus infected osteomyelitis and healthy controls, the data showed significantly higher mRNA expression of granulocyte colony-stimulating factor (G-CSF) in the whole blood from patients with S. aureus infection. Enzyme-linked immunosorbent assay (ELISA) analysis confirmed an increased level of G-CSF in the bone marrow and serum from S. aureus infected mice, which might have been due to the increased amount of F4/80+ macrophages. Interestingly, G-CSF neutralizing antibody treatment significantly rescued the bone loss after S. aureus infection, as evidenced by its roles in improving BV/TV and preserving osteocalcin- and osterix-positive stained cells. Importantly, we found that G-CSF level was significantly up-regulated in the serum from osteomyelitis patients infected by S. aureus Together, S. aureus infection might suppress the function of osteoblastic cells and induce progressive bone loss by up-regulating the level G-CSF, suggesting a therapeutic potential for G-CSF neutralization in combating bone loss in S. aureus osteomyelitis.
Collapse
|
26
|
Katayama Y. Vitamin D receptor: A critical regulator of inter-organ communication between skeletal and hematopoietic systems. J Steroid Biochem Mol Biol 2019; 190:281-283. [PMID: 30731117 DOI: 10.1016/j.jsbmb.2019.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 02/01/2019] [Accepted: 02/03/2019] [Indexed: 11/28/2022]
Abstract
The functions of vitamin D receptor (VDR) have been extensively studied, for example, in the bone biology field. It is widely known that VDR knockout mice display the characteristic features of rickets type II. However, the contribution of VDR signaling to bone marrow (BM) hematopoiesis in association with the phenomena observed in clinical hematology has not been evaluated thoroughly. Hematopoietic stem cells (HSCs) can be mobilized from the BM into and harvested from peripheral blood as a BM transplantation source for the curable treatment of hematologic malignancies. This HSC mobilization can be achieved by the administration of cytokine granulocyte colony-stimulating factor (G-CSF) for several consecutive days. We have reported that, using the murine model, G-CSF induces the high sympathetic tone in the BM and a β2-adrenergic signal into osteoblasts induces a rapid and drastic increase of VDR, which is critical for the subsequent cascade for HSC mobilization. This is an example of the transient deviation of the inter-organ communication in three different systems (nervous, skeletal, and hematopoietic) bridged by VDR in osteoblasts. It would be important to reconsider VDR as a pivotal molecule that mediates inter-organ communication to broaden the application of vitamin D signal modulators.
Collapse
Affiliation(s)
- Yoshio Katayama
- Hematology, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, 650-0017, Kobe, Japan.
| |
Collapse
|
27
|
Iberl S, Meyer AL, Müller G, Peters S, Johannesen S, Kobor I, Beier F, Brümmendorf TH, Hart C, Schelker R, Herr W, Bogdahn U, Grassinger J. Effects of continuous high-dose G-CSF administration on hematopoietic stem cell mobilization and telomere length in patients with amyotrophic lateral sclerosis - a pilot study. Cytokine 2019; 120:192-201. [PMID: 31100684 DOI: 10.1016/j.cyto.2019.05.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 04/23/2019] [Accepted: 05/05/2019] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease of complex and still poorly understood etiology. Loss of upper and lower motoneurons results in death within few years after diagnosis. Recent studies have proposed neuroprotective and disease-slowing effects of granulocyte-colony stimulating factor (G-CSF) treatment in ALS mouse models as well as humans. In this study, six ALS patients were monitored up to 3.5 years during continuous high-dose G-CSF administration. Repetitive analyses were performed including blood count parameters, CD34+ hematopoietic stem and progenitor cell (HSPC) and colony forming cell (CFC) counts, serum cytokine levels and leukocyte telomere length. We demonstrate that continuous G-CSF therapy was well tolerated and safe resulting in only mild adverse events during the observation period. However, no mobilization of CD34+ HSPC was detected as compared to baseline values. CFC mobilization was equally low and even a decrease of myeloid precursors was observed in some patients. Assessment of telomere length within ALS patients' leukocytes revealed that G-CSF did not significantly shorten telomeres, while those of ALS patients were shorter compared to age-matched healthy controls, irrespective of G-CSF treatment. During G-CSF stimulation, TNF-alpha, CRP, IL-16, sVCAM-1, sICAM-1, Tie-2 and VEGF were significantly increased in serum whereas MCP-1 levels decreased. In conclusion, our data show that continuous G-CSF treatment fails to increase circulating CD34+ HSPC in ALS patients. Cytokine profiles revealed G-CSF-mediated immunomodulatory and proteolytic effects. Interestingly, despite intense G-CSF stimulation, telomere length was not significantly shortened.
Collapse
Affiliation(s)
- Sabine Iberl
- Department of Hematology and Oncology, Internal Medicine III, University Hospital Regensburg, Regensburg, Germany.
| | - Anne-Louise Meyer
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Gunnar Müller
- Department of Hematology and Oncology, Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Sebastian Peters
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Siw Johannesen
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Ines Kobor
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Fabian Beier
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, RWTH Aachen University Medical School, Aachen, Germany
| | - Tim H Brümmendorf
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, RWTH Aachen University Medical School, Aachen, Germany
| | - Christina Hart
- Department of Hematology and Oncology, Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Roland Schelker
- Department of Hematology and Oncology, Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Wolfgang Herr
- Department of Hematology and Oncology, Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Ulrich Bogdahn
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Jochen Grassinger
- Department of Hematology and Oncology, Internal Medicine III, University Hospital Regensburg, Regensburg, Germany; Department of Oncology and Hematology, St. Elisabeth Hospital, Straubing, Germany
| |
Collapse
|
28
|
de Kruijf EJFM, Fibbe WE, van Pel M. Cytokine-induced hematopoietic stem and progenitor cell mobilization: unraveling interactions between stem cells and their niche. Ann N Y Acad Sci 2019; 1466:24-38. [PMID: 31006885 PMCID: PMC7217176 DOI: 10.1111/nyas.14059] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/15/2019] [Accepted: 02/28/2019] [Indexed: 02/06/2023]
Abstract
Peripheral blood hematopoietic stem and progenitor cells (HSPCs), mobilized by granulocyte colony‐stimulating factor, are widely used as a source for both autologous and allogeneic stem cell transplantation. The use of mobilized HSPCs has several advantages over traditional bone marrow–derived HSPCs, including a less invasive harvesting process for the donor, higher HSPC yields, and faster hematopoietic reconstitution in the recipient. For years, the mechanisms by which cytokines and other agents mobilize HSPCs from the bone marrow were not fully understood. The field of stem cell mobilization research has advanced significantly over the past decade, with major breakthroughs in the elucidation of the complex mechanisms that underlie stem cell mobilization. In this review, we provide an overview of the events that underlie HSPC mobilization and address the relevant cellular and molecular components of the bone marrow niche. Furthermore, current and future mobilizing agents will be discussed.
Collapse
Affiliation(s)
- Evert-Jan F M de Kruijf
- Section of Stem Cell Biology, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Willem E Fibbe
- Section of Stem Cell Biology, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Melissa van Pel
- Section of Stem Cell Biology, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
29
|
Lucas D. Leukocyte Trafficking and Regulation of Murine Hematopoietic Stem Cells and Their Niches. Front Immunol 2019; 10:387. [PMID: 30891044 PMCID: PMC6412148 DOI: 10.3389/fimmu.2019.00387] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 02/14/2019] [Indexed: 12/28/2022] Open
Abstract
Hematopoietic stem cells (HSC) are the most powerful type of adult stem cell found in the body. Hematopoietic stem cells are multipotent and capable of giving rise to all other types of hematopoietic cells found in the organism. A single HSC is capable of regenerating a functional hematopoietic system when transplanted into a recipient. Hematopoietic stem cells reside in the bone marrow in specific multicellular structures called niches. These niches are indispensable for maintaining and regulating HSC numbers and function. It has become increasingly clearer that HSC and their niches can also be regulated by migrating leukocytes. Here we will discuss the composition of murine bone marrow niches and how HSC and their niches are regulated by different types of leukocytes that traffic between the periphery and the niche. Unless otherwise indicated all the studies discussed below were performed in mouse models.
Collapse
Affiliation(s)
- Daniel Lucas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
30
|
The effects of short-term use of granulocyte colony-stimulating factor on bone metabolism in child cancer patients. North Clin Istanb 2018; 5:277-281. [PMID: 30859156 PMCID: PMC6371986 DOI: 10.14744/nci.2017.59320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 12/08/2017] [Indexed: 01/27/2023] Open
Abstract
OBJECTIVE: The granulocyte colony-stimulating factor (G-CSF) is the most commonly used hematopoietic growth factor recombinant DNA technology. It affects bone metabolism by modulating both osteoclast and osteoblast functions. The aim of the present study was to investigate the effects of short-term use of G-CSF on bone metabolism in children with leukemia and solid tumors. METHODS: Thirty-six patients with a malignancy who received G-CSF therapy according to chemotherapy protocols and another 20 growth factor-free cancer patients who were enrolled as controls were included in the study. The serum osteocalcin and urinary free deoxypyridinoline levels were measured before the start of G-CSF therapy, on day 3 after treatment, and 7 days after G-CSF therapy was discontinued. In the control group, the measurements were made during corticosteroid and methotrexate-free chemotherapy. RESULTS: The mean osteocalcin level (8.6±2.3 ng/mL) from before the onset of treatment decreased significantly (7.7±2.3 ng/mL) on day 3 of G-CSF therapy and significantly increased after 7 days of G-CSF therapy (7.9±2.2 ng/mL) (p<0.001 and p<0.001, respectively), which was still significantly lower than the pre-G-CSF values (p<0.001). The urinary free deoxypyridinoline level significantly increased on day 3 of G-CSF treatment (25.6±6.5 nmol/mmol Cr) and significantly decreased after 7 days of G-CSF therapy (22.6±6.4 nmol/mmol Cr) (p<0.001 and p<0.001, respectively), which was still significantly higher than the values recorded before G-CSF therapy (p<0.001). CONCLUSION: The findings show that the short-term use of G-CSF in children with cancer can affect bone metabolism and can play a role in metabolic changes. Decreased osteoblastic activity and increased osteoclastic activity suggest that osteoporosis may be associated with bone pain in these patients.
Collapse
|
31
|
Arias CF, Herrero MA, Echeverri LF, Oleaga GE, López JM. Bone remodeling: A tissue-level process emerging from cell-level molecular algorithms. PLoS One 2018; 13:e0204171. [PMID: 30231062 PMCID: PMC6145577 DOI: 10.1371/journal.pone.0204171] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 09/03/2018] [Indexed: 01/08/2023] Open
Abstract
The human skeleton undergoes constant remodeling throughout the lifetime. Processes occurring on microscopic and molecular scales degrade bone and replace it with new, fully functional tissue. Multiple bone remodeling events occur simultaneously, continuously and independently throughout the body, so that the entire skeleton is completely renewed about every ten years.Bone remodeling is performed by groups of cells called Bone Multicellular Units (BMU). BMUs consist of different cell types, some specialized in the resorption of old bone, others encharged with producing new bone to replace the former. These processes are tightly regulated so that the amount of new bone produced is in perfect equilibrium with that of old bone removed, thus maintaining bone microscopic structure.To date, many regulatory molecules involved in bone remodeling have been identified, but the precise mechanism of BMU operation remains to be fully elucidated. Given the complexity of the signaling pathways already known, one may question whether such complexity is an inherent requirement of the process or whether some subset of the multiple constituents could fulfill the essential role, leaving functional redundancy to serve an alternative safety role. We propose in this work a minimal model of BMU function that involves a limited number of signals able to account for fully functional BMU operation. Our main assumptions were i) at any given time, any cell within a BMU can select only one among a limited choice of decisions, i.e. divide, die, migrate or differentiate, ii) this decision is irreversibly determined by depletion of an appropriate internal inhibitor and iii) the dynamics of any such inhibitor are coupled to that of specific external mediators, such as hormones, cytokines, growth factors. It was thus shown that efficient BMU operation manifests as an emergent process, which results from the individual and collective decisions taken by cells within the BMU unit in the absence of any external planning.
Collapse
Affiliation(s)
- Clemente F. Arias
- Grupo Interdisciplinar de Sistemas Complejos (GISC), Universidad Complutense, 28040 Madrid, Spain
| | - Miguel A. Herrero
- Departamento de Análisis Matemático y Matemática Aplicada, Facultad de Matemáticas, Universidad Complutense, 28040 Madrid, Spain
| | - Luis F. Echeverri
- Instituto de Matemáticas, Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia, 53108 Medellín, Colombia
| | - Gerardo E. Oleaga
- Departamento de Análisis Matemático y Matemática Aplicada, Facultad de Matemáticas, Universidad Complutense, 28040 Madrid, Spain
- Instituto de Matemática Interdisciplinar, Facultad de Matemáticas, Universidad Complutense, 28040 Madrid, Spain
| | - José M. López
- Departamento de Morfología y Biología Celular, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
| |
Collapse
|
32
|
Abstract
Bones provide both skeletal scaffolding and space for hematopoiesis in its marrow. Previous work has shown that these functions were tightly regulated by the nervous system. The central and peripheral nervous systems tightly regulate compact bone remodeling, its metabolism, and hematopoietic homeostasis in the bone marrow (BM). Accumulating evidence indicates that the nervous system, which fine-tunes inflammatory responses and alterations in neural functions, may regulate autoimmune diseases. Neural signals also influence the progression of hematological malignancies such as acute and chronic myeloid leukemias. Here, we review the interplay of the nervous system with bone, BM, and immunity, and discuss future challenges to target hematological diseases through modulation of activity of the nervous system.
Collapse
Affiliation(s)
- Maria Maryanovich
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Shoichiro Takeishi
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Paul S Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
33
|
Ubellacker JM, Baryawno N, Severe N, DeCristo MJ, Sceneay J, Hutchinson JN, Haider MT, Rhee CS, Qin Y, Gregory WM, Garrido-Castro AC, Holen I, Brown JE, Coleman RE, Scadden DT, McAllister SS. Modulating Bone Marrow Hematopoietic Lineage Potential to Prevent Bone Metastasis in Breast Cancer. Cancer Res 2018; 78:5300-5314. [PMID: 30065048 DOI: 10.1158/0008-5472.can-18-0548] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 06/12/2018] [Accepted: 07/23/2018] [Indexed: 12/20/2022]
Abstract
The presence of disseminated tumor cells in breast cancer patient bone marrow aspirates predicts decreased recurrence-free survival. Although it is appreciated that physiologic, pathologic, and therapeutic conditions impact hematopoiesis, it remains unclear whether targeting hematopoiesis presents opportunities for limiting bone metastasis. Using preclinical breast cancer models, we discovered that marrow from mice treated with the bisphosphonate zoledronic acid (ZA) are metastasis-suppressive. Specifically, ZA modulated hematopoietic myeloid/osteoclast progenitor cell (M/OCP) lineage potential to activate metastasis-suppressive activity. Granulocyte-colony stimulating factor (G-CSF) promoted ZA resistance by redirecting M/OCP differentiation. We identified M/OCP and bone marrow transcriptional programs associated with metastasis suppression and ZA resistance. Analysis of patient blood samples taken at randomization revealed that women with high-plasma G-CSF experienced significantly worse outcome with adjuvant ZA than those with lower G-CSF levels. Our findings support discovery of therapeutic strategies to direct M/OCP lineage potential and biomarkers that stratify responses in patients at risk of recurrence.Significance: Bone marrow myeloid/osteoclast progenitor cell lineage potential has a profound impact on breast cancer bone metastasis and can be modulated by G-CSF and bone-targeting agents. Cancer Res; 78(18); 5300-14. ©2018 AACR.
Collapse
Affiliation(s)
- Jessalyn M Ubellacker
- Hematology Division, Brigham & Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Ninib Baryawno
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts.,Center for Regenerative Medicine and the Cancer Center, Massachusetts General Hospital, Boston, Massachusetts.,Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Nicolas Severe
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts.,Center for Regenerative Medicine and the Cancer Center, Massachusetts General Hospital, Boston, Massachusetts.,Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Molly J DeCristo
- Hematology Division, Brigham & Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Jaclyn Sceneay
- Hematology Division, Brigham & Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - John N Hutchinson
- Department of Biostatistics, Harvard T.H. Chan, School of Public Health, Boston, Massachusetts
| | - Marie-Therese Haider
- Academic Unit of Clinical Oncology, Department of Oncology & Metabolism, Weston Park Hospital, University of Sheffield, Sheffield, United Kingdom
| | - Catherine S Rhee
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts.,Center for Regenerative Medicine and the Cancer Center, Massachusetts General Hospital, Boston, Massachusetts.,Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Yuanbo Qin
- Hematology Division, Brigham & Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Walter M Gregory
- Clinical Trials Research Unit, University of Leeds, Leeds, United Kingdom
| | - Ana C Garrido-Castro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ingunn Holen
- Academic Unit of Clinical Oncology, Department of Oncology & Metabolism, Weston Park Hospital, University of Sheffield, Sheffield, United Kingdom
| | - Janet E Brown
- Academic Unit of Clinical Oncology, Department of Oncology & Metabolism, Weston Park Hospital, University of Sheffield, Sheffield, United Kingdom
| | - Robert E Coleman
- Academic Unit of Clinical Oncology, Department of Oncology & Metabolism, Weston Park Hospital, University of Sheffield, Sheffield, United Kingdom
| | - David T Scadden
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts.,Center for Regenerative Medicine and the Cancer Center, Massachusetts General Hospital, Boston, Massachusetts.,Harvard Stem Cell Institute, Cambridge, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Sandra S McAllister
- Hematology Division, Brigham & Women's Hospital, Boston, Massachusetts. .,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Harvard Stem Cell Institute, Cambridge, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| |
Collapse
|
34
|
Tsantikos E, Lau M, Castelino CM, Maxwell MJ, Passey SL, Hansen MJ, McGregor NE, Sims NA, Steinfort DP, Irving LB, Anderson GP, Hibbs ML. Granulocyte-CSF links destructive inflammation and comorbidities in obstructive lung disease. J Clin Invest 2018; 128:2406-2418. [PMID: 29708507 DOI: 10.1172/jci98224] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 03/06/2018] [Indexed: 12/13/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is an incurable inflammatory lung disease that afflicts millions of people worldwide, and it is the fourth leading cause of death. Systemic comorbidities affecting the heart, skeletal muscle, bone, and metabolism are major contributors to morbidity and mortality. Given the surprising finding in large prospective clinical biomarker studies that peripheral white blood cell count is more closely associated with disease than inflammatory biomarkers, we probed the role of blood growth factors. Using the SHIP-1-deficient COPD mouse model, which manifests a syndrome of destructive lung disease and a complex of comorbid pathologies, we have identified a critical and unexpected role for granulocyte-CSF (G-CSF) in linking these conditions. Deletion of G-CSF greatly reduced airway inflammation and lung tissue destruction, and attenuated systemic inflammation, right heart hypertrophy, loss of fat reserves, and bone osteoporosis. In human clinical translational studies, bronchoalveolar lavage fluid of patients with COPD demonstrated elevated G-CSF levels. These studies suggest that G-CSF may play a central and unforeseen pathogenic role in COPD and its complex comorbidities, and identify G-CSF and its regulators as potential therapeutic targets.
Collapse
Affiliation(s)
- Evelyn Tsantikos
- Department of Immunology and Pathology, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia
| | - Maverick Lau
- Department of Immunology and Pathology, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia.,Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia
| | - Cassandra Mn Castelino
- Department of Immunology and Pathology, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia
| | - Mhairi J Maxwell
- Department of Immunology and Pathology, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia
| | - Samantha L Passey
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia
| | - Michelle J Hansen
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia
| | - Narelle E McGregor
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Natalie A Sims
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Daniel P Steinfort
- Department of Respiratory and Sleep Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Louis B Irving
- Department of Respiratory and Sleep Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Gary P Anderson
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia
| | - Margaret L Hibbs
- Department of Immunology and Pathology, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
35
|
Bacevic M, Brkovic B, Albert A, Rompen E, Radermecker RP, Lambert F. Does Oxidative Stress Play a Role in Altered Characteristics of Diabetic Bone? A Systematic Review. Calcif Tissue Int 2017; 101:553-563. [PMID: 29063963 DOI: 10.1007/s00223-017-0327-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 09/12/2017] [Indexed: 01/22/2023]
Abstract
Diabetes mellitus (DM) has been associated with increased bone fracture rates, impaired bone regeneration, delayed bone healing, and depressed osteogenesis. However, the plausible pathogenic mechanisms remain incompletely understood. The aim of the present systematic review was to investigate whether oxidative stress (OS) plays a role in altered characteristics of diabetic bone under in vivo conditions. An electronic search of the MEDLINE (via PubMed) and Embase databases was performed. In vivo animal studies involving DM and providing information regarding assessment of OS markers combined with analyses of bone histology/histomorphometry parameters were selected. A descriptive analysis of selected articles was performed. Ten studies were included in the present review. Both bone formation and bone resorption parameters were significantly decreased in the diabetic groups of animals compared to the healthy groups. This finding was consistent regardless of different animal/bone models employed or different evaluation periods. A statistically significant increase in systemic and/or local OS status was also emphasised in the diabetic groups in comparison to the healthy ones. Markers of OS were associated with histological and/or histomorphometric parameters, including decreased trabecular bone and osteoid volumes, suppressed bone formation, defective bone mineralisation, and reduced osteoclastic activity, in diabetic animals. Additionally, insulin and antioxidative treatment proved to be efficient in reversing the deleterious effects of high glucose and associated OS. The present findings support the hypotheses that OS in the diabetic condition contributes at least partially to defective bone features, and that antioxidative supplementation can be a valuable adjunctive strategy in treating diabetic bone disease, accelerating bone healing, and improving osteointegration.
Collapse
Affiliation(s)
- Miljana Bacevic
- Dental Biomaterials Research Unit (d-BRU), Faculty of Medicine, University of Liege, Liège, Belgium
- Clinic of Oral Surgery, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - Bozidar Brkovic
- Clinic of Oral Surgery, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - Adelin Albert
- Department of Biostatistics, University Hospital of Liege, Liège, Belgium
| | - Eric Rompen
- Department of Periodontology and Oral Surgery, Faculty of Medicine, University of Liege, Liège, Belgium
| | - Regis P Radermecker
- Department of Diabetes, Nutrition and Metabolic Disorders, University Hospital of Liege, Liège, Belgium
| | - France Lambert
- Dental Biomaterials Research Unit (d-BRU), Faculty of Medicine, University of Liege, Liège, Belgium.
- Department of Periodontology and Oral Surgery, Faculty of Medicine, University of Liege, Liège, Belgium.
- Service de Médecine Dentaire, Domaine du Sart Tilman Bat B-35, 4000, Liège, Belgium.
| |
Collapse
|
36
|
Hoggatt J, Kfoury Y, Scadden DT. Hematopoietic Stem Cell Niche in Health and Disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 11:555-81. [PMID: 27193455 DOI: 10.1146/annurev-pathol-012615-044414] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Regulation of stem cells in adult tissues is a key determinant of how well an organism can respond to the stresses of physiological challenge and disease. This is particularly true of the hematopoietic system, where demands on host defenses can call for an acute increase in cell production. Hematopoietic stem cells receive the regulatory signals for cell production in adult mammals in the bone marrow, a tissue with higher-order architectural and functional organization than previously appreciated. Here, we review the data defining particular structural components and heterologous cells in the bone marrow that participate in hematopoietic stem cell function. Further, we explore the case for stromal-hematopoietic cell interactions contributing to neoplastic myeloid disease. As the hematopoietic regulatory networks in the bone marrow are revealed, it is anticipated that strategies will emerge for how to enhance or inhibit production of specific blood cells. In that way, the control of hematopoiesis will enter the domain of therapies to modulate broad aspects of hematopoiesis, both normal and malignant.
Collapse
Affiliation(s)
- Jonathan Hoggatt
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138.,Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114;
| | - Youmna Kfoury
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138.,Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114;
| | - David T Scadden
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138.,Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114;
| |
Collapse
|
37
|
Morimoto A, Oh Y, Nakamura S, Shioda Y, Hayase T, Imamura T, Kudo K, Imashuku S. Inflammatory serum cytokines and chemokines increase associated with the disease extent in pediatric Langerhans cell histiocytosis. Cytokine 2017; 97:73-79. [PMID: 28582647 DOI: 10.1016/j.cyto.2017.05.026] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 05/23/2017] [Accepted: 05/27/2017] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Langerhans cell histiocytosis (LCH) is characterized by immature dendritic cell proliferation, infiltration of LCH lesions by various inflammatory cells, and a lesional cytokine storm. It is classified into three groups on the basis of disease extent, namely, multisystem with risk-organ involvement (MS+), multisystem without risk-organ involvement (MS-), and single-system (SS) disease. We comprehensively analyzed whether serum levels of cytokines/chemokines reflect the disease extent. METHODS Serum samples from 52 children with LCH (eight, 25, and 19 with MS+, MS-, and SS, respectively) and 34 control children were analyzed quantitatively for 48 humoral factors. DNA samples extracted from biopsied LCH lesions from 12 patients were tested for BRAF V600E status. RESULTS The LCH patients had significantly higher serum levels of IL-1Ra, IL-3, IL-6, IL-8, IL-9, IL-10, IL12, IL-13, IL-15, IL-17, IL-18, TNF-α, G-CSF, M-CSF, MIF, HGF, VEGF, CCL2, CCL3, CCL7, CXCL1, and CXCL9 than the controls by univariate analysis. Of these IL-9, IL-15 and MIF were significant by multivariate analysis; but not differed between MS and SS diseases. MS disease associated with significantly higher IL-2R, IL-3, IL-8, IL-18, M-CSF, HGF, CCL2, CXCL1, and CXCL9 levels than SS disease by univariate analysis. Of these, CCL2 and M-CSF were significant by multivariate analysis. IL-18 levels were significantly higher in MS+ disease than MS- disease. The LCH patients with BRAF V600E mutation had higher serum levels of CCL7. CONCLUSION Numerous inflammatory cytokines and chemokines play a role in LCH. Of those, more specific ones reflect the disease extent (MS vs. SS and MS+ vs. MS-) or the BRAF V600E mutation status. It is thought that the most responsible cytokines and chemokines involved in the poor outcome may become future candidate therapeutic targets in LCH.
Collapse
Affiliation(s)
- Akira Morimoto
- Department of Pediatrics, Jichi Medical University, School of Medicine, Shimotsuke, Japan.
| | - Yukiko Oh
- Department of Pediatrics, Jichi Medical University, School of Medicine, Shimotsuke, Japan
| | - Sachie Nakamura
- Department of Pediatrics, Jichi Medical University, School of Medicine, Shimotsuke, Japan
| | - Yoko Shioda
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Tomomi Hayase
- Department of Pediatrics, Jichi Medical University, School of Medicine, Shimotsuke, Japan
| | - Toshihiko Imamura
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuko Kudo
- Department of Pediatrics, Fujita Health University, Toyoake, Japan
| | - Shinsaku Imashuku
- Division of Laboratory Medicine, Uji-Tokushukai Medical Center, Uji, Japan
| | | |
Collapse
|
38
|
Li SD, Chen YB, Qiu LG, Qin MQ. G-CSF Indirectly Induces Apoptosis of Osteoblasts During Hematopoietic Stem Cell Mobilization. Clin Transl Sci 2017; 10:287-291. [PMID: 28556597 PMCID: PMC5504546 DOI: 10.1111/cts.12467] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 04/02/2017] [Indexed: 12/17/2022] Open
Abstract
The objective of this study was to explore the mechanism underlying osteoblast suppression in the process of hematopoietic stem cells mobilization induced by granulocyte colony‐stimulating factor (G‐CSF). The apoptosis of human and mouse osteoblasts was examined by detecting caspase 3. The levels of serum DKK1 and osteocalcin in the supernatant of co‐culture of mouse osteoblasts and mouse bone marrow nucleated cells were measured. The number of mouse osteoblasts co‐cultured with mouse bone marrow nucleated cells was measured and the osteocalcin mRNA level was also measured. The G‐CSF‐induced decrease in osteoblast function was partly due to the apoptosis of osteoblasts. There was no significant difference in the level of serum DKK1 in healthy donors before and 5 days after mobilization. The osteocalcin gene and protein expression was significantly different in co‐cultured osteoblasts with bone marrow nucleated cells treated with and without G‐CSF. Osteoblasts undergo apoptosis during mobilization and G‐CSF affects osteoblasts through bone marrow nucleated cells.
Collapse
Affiliation(s)
- S-D Li
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Ministry of Education, Key Laboratory of Major Diseases in Children, Ministry of Education, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, Beijing, P.R. China.,State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences and Peking Union of Medical College, Tianjin, P.R. China
| | - Y-B Chen
- Department of Hepatobiliary Surgery, PLA Army General Hospital, Beijing, P.R. China
| | - L-G Qiu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences and Peking Union of Medical College, Tianjin, P.R. China
| | - M-Q Qin
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Ministry of Education, Key Laboratory of Major Diseases in Children, Ministry of Education, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, Beijing, P.R. China
| |
Collapse
|
39
|
Continuous blockade of CXCR4 results in dramatic mobilization and expansion of hematopoietic stem and progenitor cells. Blood 2017; 129:2939-2949. [PMID: 28400375 DOI: 10.1182/blood-2016-10-746909] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 03/24/2017] [Indexed: 01/24/2023] Open
Abstract
Interaction between the chemokine receptor CXCR4 and its chief ligand CXCL12 plays a critical role in the retention and migration of hematopoietic stem and progenitor cells (HSPCs) in the bone marrow (BM) microenvironment. In this study, qualitative and quantitative effects of long-term pharmacologic inhibition of the CXCR4/CXCL12 axis on the HSPC compartment were investigated by using 3 structurally unrelated small molecule CXCR4 antagonists. A >10-fold increase in mobilization efficiency was achieved by administering the antagonists as a subcutaneous continuous infusion for 2 weeks compared to a single bolus injection. A concurrent increase in self-renewing proliferation leading to a twofold to fourfold expansion of the HSPC pool in the BM was observed. The expanded BM showed a distinct repopulating advantage when tested in serial competitive transplantation experiments. Furthermore, major changes within the HSPC niche associated with previously described HSPC expansion strategies were not detected in bones treated with a CXCR4 antagonist infusion. Our data suggest that prolonged but reversible pharmacologic blockade of the CXCR4/CXCL12 axis represents an approach that releases HSPC with efficiency superior to any other known mobilization strategy and may also serve as an effective method to expand the BM HSPC pool.
Collapse
|
40
|
Lobo RC, Hubbard NE, Damonte P, Mori H, Pénzváltó Z, Pham C, Koehne AL, Go AC, Anderson SE, Cala PM, Borowsky AD. Glucose Uptake and Intracellular pH in a Mouse Model of Ductal Carcinoma In situ (DCIS) Suggests Metabolic Heterogeneity. Front Cell Dev Biol 2016; 4:93. [PMID: 27630987 PMCID: PMC5005977 DOI: 10.3389/fcell.2016.00093] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 08/18/2016] [Indexed: 02/04/2023] Open
Abstract
Mechanisms for the progression of ductal carcinoma in situ (DCIS) to invasive breast carcinoma remain unclear. Previously we showed that the transition to invasiveness in the mammary intraepithelial neoplastic outgrowth (MINO) model of DCIS does not correlate with its serial acquisition of genetic mutations. We hypothesized instead that progression to invasiveness depends on a change in the microenvironment and that precancer cells might create a more tumor-permissive microenvironment secondary to changes in glucose uptake and metabolism. Immunostaining for glucose transporter 1 (GLUT1) and the hypoxia marker carbonic anhydrase 9 (CAIX) in tumor, normal mammary gland and MINO (precancer) tissue showed differences in expression. The uptake of the fluorescent glucose analog dye, 2-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl) amino]-2-deoxy-D-glucose (2-NBDG), reflected differences in the cellular distributions of glucose uptake in normal mammary epithelial cells (nMEC), MINO, and Met1 cancer cells, with a broad distribution in the MINO population. The intracellular pH (pHi) measured using the fluorescent ratio dye 2',7'-bis(2-carboxyethyl)-5(6)-155 carboxyfluorescein (BCECF) revealed expected differences between normal and cancer cells (low and high, respectively), and a mixed distribution in the MINO cells, with a subset of cells in the MINO having an increased rate of acidification when proton efflux was inhibited. Invasive tumor cells had a more alkaline baseline pHi with high rates of proton production coupled with higher rates of proton export, compared with nMEC. MINO cells displayed considerable variation in baseline pHi that separated into two distinct populations: MINO high and MINO low. MINO high had a noticeably higher mean acidification rate compared with nMEC, but relatively high baseline pHi similar to tumor cells. MINO low cells also had an increased acidification rate compared with nMEC, but with a more acidic pHi similar to nMEC. These findings demonstrate that MINO is heterogeneous with respect to intracellular pH regulation which may be associated with an acidified regional microenvironment. A change in the pH of the microenvironment might contribute to a tumor-permissive or tumor-promoting progression. We are not aware of any previous work showing that a sub-population of cells in in situ precancer exhibits a higher than normal proton production and export rate.
Collapse
Affiliation(s)
- Rebecca C Lobo
- Center for Comparative Medicine, University of California at Davis Davis, CA, USA
| | - Neil E Hubbard
- Center for Comparative Medicine, University of California at Davis Davis, CA, USA
| | - Patrizia Damonte
- Center for Comparative Medicine, University of California at Davis Davis, CA, USA
| | - Hidetoshi Mori
- Center for Comparative Medicine, University of California at Davis Davis, CA, USA
| | - Zsófia Pénzváltó
- Center for Comparative Medicine, University of California at Davis Davis, CA, USA
| | - Cindy Pham
- Department of Human Physiology and Membrane Biology, University of California at Davis Davis, CA, USA
| | - Amanda L Koehne
- Center for Comparative Medicine, University of California at Davis Davis, CA, USA
| | - Aiza C Go
- Center for Comparative Medicine, University of California at Davis Davis, CA, USA
| | - Steve E Anderson
- Department of Human Physiology and Membrane Biology, University of California at Davis Davis, CA, USA
| | - Peter M Cala
- Department of Human Physiology and Membrane Biology, University of California at Davis Davis, CA, USA
| | - Alexander D Borowsky
- Center for Comparative Medicine, University of California at DavisDavis, CA, USA; Department of Pathology, School of Medicine, University of California at DavisSacramento, CA, USA
| |
Collapse
|
41
|
Liu XL, Rao NJ, Huo L, Hu X, Lu WW, Zheng LW. The influence of locally applied granulocyte-colony stimulating factor on osteoporotic bone. Clin Oral Implants Res 2016; 28:586-593. [PMID: 27038192 DOI: 10.1111/clr.12839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2016] [Indexed: 12/25/2022]
Abstract
OBJECTIVES The compromised capacity of bone healing in osteoporotic population renders a serious concern of patients and clinicians. This study aimed to investigate the influence of G-CSF on bone reconstruction using an osteoporotic animal model. MATERIALS AND METHODS Sixty skeletal mature female Spraque-Dawley rats underwent bilateral ovariectomy (OVX) and were assigned into three groups (n = 20). Three months after OVX, defects of 5 mm in cranial and 2 mm in femur were surgically created on all the animals. The defects were left unfilled, filled with gelatin sponge (GS), or filled with granulocyte-colony stimulating factor (G-CSF) infused GS. Specimens were retrieved for histomorphometric and micro-CT analyses at weeks 1, 4, 8, and 12 after surgery. RESULTS At early stage of week 1 to week 8, the histomorphometric and micro-CT analysis demonstrated more advanced bone formation in femur in the control group; by week 12, all groups achieved cortical closure. In cranial bone, more advanced bone formation was exhibited in G-CSF-treated group at both early and late stages, although this observation was not statistically significant. CONCLUSIONS The results indicated that in osteoporotic bone, G-CSF may advance bone healing in cranial bone where spontaneous bone formation was insufficient.
Collapse
Affiliation(s)
- Xi Ling Liu
- Discipline of Oral Diagnosis and Polyclinics, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China.,Key Laboratory of Oral and Maxillofacial Tissue Engineering, Peking University Shenzhen Hospital, Shenzhen, China
| | - Nian Jing Rao
- Discipline of Oral Diagnosis and Polyclinics, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Lei Huo
- Discipline of Oral Diagnosis and Polyclinics, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Xiang Hu
- Discipline of Oral Diagnosis and Polyclinics, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Weijia William Lu
- Department of Orthopedics and Traumatology, The University of Hong Kong, Hong Kong SAR, China
| | - Li Wu Zheng
- Discipline of Oral Diagnosis and Polyclinics, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
42
|
Effect of Granulocyte-Colony Stimulating Factor on Endothelial Cells and Osteoblasts. BIOMED RESEARCH INTERNATIONAL 2016; 2016:8485721. [PMID: 27006951 PMCID: PMC4783536 DOI: 10.1155/2016/8485721] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 01/28/2016] [Accepted: 01/31/2016] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Some animal studies showed that granulocyte-colony stimulating factor (G-CSF) provides beneficial environment for bone healing. It has been well documented that endothelial cells and osteoblasts play critical roles in multiple phases of bone healing. However, the biological effects of G-CSF on these cells remain controversial. This study aimed to investigate the influence of G-CSF at various concentrations on endothelial cells and osteoblasts. MATERIALS AND METHODS Human umbilical vein endothelial cells (HUVECs) and human osteoblasts (hOBs) were treated with G-CSF at 1000, 100, 10, and 0 ng/mL, respectively. The capacity of cell proliferation, migration, and tube formation of HUVECs was evaluated at 72, 8, and 6 hours after treatment, respectively. The capacity of proliferation, differentiation, and mineralization of hOBs was evaluated at 24 hours, 72 hours, and 21 days after treatment, respectively. RESULTS HUVECs treated with 100 and 1000 ng/mL G-CSF showed a significantly higher value comparing with controls in migration assay (p < 0.001, p < 0.01, resp.); the group treated with 1000 ng/mL G-CSF showed a significantly lower value on tube formation. No significant difference was detected in groups of hOBs. CONCLUSIONS G-CSF showed favorable effects only on the migration of HUVECs, and no direct influence was found on hOBs.
Collapse
|
43
|
Gemery JM, Forauer AR, Silas AM, Hoffer EK. Hypersplenism in liver disease and SLE revisited: current evidence supports an active rather than passive process. BMC HEMATOLOGY 2016; 16:3. [PMID: 26865982 PMCID: PMC4748462 DOI: 10.1186/s12878-016-0042-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 01/20/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Active and passive theories have been advanced to explain splenomegaly and cytopenias in liver disease. Dameshek proposed active downregulation of hematopoiesis. Doan proposed passive trapping of blood components in a spleen enlarged by portal hypertension. Recent findings do not support a passive process. DISCUSSION Cytopenias and splenomegaly in both liver disease and systemic lupus erythematosus (SLE) poorly correlate with portal hypertension, and likely reflect an active process allocating stem cell resources in response to injury. Organ injury is repaired partly by bone-marrow-derived stem cells. Signaling would thus be needed to allocate resources between repair and routine marrow activities, hematologic and bone production. Granulocyte-colony stimulating factor (G-CSF) may play a central role: mobilizing stem cells, increasing spleen size and downregulating bone production. Serum G-CSF rises with liver injury, and is elevated in chronic liver disease and SLE. Signaling, not sequestration, likely accounts for splenomegaly and osteopenia in liver disease and SLE. The downregulation of a non-repair use of stem cells, bone production, suggests that repair efforts are prioritized. Other non-repair uses might be downregulated, namely hematologic production, as Dameshek proposed. SUMMARY Recognition that an active process may exist to allocate stem-cell resources would provide new approaches to diagnosis and treatment of cytopenias in liver disease, SLE and potentially other illnesses.
Collapse
Affiliation(s)
- John M. Gemery
- />Division of Interventional Radiology, Department of Radiology, Dartmouth-Hitchcock Medical Center, One Medical Center Drive, Lebanon, NH 03766 USA
- />Geisel School of Medicine at Dartmouth, One Rope Ferry Road, Hanover, NH 03755 USA
| | - Andrew R. Forauer
- />Geisel School of Medicine at Dartmouth, One Rope Ferry Road, Hanover, NH 03755 USA
| | - Anne M. Silas
- />Geisel School of Medicine at Dartmouth, One Rope Ferry Road, Hanover, NH 03755 USA
| | - Eric K. Hoffer
- />Geisel School of Medicine at Dartmouth, One Rope Ferry Road, Hanover, NH 03755 USA
| |
Collapse
|
44
|
Calvi LM, Link DC. The hematopoietic stem cell niche in homeostasis and disease. Blood 2015; 126:2443-51. [PMID: 26468230 PMCID: PMC4661168 DOI: 10.1182/blood-2015-07-533588] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 10/06/2015] [Indexed: 12/12/2022] Open
Abstract
The bone marrow microenvironment contains a heterogeneous population of stromal cells organized into niches that support hematopoietic stem cells (HSCs) and other lineage-committed hematopoietic progenitors. The stem cell niche generates signals that regulate HSC self-renewal, quiescence, and differentiation. Here, we review recent studies that highlight the heterogeneity of the stromal cells that comprise stem cell niches and the complexity of the signals that they generate. We highlight emerging data that stem cell niches in the bone marrow are not static but instead are responsive to environmental stimuli. Finally, we review recent data showing that hematopoietic niches are altered in certain hematopoietic malignancies, and we discuss how these alterations might contribute to disease pathogenesis.
Collapse
Affiliation(s)
- Laura M Calvi
- Department of Medicine and Pharmacology & Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY; and
| | - Daniel C Link
- Departments of Medicine and Pathology & Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
45
|
Cetean S, Căinap C, Constantin AM, Căinap S, Gherman A, Oprean L, Hangan A, Oprean R. The importance of the granulocyte-colony stimulating factor in oncology. ACTA ACUST UNITED AC 2015; 88:468-72. [PMID: 26732055 PMCID: PMC4689238 DOI: 10.15386/cjmed-531] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/14/2015] [Accepted: 09/14/2015] [Indexed: 12/24/2022]
Abstract
Granulocyte-colony stimulating factor (G-CSF) is a glycoprotein, the second CSF, sharing some common effects with granulocyte macrophage-colony stimulating factor (GM-CSF), interleukin-3 (IL-3) and interleukin-5 (IL-5). G-CSF is mainly produced by fibroblasts and endothelial cells from bone marrow stroma and by immunocompetent cells (monocytes, macrophages). The receptor for G-CSF (G-CSFR) is part of the cytokine and hematopoietin receptor superfamily and G-CSFR mutations cause severe congenital neutropenia. The main action of G-CSF - G-CSFR linkage is stimulation of the production, mobilization, survival and chemotaxis of neutrophils, but there are many other G-CSF effects: growth and migration of endothelial cells, decrease of norepinephrine reuptake, increase in osteoclastic activity and decrease in osteoblast activity. In oncology, G-CSF is utilized especially for the primary prophylaxis of chemotherapy-induced neutropenia, but it can be used for hematopoietic stem cell transplantation, it can produce monocytic differentiation of some myeloid leukemias and it can increase some drug resistance. The therapeutic indications of G-CSF are becoming more and more numerous: non neutropenic patients infections, reproductive medicine, neurological disturbances, regeneration therapy after acute myocardial infarction and of skeletal muscle, and hepatitis C therapy.
Collapse
Affiliation(s)
- Sînziana Cetean
- Department of General and Inorganic Chemistry, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania; Prof. Dr. Ion Chiricuta Oncology Institute, Cluj-Napoca, Romania
| | - Călin Căinap
- Prof. Dr. Ion Chiricuta Oncology Institute, Cluj-Napoca, Romania; Department of Oncology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Anne-Marie Constantin
- Department of Morphological Sciences, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Simona Căinap
- Department of Infant Care, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alexandra Gherman
- Prof. Dr. Ion Chiricuta Oncology Institute, Cluj-Napoca, Romania; Department of Oncology and Radiotherapy, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Luminiţa Oprean
- Department of General and Inorganic Chemistry, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Adriana Hangan
- Department of General and Inorganic Chemistry, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Radu Oprean
- Department of Analytical Chemistry, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
46
|
Asada N, Sato M, Katayama Y. Communication of bone cells with hematopoiesis, immunity and energy metabolism. BONEKEY REPORTS 2015; 4:748. [PMID: 26512322 DOI: 10.1038/bonekey.2015.117] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 08/28/2015] [Indexed: 12/20/2022]
Abstract
The bone contains the bone marrow. The functional communication between bone cells and hematopoiesis has been extensively studied in the past decade or so. Osteolineage cells and their modulators, such as the sympathetic nervous system, macrophages and osteoclasts, form a complex unit to maintain the homeostasis of hematopoiesis, called the 'microenvironment'. Recently, bone-embedded osteocytes, the sensors of gravity and mechanical stress, have joined the microenvironment, and they are demonstrated to contribute to whole body homeostasis through the control of immunity and energy metabolism. The inter-organ communication orchestrated by the bone is summarized in this article.
Collapse
Affiliation(s)
- Noboru Asada
- Division of Hematology, Department of Medicine, Kobe University Graduate School of Medicine , Kobe, Japan
| | - Mari Sato
- Division of Hematology, Department of Medicine, Kobe University Graduate School of Medicine , Kobe, Japan
| | - Yoshio Katayama
- Division of Hematology, Department of Medicine, Kobe University Graduate School of Medicine , Kobe, Japan ; Department of Hematology, Kobe University Hospital , Kobe, Japan ; PRESTO, Japan Science and Technology Agency , Kawaguchi, Japan
| |
Collapse
|
47
|
Uy GL, Hsu YMS, Schmidt AP, Stock W, Fletcher TR, Trinkaus KM, Westervelt P, DiPersio JF, Link DC. Targeting bone marrow lymphoid niches in acute lymphoblastic leukemia. Leuk Res 2015; 39:1437-42. [PMID: 26467815 DOI: 10.1016/j.leukres.2015.09.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 09/15/2015] [Accepted: 09/28/2015] [Indexed: 01/24/2023]
Abstract
In acute lymphoblastic leukemia (ALL) the bone marrow microenvironment provides growth and survival signals that may confer resistance to chemotherapy. Granulocyte colony-stimulating factor (G-CSF) potently inhibits lymphopoiesis by targeting stromal cells that comprise the lymphoid niche in the bone marrow. To determine whether lymphoid niche disruption by G-CSF sensitizes ALL cells to chemotherapy, we conducted a pilot study of G-CSF in combination with chemotherapy in patients with relapsed or refractory ALL. Thirteen patients were treated on study; three patients achieved a complete remission (CR/CRi) for an overall response rate of 23%. In the healthy volunteers, G-CSF treatment disrupted the lymphoid niche, as evidenced by reduced expression of CXCL12, interleukin-7, and osteocalcin. However, in most patients with relapsed/refractory ALL expression of these genes was markedly suppressed at baseline. Thus, although G-CSF treatment was associated with ALL cell mobilization into the blood, and increased apoptosis of bone marrow resident ALL cells, alterations in the bone marrow microenvironment were modest and highly variable. These data suggest that disruption of lymphoid niches by G-CSF to sensitize ALL cells to chemotherapy may be best accomplished in the consolidation where the bone marrow microenvironment is more likely to be normal.
Collapse
Affiliation(s)
- Geoffrey L Uy
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, United States
| | - Yen-Michael S Hsu
- Department of Pathology, Washington University School of Medicine, St. Louis, MO, United States
| | - Amy P Schmidt
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, United States
| | - Wendy Stock
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, United States
| | - Theresa R Fletcher
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, United States
| | - Kathryn M Trinkaus
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States
| | - Peter Westervelt
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, United States
| | - John F DiPersio
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, United States
| | - Daniel C Link
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, United States.
| |
Collapse
|
48
|
Li S, Zou D, Li C, Meng H, Sui W, Feng S, Cheng T, Zhai Q, Qiu L. Targeting stem cell niche can protect hematopoietic stem cells from chemotherapy and G-CSF treatment. Stem Cell Res Ther 2015; 6:175. [PMID: 26373707 PMCID: PMC4572669 DOI: 10.1186/s13287-015-0164-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 01/25/2015] [Accepted: 08/19/2015] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Hematopoietic stem/progenitor cells (HSPCs) reside in a tightly controlled local microenvironment called bone marrow niche. The specialized microenvironment or niche not only provides a favorable habitat for HSPC maintenance and development but also governs stem cell function. METHOD We investigated the effect of cytotoxic drugs on bone marrow niche. To mimic the multiple rounds of chemotherapy followed by autologous hematopoietic stem cells (HSCs) transplantation in a clinical setting, we further verified the hypothesis that targeting the niche might improve stem cell-based therapies in mouse models. RESULTS We found that multiple rounds of cytotoxic drug treatment significantly disrupted niche and serum osteocalcin level was significantly reduced after treatment in autologous HSPCs transplanted patients (P = 0.01). In mouse models, the number of CD45(-)Ter119(-)OPN(+) osteoblasts was significantly reduced after multiple rounds of chemotherapies and granulocyte colony stimulating factor (G-CSF) treatment (P < 0.01). Parathyroid hormone (PTH) or receptor activator of nuclear factor kappa-B ligand (RANKL) treatment significantly increased the number of HSCs mobilized into peripheral blood (PB) for stem cell harvesting and protected stem cells from repeated exposure to cytotoxic chemotherapy. Treatments with G-CSF and PTH significantly increased the preservation of the HSC pool (P < 0.05). Moreover, recipient mice transplanted with circulation HSPCs that were previously treated with PTH and RANKL showed robust myeloid and lymphatic cell engraftment compared to the mice transplanted with HSCs after chemotherapy or G-CSF treatment. CONCLUSION These data provide new evidence that the niche may be an important target for drug-based stem cell therapy.
Collapse
Affiliation(s)
- Sidan Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences and Peking Union of Medical College, 288 Nanjing Road, Tianjin, 30020, China. .,Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Ministry of Education, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, Beijing, China.
| | - Dehui Zou
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences and Peking Union of Medical College, 288 Nanjing Road, Tianjin, 30020, China.
| | - Changhong Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences and Peking Union of Medical College, 288 Nanjing Road, Tianjin, 30020, China.
| | - Hengxing Meng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences and Peking Union of Medical College, 288 Nanjing Road, Tianjin, 30020, China.
| | - Weiwei Sui
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences and Peking Union of Medical College, 288 Nanjing Road, Tianjin, 30020, China.
| | - Sizhou Feng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences and Peking Union of Medical College, 288 Nanjing Road, Tianjin, 30020, China.
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences and Peking Union of Medical College, 288 Nanjing Road, Tianjin, 30020, China.
| | - Qiongli Zhai
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences and Peking Union of Medical College, 288 Nanjing Road, Tianjin, 30020, China.
| |
Collapse
|
49
|
Klamer S, Voermans C. The role of novel and known extracellular matrix and adhesion molecules in the homeostatic and regenerative bone marrow microenvironment. Cell Adh Migr 2015; 8:563-77. [PMID: 25482635 PMCID: PMC4594522 DOI: 10.4161/19336918.2014.968501] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Maintenance of haematopoietic stem cells and differentiation of committed progenitors occurs in highly specialized niches. The interactions of haematopoietic stem and progenitor cells (HSPCs) with cells, growth factors and extracellular matrix (ECM) components of the bone marrow (BM) microenvironment control homeostasis of HSPCs. We only start to understand the complexity of the haematopoietic niche(s) that comprises endosteal, arterial, sinusoidal, mesenchymal and neuronal components. These distinct niches produce a broad range of soluble factors and adhesion molecules that modulate HSPC fate during normal hematopoiesis and BM regeneration. Adhesive interactions between HSPCs and the microenvironment will influence their localization and differentiation potential. In this review we highlight the current understanding of the functional role of ECM- and adhesion (regulating) molecules in the haematopoietic niche during homeostatic and regenerative hematopoiesis. This knowledge may lead to the improvement of current cellular therapies and more efficient development of future cellular products.
Collapse
Affiliation(s)
- Sofieke Klamer
- a Department of Hematopoiesis; Sanquin Research; Landsteiner Laboratory; Academic Medical Centre ; University of Amsterdam ; Amsterdam , The Netherlands
| | | |
Collapse
|
50
|
Hanoun M, Maryanovich M, Arnal-Estapé A, Frenette PS. Neural regulation of hematopoiesis, inflammation, and cancer. Neuron 2015; 86:360-73. [PMID: 25905810 DOI: 10.1016/j.neuron.2015.01.026] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although the function of the autonomic nervous system (ANS) in mediating the flight-or-fight response was recognized decades ago, the crucial role of peripheral innervation in regulating cell behavior and response to the microenvironment has only recently emerged. In the hematopoietic system, the ANS regulates stem cell niche homeostasis and regeneration and fine-tunes the inflammatory response. Additionally, emerging data suggest that cancer cells take advantage of innervating neural circuitry to promote their progression. These new discoveries outline the need to redesign therapeutic strategies to target this underappreciated stromal constituent. Here, we review the importance of neural signaling in hematopoietic homeostasis, inflammation, and cancer.
Collapse
Affiliation(s)
- Maher Hanoun
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Maria Maryanovich
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Anna Arnal-Estapé
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Paul S Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|