1
|
Lit KK, Zhirenova Z, Blocki A. Insulin-like growth factor-binding protein 7 (IGFBP7): A microenvironment-dependent regulator of angiogenesis and vascular remodeling. Front Cell Dev Biol 2024; 12:1421438. [PMID: 39045455 PMCID: PMC11263173 DOI: 10.3389/fcell.2024.1421438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/10/2024] [Indexed: 07/25/2024] Open
Abstract
Insulin-like Growth Factor-Binding Protein 7 (IGFBP7) is an extracellular matrix (ECM) glycoprotein, highly enriched in activated vasculature during development, physiological and pathological tissue remodeling. Despite decades of research, its role in tissue (re-)vascularization is highly ambiguous, exhibiting pro- and anti-angiogenic properties in different tissue remodeling states. IGFBP7 has multiple binding partners, including structural ECM components, cytokines, chemokines, as well as several receptors. Based on current evidence, it is suggested that IGFBP7's bioactivity is strongly dependent on the microenvironment it is embedded in. Current studies indicate that during physiological angiogenesis, IGFBP7 promotes endothelial cell attachment, luminogenesis, vessel stabilization and maturation. Its effects on other stages of angiogenesis and vessel function remain to be determined. IGFBP7 also modulates the pro-angiogenic properties of other signaling factors, such as VEGF-A and IGF, and potentially acts as a growth factor reservoir, while its actual effects on the factors' signaling may depend on the environment IGFBP7 is embedded in. Besides (re-)vascularization, IGFBP7 clearly promotes progenitor and stem cell commitment and may exhibit anti-inflammatory and anti-fibrotic properties. Nonetheless, its role in inflammation, immunomodulation, fibrosis and cellular senescence is again likely to be context-dependent. Future studies are required to shed more light on the intricate functioning of IGFBP7.
Collapse
Affiliation(s)
- Kwok Keung Lit
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine (CNRM), Hong Kong Science Park, Shatin, Hong Kong SAR, China
| | - Zhamilya Zhirenova
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine (CNRM), Hong Kong Science Park, Shatin, Hong Kong SAR, China
| | - Anna Blocki
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine (CNRM), Hong Kong Science Park, Shatin, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| |
Collapse
|
2
|
Shoeib SM, Hassan A, Habeeb E, Ragab RA, Elakshar S, Sherief D. Urinary Insulin-Like Growth Factor-Binding Protein 7 (IGFBp7), Urinary Tissue Inhibitor of Matrix Metalloproteinase 2 (TIMP2), and Serum Transgelin as Novel Biomarkers of Kidney Injury in Multiple Myeloma. Indian J Hematol Blood Transfus 2024; 40:246-254. [PMID: 38708150 PMCID: PMC11065805 DOI: 10.1007/s12288-023-01701-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 10/04/2023] [Indexed: 05/07/2024] Open
Abstract
Renal dysfunction is a common complication of MM and is associated with poor prognosis, particularly when progressive. Early identification of renal dysfunction is essential for prompt treatment for disease control and restoration of renal function. Urinary insulin-like growth factor-binding protein 7 (IGFBP-7), urinary tissue inhibitor of matrix metalloproteinase 2 (TIMP-2), and serum transgelin levels were measured using enzyme-linked immunosorbent assays and evaluated as biomarkers for the prediction of renal impairment in patients with multiple myeloma. U IGFBP-7/creatinine ratio, U TIMP2/creatinine ratio, and serum transgelin levels were higher in patients with MM than healthy controls, and predicted renal insufficiency in MM. Serum transgelin, urinary IGFBp7, and TIMP2 levels may have utility as biomarkers of renal tubular injury and predict future renal impairment in patients with MM.
Collapse
Affiliation(s)
- Sarah M. Shoeib
- Department of Clinical Pathology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Asmaa Hassan
- Department of Clinical Pathology, Faculty of Medicine, Kafr El-Sheikh University, Kafr El-Sheikh, Egypt
| | - Eman Habeeb
- Department of Clinical Pathology, Faculty of Medicine, Kafr El-Sheikh University, Kafr El-Sheikh, Egypt
| | - Rasha Abdallah Ragab
- Department of Internal Medicine, Faculty of Medicine, Kafr El-Sheikh University, Kafr El-Sheikh, Egypt
| | - Sara Elakshar
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Dalia Sherief
- Department of Clinical Pathology, Faculty of Medicine, Kafr El-Sheikh University, Kafr El-Sheikh, Egypt
| |
Collapse
|
3
|
Xu C, Gao M, Zhang J, Fu Y. IL5RA as an immunogenic cell death-related predictor in progression and therapeutic response of multiple myeloma. Sci Rep 2023; 13:8528. [PMID: 37236993 DOI: 10.1038/s41598-023-35378-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Previous studies have shown the potential of immunogenic cell death-related modalities in myeloma. The significance of IL5RA in myeloma and immunogenic cell death remains unknown. We analyzed IL5RA expression, the gene expression profile, and secretory protein genes related to IL5RA level using GEO data. Immunogenic cell death subgroup classification was performed using the ConsensusClusterPlus and pheatmap R package. Enrichment analyses were based on GO/KEGG analysis. After IL5RA-shRNA transfection in myeloma cells, cell proliferation, apoptosis, and drug sensitivity were detected. P < 0.05 was considered statistically significant. IL5RA was upregulated in myeloma and progressed smoldering myeloma. We observed enrichment in pathways such as the PI3K-Akt signaling pathway, and Natural killer cell mediated cytotoxicity in the high-IL5RA group. IL5RA was also closely associated with secretory protein genes such as CST6. We observed the enrichment of cellular apoptosis and hippo signaling pathway on differential genes in the immunogenic cell death cluster. Furthermore, IL5RA was associated with immune infiltration, immunogenic cell death-related genes, immune-checkpoint-related genes, and m6A in myeloma. In vitro and in vivo experiments showed the involvement of IL5RA in apoptosis, proliferation, and drug resistance of myeloma cells. IL5RA shows the potential to be an immunogenic cell death-related predictor for myeloma.
Collapse
Affiliation(s)
- Cong Xu
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Meng Gao
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Junhua Zhang
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China.
| | - Yunfeng Fu
- Department of Hematology, The Third Xiangya Hospital of Central South University, Changsha, China.
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
4
|
Xia H, Tian Y, Lin Y, Huang Q, Xue Y. Evaluating Osteogenic Differentiation of Osteoblastic Precursors Upon Intermittent Administration of PTH/IGFBP7. Front Pharmacol 2022; 13:839035. [PMID: 35462909 PMCID: PMC9019492 DOI: 10.3389/fphar.2022.839035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 03/23/2022] [Indexed: 11/17/2022] Open
Abstract
Parathyroid hormone (PTH) 1–34 is the first anabolic agent approved for the treatment of osteoporosis. Preclinical evidence shows a potential association between PTH and osteosarcoma. The mechanisms mediating the bone- and neoplasm-forming effects of PTH remain incompleted understood, few studies on the role of Insulin-like growth factor-binding protein 7 (IGFBP7) in mediating the anabolic effects of PTH has been reported. Intermittent PTH administration was found to increase the expression of IGFBP7 in mesenchymal stem cells (MSCs) and pre-osteoblasts. The results indicated that the anabolic effects of PTH were interrupted when knockdown of IGFBP7, while supplementation with IGFBP7 protein could enhance the bone-forming efficacy of PTH and regulate the signaling pathways. Moreover, bone healing was accelerated by the administration of IGFBP7 along with PTH in a mouse model of fracture. The obtained results proved that IGFBP7 was necessary for the anabolic effects of PTH, and combined administration of PTH and IGFBP7 showed stronger bone-forming effects relative to administration of PTH alone.
Collapse
Affiliation(s)
- Han Xia
- Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yueyang Tian
- Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yile Lin
- Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Qia Huang
- Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuan Xue
- Tianjin Key Laboratory of Spine and Spinal Cord, Department of Orthopedic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
5
|
Woziwodzka K, Małyszko J, Koc-Żórawska E, Żórawski M, Dumnicka P, Jurczyszyn A, Batko K, Mazur P, Banaszkiewicz M, Krzanowski M, Gołasa P, Małyszko JA, Drożdż R, Krzanowska K. Renal Impairment Detectors: IGFBP-7 and NGAL as Tubular Injury Markers in Multiple Myeloma Patients. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:1348. [PMID: 34946293 PMCID: PMC8703512 DOI: 10.3390/medicina57121348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/03/2021] [Accepted: 12/08/2021] [Indexed: 02/07/2023]
Abstract
Background and Objectives: Urine insulin-like growth factor-binding protein 7 (IGFBP-7), tissue inhibitor of matrix metalloproteinase 2 (TIMP-2), and neutrophil gelatinase-associated lipocalin (NGAL) monomer are novel tubular kidney injury biomarkers. In multiple myeloma (MM), immunoglobulin free light chains (FLCs) play an integral role in renal impairment. This study aimed to investigate the correlation between new biomarkers and acclaimed parameters of renal failure, MM stage, and prognosis. Materials and Methods: The examined parameters included: urinary and serum cystatin-C, IGFBP-7, and TIMP-2, and urinary NGAL monomer in 124 enrolled patients. Results: Urinary and serum IGFBP-7 and urinary NGAL were higher among patients with an estimated glomerular filtration rate (eGFR) < 60 mL/min/1.73 m2, and positively correlated with urine light chains. Serum and urine IGFBP-7 and urine NGAL were greater among patients with a higher disease stage. In the whole study group, urinary concentrations of the studied markers were positively correlated with each other. In multiple linear regression, urinary IGFBP-7 and NGAL were associated with lower eGFR, independently of other urinary markers. Conclusions: Urinary IGFBP-7 and NGAL monomer may be useful markers of tubular renal damage in patients with MM. Biomarker-based diagnostics may contribute to earlier treatment that may improve renal outcomes and life expectancy in MM.
Collapse
Affiliation(s)
- Karolina Woziwodzka
- Department of Nephrology, Jagiellonian University Medical College, 30-688 Kraków, Poland; (K.W.); (K.B.); (M.B.); (M.K.); (P.G.)
| | - Jolanta Małyszko
- Department of Nephrology, Dialysis and Internal Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | - Ewa Koc-Żórawska
- Second Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Bialystok, 15-276 Bialystok, Poland;
| | - Marcin Żórawski
- Department of Clinical Medicine, Medical University of Bialystok, 15-254 Bialystok, Poland;
| | - Paulina Dumnicka
- Department of Medical Diagnostics, Jagiellonian University Medical College, 30-688 Kraków, Poland; (P.D.); (P.M.); (R.D.)
| | - Artur Jurczyszyn
- Departament of Hematology, Jagiellonian University Medical College, Kopernika 17, 30-501 Kraków, Poland;
| | - Krzysztof Batko
- Department of Nephrology, Jagiellonian University Medical College, 30-688 Kraków, Poland; (K.W.); (K.B.); (M.B.); (M.K.); (P.G.)
| | - Paulina Mazur
- Department of Medical Diagnostics, Jagiellonian University Medical College, 30-688 Kraków, Poland; (P.D.); (P.M.); (R.D.)
| | - Małgorzata Banaszkiewicz
- Department of Nephrology, Jagiellonian University Medical College, 30-688 Kraków, Poland; (K.W.); (K.B.); (M.B.); (M.K.); (P.G.)
| | - Marcin Krzanowski
- Department of Nephrology, Jagiellonian University Medical College, 30-688 Kraków, Poland; (K.W.); (K.B.); (M.B.); (M.K.); (P.G.)
| | - Paulina Gołasa
- Department of Nephrology, Jagiellonian University Medical College, 30-688 Kraków, Poland; (K.W.); (K.B.); (M.B.); (M.K.); (P.G.)
| | - Jacek A. Małyszko
- First Department of Nephrology and Transplantology with Dialysis Unit, Medical University of Bialystok, 15-540 Bialystok, Poland;
| | - Ryszard Drożdż
- Department of Medical Diagnostics, Jagiellonian University Medical College, 30-688 Kraków, Poland; (P.D.); (P.M.); (R.D.)
| | - Katarzyna Krzanowska
- Department of Nephrology, Jagiellonian University Medical College, 30-688 Kraków, Poland; (K.W.); (K.B.); (M.B.); (M.K.); (P.G.)
| |
Collapse
|
6
|
Dong X, Lu G, Su X, Liu J, Chen X, Tian Y, Chang Y, Wang L, Wang W, Zhou J. Identification of key miRNA signature and pathways involved in multiple myeloma by integrated bioinformatics analysis. Hematology 2021; 26:976-984. [PMID: 34871535 DOI: 10.1080/16078454.2021.2003980] [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: 10/19/2022] Open
Abstract
Multiple myeloma (MM) is one of the most common types of hematologic malignancy for which the underlying molecular mechanisms remain largely unclear. Dysregulated miRNA expression has been shown to be involved in MM tumorigenesis, progression and drug response. Therefore, a comprehensive analysis based on miRNA-level integrated strategy was performed. This study aimed to elucidate key miRNA signatures and pathways in MM by integrated bioinformatics analysis. Expression profiles GSE24371, GSE49261 and GSE54156 were obtained from the Gene Expression Omnibus database, and differentially expressed miRNAs (DEMirs) with p < 0.05 were identified. The target genes of these DEMirs were obtained from ENCORI database, and functional enrichment, subpathway enrichment and protein-protein interaction network construction were performed. The key target genes were identified by random walk algorithm and survival verification was performed. and discussion: First, six up-regulated and four down-regulated DEMirs shared between any two GSE data sets were identified. Second, target genes (DEMirTGs) by up-regulated and down-regulated DEMirs were obtained. Functional and subpathway enrichment analysis showed that these up-regulated DEMirs are consistently involved in the Wnt signaling pathway. Moreover, enrichment of the down-regulated DEMirs is mainly in the MAPK signaling pathway. Finally, a protein-protein interaction sub-network for these DEMirTGs was constructed, the correlations between the two key genes were identified and survival in MM was evaluated using multiple independent data sets. We identified miRNA signatures and key target genes that were closely related to MM biology, and these genes might serve as potential therapeutic targets for MM patients.
Collapse
Affiliation(s)
- Xiushuai Dong
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China.,Department of Hematology, The Second Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Gang Lu
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Xianwei Su
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Jie Liu
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Xi Chen
- Department of Hematology, The Second Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Yaoyao Tian
- Department of Hematology, The Second Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Yuying Chang
- Department of Hematology, The Second Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Lianjie Wang
- Department of Hematology, The Second Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Wei Wang
- Department of Hematology, The Second Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Jin Zhou
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| |
Collapse
|
7
|
Abboud A, Kui N, Gaggin HK, Ibrahim NE, Chen-Tournoux AA, Christenson RH, Hollander JE, Levy PD, Nagurney JT, Nowak RM, Pang PS, Peacock WF, Walters EL, Januzzi JL. Multiple Cardiac Biomarker Testing Among Patients With Acute Dyspnea From the ICON-RELOADED Study. J Card Fail 2021; 28:226-233. [PMID: 34634446 DOI: 10.1016/j.cardfail.2021.08.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 01/23/2023]
Abstract
BACKGROUND Among patients with acute dyspnea, concentrations of N-terminal pro-B-type natriuretic peptide (NT-proBNP), high-sensitivity cardiac troponin T, and insulin-like growth factor binding protein-7 predict cardiovascular outcomes and death. Understanding the optimal means to interpret these elevated biomarkers in patients presenting with acute dyspnea remains unknown. METHODS AND RESULTS Concentrations of NT-proBNP, high-sensitivity cardiac troponin T, and insulin-like growth factor binding protein-7 were analyzed in 1448 patients presenting with acute dyspnea from the prospective, multicenter International Collaborative of NT-proBNP-Re-evaluation of Acute Diagnostic Cut-Offs in the Emergency Department (ICON-RELOADED) Study. Eight biogroups were derived based upon patterns in biomarker elevation at presentation and compared for differences in baseline characteristics. Of 441 patients with elevations in all 3 biomarkers, 218 (49.4%) were diagnosed with acute heart failure (HF). The frequency of acute HF diagnosis in this biogroup was higher than those with elevations in 2 biomarkers (18.8%, 44 of 234), 1 biomarker (3.8%, 10 of 260), or no elevated biomarkers (0.4%, 2 of 513). The absolute number of elevated biomarkers on admission was prognostic of the composite end point of mortality and HF rehospitalization. In adjusted models, patients with one, 2, and 3 elevated biomarkers had 3.74 (95% confidence interval [CI], 1.26-11.1, P = .017), 12.3 (95% CI, 4.60-32.9, P < .001), and 12.6 (95% CI, 4.54-35.0, P < .001) fold increased risk of 180-day mortality or HF rehospitalization. CONCLUSIONS A multimarker panel of NT-proBNP, hsTnT, and IGBFP7 provides unique clinical, diagnostic, and prognostic information in patients presenting with acute dyspnea. Differences in the number of elevated biomarkers at presentation may allow for more efficient clinical risk stratification of short-term mortality and HF rehospitalization.
Collapse
Affiliation(s)
- Andrew Abboud
- From the Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Naishu Kui
- Baim Institute for Clinical Research, Boston, Massachusetts
| | - Hanna K Gaggin
- From the Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | | | - John T Nagurney
- From the Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | | | - Peter S Pang
- Indiana University School of Medicine & Indianapolis EMS, Indianapolis, Indiana
| | | | | | - James L Januzzi
- From the Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Baim Institute for Clinical Research, Boston, Massachusetts.
| |
Collapse
|
8
|
Ghafouri-Fard S, Abak A, Mohaqiq M, Shoorei H, Taheri M. The Interplay Between Non-coding RNAs and Insulin-Like Growth Factor Signaling in the Pathogenesis of Neoplasia. Front Cell Dev Biol 2021; 9:634512. [PMID: 33768092 PMCID: PMC7985092 DOI: 10.3389/fcell.2021.634512] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/02/2021] [Indexed: 12/11/2022] Open
Abstract
The insulin-like growth factors (IGFs) are polypeptides with similar sequences with insulin. These factors regulate cell growth, development, maturation, and aging via different processes including the interplay with MAPK, Akt, and PI3K. IGF signaling participates in the pathogenesis of neoplasia, insulin resistance, diabetes mellitus, polycystic ovarian syndrome, cerebral ischemic injury, fatty liver disease, and several other conditions. Recent investigations have demonstrated the interplay between non-coding RNAs and IGF signaling. This interplay has fundamental roles in the development of the mentioned disorders. We designed the current study to search the available data about the role of IGF-associated non-coding RNAs in the evolution of neoplasia and other conditions. As novel therapeutic strategies have been designed for modification of IGF signaling, identification of the impact of non-coding RNAs in this pathway is necessary for the prediction of response to these modalities.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefe Abak
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Mohaqiq
- School of Advancement, Centennial College, Ashtonbee Campus, Toronto, ON, Canada
- Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University, Winston-Salem, NC, United States
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Biranjd University of Medical Sciences, Birjand, Iran
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
The Roles of Insulin-Like Growth Factor Binding Protein Family in Development and Diseases. Adv Ther 2021; 38:885-903. [PMID: 33331986 DOI: 10.1007/s12325-020-01581-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/19/2020] [Indexed: 12/17/2022]
Abstract
The insulin-like growth factor (IGF) system comprises ligands of IGF-I/II, IGF receptors (IGFR), IGF binding proteins (IGFBPs), and IGFBP hydrolases. The IGF system plays multiple roles during various disease development as IGFs are widely involved in cell proliferation and differentiation through regulating DNA transcription. Meanwhile, IGFBPs, which are mainly synthesized in the liver, can bind to IGFs and perform two different functions: either inhibition of IGFs by forming inactive compounds with IGF or enhancement of the function of IGFs by strengthening the IGF-IGFR interaction. Interestingly, IGFBPs may have wider functions through IGF-independent mechanisms. Studies have shown that IGFBPs play important roles in cardiovascular disease, tumor progression, fetal growth, and neuro-nutrition. In this review, we emphasize that different IGFBP family members have common or unique functions in numerous diseases; moreover, IGFBPs may serve as biomarkers for disease diagnosis and prediction.
Collapse
|
10
|
Guercio BJ, Zhang S, Ou FS, Venook AP, Niedzwiecki D, Lenz HJ, Innocenti F, Pollak MN, Nixon AB, Mullen BC, O'Neil BH, Shaw JE, Polite BN, Benson AB, Atkins JN, Goldberg RM, Brown JC, O'Reilly EM, Mayer RJ, Blanke CD, Fuchs CS, Meyerhardt JA. IGF-Binding Proteins, Adiponectin, and Survival in Metastatic Colorectal Cancer: Results From CALGB (Alliance)/SWOG 80405. JNCI Cancer Spectr 2020; 5:pkaa074. [PMID: 33426464 PMCID: PMC7785047 DOI: 10.1093/jncics/pkaa074] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 07/07/2020] [Accepted: 08/14/2020] [Indexed: 12/16/2022] Open
Abstract
Background Energy balance-related biomarkers are associated with risk and prognosis of various malignancies. Their relationship to survival in metastatic colorectal cancer (mCRC) requires further study. Methods Baseline plasma insulin-like growth factor (IGF)-1, IGF-binding protein (IGFBP)-3, IGFBP-7, C-peptide, and adiponectin were measured at time of trial registration in a prospective cohort of patients with mCRC participating in a National Cancer Institute–sponsored trial of first-line systemic therapy. We used Cox proportional hazards regression to adjust for confounders and examine associations of each biomarker with overall survival (OS) and progression-free survival (PFS). P values are 2-sided. Results Median follow-up for 1086 patients was 6.2 years. Compared with patients in the lowest IGFBP-3 quintile, patients in the highest IGFBP-3 quintile experienced an adjusted hazard ratio (HR) for OS of 0.57 (95% confidence interval [CI] = 0.42 to 0.78; Pnonlinearity < .001) and for PFS of 0.61 (95% CI = 0.45 to 0.82; Ptrend = .003). Compared with patients in the lowest IGFBP-7 quintile, patients in the highest IGFBP-7 quintile experienced an adjusted hazard ratio for OS of 1.60 (95% CI = 1.30 to 1.97; Ptrend < .001) and for PFS of 1.38 (95% CI = 1.13 to 1.69; Ptrend < .001). Plasma C-peptide and IGF-1 were not associated with patient outcomes. Adiponectin was not associated with OS; there was a nonlinear U-shaped association between adiponectin and PFS (Pnonlinearity = .03). Conclusions Among patients with mCRC, high plasma IGFBP-3 and low IGFBP-7 were associated with longer OS and PFS. Extreme levels of adiponectin were associated with shorter PFS. These findings suggest potential avenues for prognostic and therapeutic innovation.
Collapse
Affiliation(s)
- Brendan J Guercio
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sui Zhang
- Department of Medical Oncology, Dana-Farber/Partners CancerCare, Boston, MA, USA
| | - Fang-Shu Ou
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN, USA
| | - Alan P Venook
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Donna Niedzwiecki
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | | | | | - Michael N Pollak
- Department of Oncology, McGill University, Montreal, QC H3T 1E2, Canada
| | - Andrew B Nixon
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Brian C Mullen
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN, USA
| | - Bert H O'Neil
- Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - James E Shaw
- MedStar Washington Hospital Center, Washington, DC, USA
| | - Blase N Polite
- Pritzker School of Medicine, University of Chicago Comprehensive Cancer Center, Chicago, IL, USA
| | - Al Bowen Benson
- Department of Medicine, Northwestern University, Chicago, IL, USA
| | - James N Atkins
- Southeast Clinical Oncology Research (SCOR) Consortium, National Cancer Institute Community Oncology Research Program (NCORP), Winston-Salem, NC, USA
| | | | - Justin C Brown
- Department of Population and Public Health, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert J Mayer
- Department of Medical Oncology, Dana-Farber/Partners CancerCare, Boston, MA, USA
| | - Charles D Blanke
- SWOG Cancer Research Network and Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Charles S Fuchs
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Jeffrey A Meyerhardt
- Department of Medical Oncology, Dana-Farber/Partners CancerCare, Boston, MA, USA
| |
Collapse
|
11
|
Janmaat VT, Liu H, da Silva RA, Wisse PHA, Spaander MCW, Ten Hagen TLM, Smits R, Bruno MJ, Fuhler GM, Peppelenbosch MP. HOXA9 mediates and marks premalignant compartment size expansion in colonic adenomas. Carcinogenesis 2020; 40:1514-1524. [PMID: 31099823 DOI: 10.1093/carcin/bgz038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/27/2019] [Accepted: 02/14/2019] [Indexed: 12/21/2022] Open
Abstract
The transformation of normal colonic epithelium to colorectal cancer (CRC) involves a relatively ordered progression, and understanding the molecular alterations involved may aid rational design of strategies aimed at preventing or counteracting disease. Homeobox A9 (HOXA9) is an oncogene in leukemia and has been implicated in CRC pathology, although its role in disease etiology remains obscure at best. We observe that HOXA9 expression is increased in colonic adenomas compared with location-matched healthy colon epithelium. Its forced expression results in dramatic genetic and signaling changes, with increased expression of growth factors IGF1 and FLT3, super-activity of the AKT survival pathway and a concomitant increase in compartment size. Furthermore, a reduced mRNA expression of the epithelial to mesenchymal transition marker N-cadherin as well as reduced activity of the actin cytoskeletal mediator PAK was seen, which is in apparent agreement with an observed reduced migratory response in HOXA9-overexpressing cells. Thus, HOXA9 appears closely linked with adenoma growth while impairing migration and metastasis and hence is both a marker and driver of premalignant polyp growth. Colonic polyps grow but remain premalignant for up to decades. Here, we show that HOXA9 drives growth in premalignant polyps, but simultaneously prevents further transformation.
Collapse
Affiliation(s)
- Vincent T Janmaat
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, The Netherlands
| | - Hui Liu
- Department of Surgery, Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Erasmus MC - University Medical Center Rotterdam, The Netherlands
| | - Rodrigo A da Silva
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, The Netherlands
| | - Pieter H A Wisse
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, The Netherlands
| | - Manon C W Spaander
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, The Netherlands
| | - Timo L M Ten Hagen
- Department of Surgery, Laboratory Experimental Surgical Oncology, Section Surgical Oncology, Erasmus MC - University Medical Center Rotterdam, The Netherlands
| | - Ron Smits
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, The Netherlands
| | - Marco J Bruno
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, The Netherlands
| | - Gwenny M Fuhler
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, The Netherlands
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, The Netherlands
| |
Collapse
|
12
|
Ye C, Hou W, Chen M, Lu J, Chen E, Tang L, Hang K, Ding Q, Li Y, Zhang W, He R. IGFBP7 acts as a negative regulator of RANKL-induced osteoclastogenesis and oestrogen deficiency-induced bone loss. Cell Prolif 2019; 53:e12752. [PMID: 31889368 PMCID: PMC7046308 DOI: 10.1111/cpr.12752] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/04/2019] [Accepted: 12/11/2019] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Insulin-like growth factor-binding protein 7 (IGFBP7) is a low-affinity insulin growth factor (IGF) binder that may play an important role in bone metabolism. We previously reported that IGFBP7 enhanced osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) via the Wnt/β-catenin signalling pathway. In this study, we tried to reveal its function in osteoclast differentiation and osteoporosis. METHODS We used both in vitro and in vivo studies to investigate the effects of IGFBP7 on RANKL-induced osteoclastogenesis and osteoporosis, together with the underlying molecular mechanisms of these processes. RESULTS We show that IGFBP7 inhibited receptor activation of nuclear factor-κB (NF-κB) ligand (RANKL)-induced osteoclastogenesis, F-actin ring formation and bone resorption, which was confirmed by using recombinant IGFBP7 protein, lentivirus and siRNA. The NF-κB signalling pathway was inhibited during this process. Moreover, in a mouse ovariectomy-induced osteoporosis model, IGFBP7 treatment attenuated osteoporotic bone loss by inhibiting osteoclast activity. CONCLUSIONS Taken together, these findings show that IGFBP7 suppressed osteoclastogenesis in vitro and in vivo and suggest that IGFBP7 is a negative regulator of osteoclastogenesis and plays a protective role in osteoporosis. These novel insights into IGFBP7 may facilitate the development of potential treatment strategies for oestrogen deficiency-induced osteoporosis and other osteoclast-related disorders.
Collapse
Affiliation(s)
- Chenyi Ye
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Weiduo Hou
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Mo Chen
- Department of Rheumatology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jinwei Lu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Erman Chen
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Lan Tang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Kai Hang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Qianhai Ding
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Yan Li
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Wei Zhang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Rongxin He
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| |
Collapse
|
13
|
IGFBP7 Drives Resistance to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibition in Lung Cancer. Cancers (Basel) 2019; 11:cancers11010036. [PMID: 30609749 PMCID: PMC6356910 DOI: 10.3390/cancers11010036] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 12/17/2018] [Accepted: 12/24/2018] [Indexed: 12/14/2022] Open
Abstract
Patients with epidermal growth factor receptor (EGFR) mutation-positive lung cancer show a dramatic response to EGFR-tyrosine kinase inhibitors (TKIs). However, acquired drug resistance eventually develops. This study explored the novel mechanisms related to TKI resistance. To identify the genes associated with TKI resistance, an integrative approach was used to analyze public datasets. Molecular manipulations were performed to investigate the roles of insulin-like growth factor binding protein 7 (IGFBP7) in lung adenocarcinoma. Clinical specimens were collected to validate the impact of IGFBP7 on the efficacy of EGFR TKI treatment. IGFBP7 mRNA expression in cancer cells isolated from malignant pleural effusions after acquired resistance to EGFR-TKI was significantly higher than in cancer cells from treatment-naïve effusions. IGFBP7 expression was markedly increased in cells with long-term TKI-induced resistance compared to in TKI-sensitive parental cells. Reduced IGFBP7 in TKI-resistant cells reversed the resistance to EGFR-TKIs and increased EGFR-TKI-induced apoptosis by up-regulating B-cell lymphoma 2 interacting mediator of cell death (BIM) and activating caspases. Suppression of IGFBP7 attenuated the phosphorylation of insulin-like growth factor 1 receptor (IGF-IR) and downstream protein kinase B (AKT) in TKI-resistant cells. Clinically, higher serum IGFBP7 levels and tumors with positive IGFBP7-immunohistochemical staining were associated with poor TKI-treatment outcomes. IGFBP7 confers resistance to EGFR-TKIs and is a potential therapeutic target for treating EGFR-TKI-resistant cancers.
Collapse
|
14
|
Cai X, Wang L, Wang X, Hou F. Silence of IGFBP7 suppresses apoptosis and epithelial mesenchymal transformation of high glucose induced-podocytes. Exp Ther Med 2018; 16:1095-1102. [PMID: 30112052 PMCID: PMC6090473 DOI: 10.3892/etm.2018.6298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/16/2018] [Indexed: 12/15/2022] Open
Abstract
Insulin-like growth factor-binding protein 7 (IGFBP7) has been identified as a secreted protein associated with a number of cellular processes. However, the specific regulatory mechanisms of IGFBP7 on podocytes of diabetic nephropathy (DN) are yet to be elucidated. In the present study, podocytes were identified initially via an immunofluorescence assay using an anti-synaptopodin antibody. It was subsequently demonstrated that glucose promoted podocyte proliferation in a time- and dose-dependent manner via MTT assay. In addition, IGFBP7 expression was silenced in podocytes via siRNA, the effects of which were evaluated using western blotting and reverse transcription-quantitative polymerase chain reaction. It was demonstrated that silencing IGFBP7 inhibited apoptosis and epithelial mesenchymal transformation (EMT) of podocytes mediated by high glucose (HG). Transforming growth factor (TGF)-β1/mothers against decapentaplegic homolog (Smad) signaling was associated with proliferation, apoptotic activities and EMT. Therefore, the expression levels of TGF-β1/Smad pathway were detected, and it was observed that silencing IGFBP7 suppressed the TGF-β1/Smad pathway in podocytes induced by HG. These findings suggested that IGFBP7 may serve as a potential therapeutic target for DN.
Collapse
Affiliation(s)
- Xiaojun Cai
- Department of Endocrinology, Heilongjiang Provincial Academy of Chinese Medical Science, Harbin, Heilongjiang 150036, P.R. China
| | - Lei Wang
- Department of Endocrinology, Heilongjiang Provincial Academy of Chinese Medical Science, Harbin, Heilongjiang 150036, P.R. China
| | - Xuling Wang
- Department of Endocrinology, Heilongjiang Provincial Academy of Chinese Medical Science, Harbin, Heilongjiang 150036, P.R. China
| | - Fengyan Hou
- Department of Endocrinology, Heilongjiang Provincial Academy of Chinese Medical Science, Harbin, Heilongjiang 150036, P.R. China
| |
Collapse
|
15
|
Infante A, Rodríguez CI. Secretome analysis of in vitro aged human mesenchymal stem cells reveals IGFBP7 as a putative factor for promoting osteogenesis. Sci Rep 2018; 8:4632. [PMID: 29545581 PMCID: PMC5854613 DOI: 10.1038/s41598-018-22855-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 03/02/2018] [Indexed: 12/13/2022] Open
Abstract
Aging is a complex biological process, which involves multiple mechanisms with different levels of regulation. Senescent cells are known to secrete senescence-associated proteins, which exert negative influences on surrounding cells. Mesenchymal stem cells (MSCs), the common progenitors for bone, cartilage and adipose tissue (which are especially affected tissues in aging), are known to secrete a broad spectrum of biologically active proteins with both paracrine and autocrine functions in many biological processes. In this report, we have studied the secreted factors (secretome) from human MSCs (hMSCs) and hMSCs-derived adipocytes which were induced to accumulate prelamin A, the immature form of the nuclear lamina protein called Lamin A, known to induce premature aging syndromes in humans and in murine models. Proteomic analysis from two different techniques, antibody arrays and LS-MS, showed that prelamin A accumulation in hMSCs promotes the differential secretion of factors previously identified as secreted by hMSCs undergoing osteogenesis. Moreover, this secretome was able to modulate osteogenesis of normal hMSCs in vitro. Finally, we found that one of the overexpressed secreted factors of this human aging in vitro stem cell model, IGFBP-7, is an osteogenic factor, essential for the viability of hMSCs during osteogenesis.
Collapse
Affiliation(s)
- Arantza Infante
- Stem Cells and Cell Therapy Laboratory, BioCruces Health Research Institute, Cruces University Hospital, Barakaldo, 48903, Spain
| | - Clara I Rodríguez
- Stem Cells and Cell Therapy Laboratory, BioCruces Health Research Institute, Cruces University Hospital, Barakaldo, 48903, Spain.
| |
Collapse
|
16
|
Bieghs L, Johnsen HE, Maes K, Menu E, Van Valckenborgh E, Overgaard MT, Nyegaard M, Conover CA, Vanderkerken K, De Bruyne E. The insulin-like growth factor system in multiple myeloma: diagnostic and therapeutic potential. Oncotarget 2018; 7:48732-48752. [PMID: 27129151 PMCID: PMC5217049 DOI: 10.18632/oncotarget.8982] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 04/16/2016] [Indexed: 12/14/2022] Open
Abstract
Multiple myeloma (MM) is a highly heterogeneous plasma cell malignancy. The MM cells reside in the bone marrow (BM), where reciprocal interactions with the BM niche foster MM cell survival, proliferation, and drug resistance. As in most cancers, the insulin-like growth factor (IGF) system has been demonstrated to play a key role in the pathogenesis of MM. The IGF system consists of IGF ligands, IGF receptors, IGF binding proteins (IGFBPs), and IGFBP proteases and contributes not only to the survival, proliferation, and homing of MM cells, but also MM-associated angiogenesis and osteolysis. Furthermore, increased IGF-I receptor (IGF-IR) expression on MM cells correlates with a poor prognosis in MM patients. Despite the prominent role of the IGF system in MM, strategies targeting the IGF-IR using blocking antibodies or small molecule inhibitors have failed to translate into the clinic. However, increasing preclinical evidence indicates that IGF-I is also involved in the development of drug resistance against current standard-of-care agents against MM, including proteasome inhibitors, immunomodulatory agents, and corticoids. IGF-IR targeting has been able to overcome or revert this drug resistance in animal models, enhancing the efficacy of standard-of-care agents. This finding has generated renewed interest in the therapeutic potential of IGF-I targeting in MM. The present review provides an update of the impact of the different IGF system components in MM and discusses the diagnostic and therapeutic potentials.
Collapse
Affiliation(s)
- Liesbeth Bieghs
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium.,Department of Hematology, Aalborg Hospital, Aalborg University, Denmark.,Department of Biomedicin, Aarhus University, Aarhus, Denmark
| | - Hans E Johnsen
- Department of Hematology, Aalborg Hospital, Aalborg University, Denmark.,Clinical Cancer Research Center, Aalborg University Hospital, Denmark.,Department of Clinical Medicine, Aalborg University, Denmark
| | - Ken Maes
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Els Van Valckenborgh
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Mette Nyegaard
- Department of Biomedicin, Aarhus University, Aarhus, Denmark
| | - Cheryl A Conover
- Division of Endocrinology, Metabolism and Nutrition, Endocrine Research Unit, Mayo Clinic, Rochester, NY, USA
| | - Karin Vanderkerken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Elke De Bruyne
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
17
|
Zhang W, Chen E, Chen M, Ye C, Qi Y, Ding Q, Li H, Xue D, Gao X, Pan Z. IGFBP7 regulates the osteogenic differentiation of bone marrow-derived mesenchymal stem cells via Wnt/β-catenin signaling pathway. FASEB J 2018; 32:2280-2291. [PMID: 29242275 DOI: 10.1096/fj.201700998rr] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Insulin-like growth factor-binding protein 7 (IGFBP7), a low-affinity IGF binder, may play an important role in bone metabolism. However, its function in osteogenic differentiation of human bone marrow-derived mesenchymal stem cells (BMSCs) remains unclear. Therefore, we investigated its effects on osteogenic differentiation. Overexpression of IGFBP7 enhanced the expression of osteo-specific genes and proteins, and IGFBP7 knockdown decreased osteogenesis-specific markers. More mineral deposits and higher alkaline phosphatase activity were observed after the up-regulation of IGFBP7. Moreover, β-catenin levels were up-regulated by the overexpression of IGFBP7 or the addition of extracellular IGFBP7 protein and were reduced by the depletion of IGFBP7. The increase in osteogenic differentiation due to the overexpression of IGFBP7 was partially decreased by specific Wnt/β-catenin signaling inhibitors. Using a rat tibial osteotomy model, a sheet of IGFBP7-overexpressing BMSCs improved bone healing, as demonstrated by imaging, biomechanical, and histologic analyses. Taken together, these findings indicate that IGFBP7 regulates the osteogenic differentiation of BMSCs partly via the Wnt/β-catenin signaling pathway.-Zhang, W., Chen, E., Chen, M., Ye, C., Qi, Y., Ding, Q., Li, H., Xue, D., Gao, X., Pan, Z. IGFBP7 regulates the osteogenic differentiation of bone marrow-derived mesenchymal stem cells via Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research Institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Erman Chen
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research Institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Mo Chen
- Department of Rheumatology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Nephrology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Chenyi Ye
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research Institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Yiying Qi
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research Institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Qianhai Ding
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research Institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Hang Li
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research Institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Deting Xue
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research Institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Xiang Gao
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research Institute of Orthopaedics, Zhejiang University, Hangzhou, China
| | - Zhijun Pan
- Department of Orthopedics, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Research Institute of Orthopaedics, Zhejiang University, Hangzhou, China
| |
Collapse
|
18
|
Clinical implications of c-maf expression in plasma cells from patients with multiple myeloma. Exp Hematol Oncol 2017; 6:16. [PMID: 28560070 PMCID: PMC5446722 DOI: 10.1186/s40164-017-0076-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 05/12/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a type of hematological malignancy with significant heterogeneity in clinical features and prognosis. Cytogenetic abnormalities are the major factors affecting patient outcomes. Studies have shown that immunohistochemistry (IHC)-based detection of cancer-related genes expression could be alternative indicators for the prognosis of MM. METHODS Nuclear expression of c-maf protein in the bone marrow plasma cells of 128 multiple myeloma patients were examined by IHC, and its association with the clinicopathological features of MM patients was analyzed as well. RESULTS Among the 128 patients, the positive rate of c-maf protein expression was up to 30.5%, which had no correlation with patient age, M protein type, Durie-Salmon staging system, the International Staging System, abnormal plasma cell ratio in the bone marrow, or the level of peripheral blood hemoglobin, serum calcium or lactate dehydrogenase. However, the c-maf-positive patients had a significantly higher rate of hypoproteinemia (p = 0.026) and higher serum β2-microglobulin levels (>2500 μg/L) (p = 0.007). Patients with negative c-maf expression had higher remission rates upon the treatment of non-bortezomib-based regimens although no effect of c-maf expression on progression-free survival or overall survival was observed. CONCLUSION Patients with negative c-maf expression had higher remission rates upon the treatment of non-bortezomib-based regimens although no effect of c-maf expression on survival was observed. A further large-scale prospective study is required to verify these findings.
Collapse
|
19
|
Hanbali A, Hassanein M, Rasheed W, Aljurf M, Alsharif F. The Evolution of Prognostic Factors in Multiple Myeloma. Adv Hematol 2017; 2017:4812637. [PMID: 28321258 PMCID: PMC5339490 DOI: 10.1155/2017/4812637] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/22/2017] [Accepted: 02/05/2017] [Indexed: 12/23/2022] Open
Abstract
Multiple myeloma (MM) is a heterogeneous hematologic malignancy involving the proliferation of plasma cells derived by different genetic events contributing to the development, progression, and prognosis of this disease. Despite improvement in treatment strategies of MM over the last decade, the disease remains incurable. All efforts are currently focused on understanding the prognostic markers of the disease hoping to incorporate the new therapeutic modalities to convert the disease into curable one. We present this comprehensive review to summarize the current standard prognostic markers used in MM along with novel techniques that are still in development and highlight their implications in current clinical practice.
Collapse
Affiliation(s)
- Amr Hanbali
- King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Mona Hassanein
- King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Walid Rasheed
- King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Mahmoud Aljurf
- King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Fahad Alsharif
- King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
20
|
Zhou X, Dai X, Wu X, Ji J, Karaplis A, Goltzman D, Yang X, Miao D. Overexpression of Bmi1 in Lymphocytes Stimulates Skeletogenesis by Improving the Osteogenic Microenvironment. Sci Rep 2016; 6:29171. [PMID: 27373231 PMCID: PMC4931581 DOI: 10.1038/srep29171] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 06/15/2016] [Indexed: 12/13/2022] Open
Abstract
To investigate whether overexpression of Bmi1 in lymphocytes can stimulate skeletogenesis by improving the osteogenic microenvironment, we examined the skeletal phenotype of EμBmi1 transgenic mice with overexpression of Bmi1 in lymphocytes. The size of the skeleton, trabecular bone volume and osteoblast number, indices of proliferation and differentiation of bone marrow mesenchymal stem cells (BM-MSCs) were increased significantly, ROS levels were reduced and antioxidative capacity was enhanced in EμBmi1 mice compared to WT mice. In PTHrP1-84 knockin (Pthrp(KI/KI)) mice, the expression levels of Bmi1 are reduced and potentially can mediate the premature osteoporosis observed. We therefore generated a Pthrp(KI/KI) mice overexpressing Bmi1 in lymphocytes and compared them with Pthrp(KI/KI) and WT littermates. Overexpression of Bmi1 in Pthrp(KI/KI) mice resulted in a longer lifespan, increased body weight and improvement in skeletal growth and parameters of osteoblastic bone formation with reduced ROS levels and DNA damage response parameters. Our results demonstrate that overexpression of Bmi1 in lymphocytes can stimulate osteogenesis in vivo and partially rescue defects in skeletal growth and osteogenesis in Pthrp(KI/KI) mice. These studies therefore indicate that overexpression of Bmi1 in lymphocytes can stimulate skeletogenesis by inhibiting oxidative stress and improving the osteogenic microenvironment.
Collapse
Affiliation(s)
- Xichao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
- The State Key Laboratory of Reproductive Medicine, the Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Xiuliang Dai
- The State Key Laboratory of Reproductive Medicine, the Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Xuan Wu
- The State Key Laboratory of Reproductive Medicine, the Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Ji Ji
- The State Key Laboratory of Reproductive Medicine, the Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
- Department of Fundamentals of Nursing, School of Nursing, Nanjing Medical University, Nanjing, China
| | - Andrew Karaplis
- The Department of Medicine, McGill University, Montreal, Canada
| | - David Goltzman
- The Department of Medicine, McGill University, Montreal, Canada
| | - Xiangjiao Yang
- The Department of Medicine, McGill University, Montreal, Canada
- Rosalind & Morris Goodman Cancer Research Center, Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Dengshun Miao
- The State Key Laboratory of Reproductive Medicine, the Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
21
|
Screening for and validation of a hepatic fibrosis-related pathway induced by insulin-like growth factor-binding protein-related protein 1. Eur J Gastroenterol Hepatol 2016; 28:762-72. [PMID: 27097355 DOI: 10.1097/meg.0000000000000631] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Our previous studies characterized insulin-like growth factor-binding protein-related protein 1 (IGFBPrP1) as a molecule that promotes hepatic fibrogenesis, but its mechanism has not been fully elucidated. Here, we have investigated the effect of IGFBPrP1 on gene expression in the hepatic fibrosis-related pathway. MATERIALS AND METHODS Sprague-Dawley rats received injections of an adenovirus carrying IGFBPrP1 or EGFP cDNA into their tail veins. In hepatic preparations, hepatic stellate cell activation was determined by α-smooth muscle actin expression and hepatic fibrosis by Sirius red staining and hydroxyproline content analysis. IGFBPrP1-inducible genes of the hepatic fibrosis-related pathway were assessed by PCR array. Expression of IGFBPrP1 and transforming growth factor β1 (TGFβ1) and array results were evaluated by quantitative real-time PCR and western blotting. RESULTS IGFBPrP1-overexpressing rats showed an increase in α-smooth muscle actin expression and collagen and hydroxyproline content in the liver. The PCR array results indicated that some genes were upregulated and some were downregulated in Ad-IGFBPrP1-infected rats. Among these, Egr1, MAP2K2 (MEK2) and MAPK3 (ERK1) expression increased, whereas PTEN and Hhip mRNA expression decreased. Egr1 protein levels increased and peaked 2 weeks after infection and then decreased gradually. PTEN protein decreased gradually in Ad-IGFBPrP1-infected rats with a concurrent increase in the degree of hepatic fibrosis. TGFβ1 levels increased during hepatic fibrosis development in liver tissues. CONCLUSION Egr1, PTEN, Hhip, MAP2K2 (MEK2) and MAPK3 (ERK1) were identified as candidate genes of the IGFBPrP1-induced hepatic fibrosis-related pathway. IGFBPrP1 promoted hepatic fibrosis mainly by enhancing the TGFβ1 expression that it triggered.
Collapse
|
22
|
Bieghs L, Brohus M, Kristensen IB, Abildgaard N, Bøgsted M, Johnsen HE, Conover CA, De Bruyne E, Vanderkerken K, Overgaard MT, Nyegaard M. Abnormal IGF-Binding Protein Profile in the Bone Marrow of Multiple Myeloma Patients. PLoS One 2016; 11:e0154256. [PMID: 27111220 PMCID: PMC4844248 DOI: 10.1371/journal.pone.0154256] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 04/11/2016] [Indexed: 01/02/2023] Open
Abstract
Insulin-like growth factor (IGF) signalling plays a key role in homing, progression, and treatment resistance in multiple myeloma (MM). In the extracellular environment, the majority of IGF molecules are bound to one of six IGF-binding proteins (IGFBP1-6), leaving a minor fraction of total IGF free and accessible for receptor activation. In MM, high IGF-receptor type 1 expression levels correlate with a poor prognosis, but the status and role of IGF and IGFBPs in the pathobiology of MM is unknown. Here we measured total IGF1, IGF2, and intact IGFBP levels in blood and bone marrow samples from MM (n = 17), monoclonal gammopathy of undetermined significance (MGUS) (n = 37), and control individuals (n = 15), using ELISA (IGFs) and 125I-IGF1 Western Ligand Blotting (IGFBPs). MGUS and MM patients displayed a significant increase in intact IGFBP-2 (2.5-3.8 fold) and decrease in intact IGFBP-3 (0.6-0.5 fold) in the circulation compared to control individuals. Further, IGFBP-2 as well as total IGFBP levels were significantly lower in bone marrow compared to circulation in MM and MGUS only, whereas IGF1, IGF2, and IGFBP-3 were equally distributed between the two compartments. In conclusion, the profound change in IGFBP profile strongly suggests an increased IGF bioavailability in the bone marrow microenvironment in MGUS and MM, despite no change in growth factor concentration.
Collapse
Affiliation(s)
- Liesbeth Bieghs
- Department of Hematology, Aalborg Hospital, Aalborg University, Alborg, Denmark
- Department of Hematology and Immunology-Myeloma Center Brussel, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Malene Brohus
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Ida B. Kristensen
- Department of Haematology, Odense University Hospital, Odense, Denmark
| | - Niels Abildgaard
- Department of Haematology, Odense University Hospital, Odense, Denmark
| | - Martin Bøgsted
- Department of Hematology, Aalborg Hospital, Aalborg University, Alborg, Denmark
| | - Hans E. Johnsen
- Department of Hematology, Aalborg Hospital, Aalborg University, Alborg, Denmark
| | - Cheryl A. Conover
- Division of Endocrinology, Metabolism and Nutrition, Endocrine Research Unit, Mayo Clinic, Rochester, MN, United States of America
| | - Elke De Bruyne
- Department of Hematology and Immunology-Myeloma Center Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karin Vanderkerken
- Department of Hematology and Immunology-Myeloma Center Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Mette Nyegaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
23
|
Bolomsky A, Schlangen K, Schreiner W, Zojer N, Ludwig H. Targeting of BMI-1 with PTC-209 shows potent anti-myeloma activity and impairs the tumour microenvironment. J Hematol Oncol 2016; 9:17. [PMID: 26935956 PMCID: PMC4776359 DOI: 10.1186/s13045-016-0247-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/23/2016] [Indexed: 11/30/2022] Open
Abstract
Background The polycomb complex protein BMI-1 (BMI-1) is a putative oncogene reported to be overexpressed in multiple myeloma (MM). Silencing of BMI-1 was shown to impair the growth and survival of MM cells. However, therapeutic agents specifically targeting BMI-1 were not available so far. Here, we investigated PTC-209, a novel small molecule inhibitor of BMI-1, for its activity in MM. Methods BMI-1 expression was analysed in human MM cell lines and primary MM cells by using publically available gene expression profiling (GEP) data. The anti-MM activity of PTC-209 was investigated by viability testing, cell cycle analysis, annexin V and 7-AAD staining, quantification of cleaved poly(ADP-ribose) polymerase (PARP), JC-1 as well as colony formation assays. Deregulation of central myeloma growth and survival genes was studied by quantitative PCR and flow cytometry, respectively. In addition, the impact of PTC-209 on in vitro osteoclast, osteoblast and tube formation was analysed. Results We confirmed overexpression of BMI-1 in MM patients by using publically available GEP datasets. Of note, BMI-1 expression was further increased at relapse which translated into significantly shorter overall survival in relapsed/refractory patients treated with bortezomib or dexamethasone. Treatment with PTC-209 significantly decreased viable cell numbers in human MM cell lines, induced a G1 cell cycle arrest, promoted apoptosis and demonstrated synergistic activity with pomalidomide and carfilzomib. The anti-MM activity of PTC-209 was accompanied by a significant decrease of cyclin D1 (CCND1) and v-myc avian myelocytomatosis viral oncogene homolog (MYC) expression as well as upregulation of cyclin-dependent kinase inhibitor 1A (CDKN1A) and cyclin-dependent kinase inhibitor 1B (CDKN1B). We also observed upregulation of NOXA (up to 3.6 ± 1.2-fold induction, P = 0.009) and subsequent downregulation of myeloid cell leukemia 1 (MCL-1) protein levels, which likely mediates the apoptotic effects of PTC-209. Importantly, the anti-MM activity was upheld in the presence of stromal support or myeloma growth factors insulin-like growth factor 1 (IGF-1) and interleukin 6 (IL-6). In the MM microenvironment, PTC-209 impaired tube formation, impaired osteoclast development and decreased osteoblast formation in a dose-dependent manner (P < 0.01 at 1 μM, respectively). The latter might be attributed to an induction of DKK1 and was reversed by concurrent anti-DKK1 antibody treatment. Conclusions We confirmed overexpression of BMI-1 in MM highlighting its role as an attractive drug target and reveal therapeutic targeting of BMI-1 by PTC-209 as a promising novel therapeutic intervention for MM. Electronic supplementary material The online version of this article (doi:10.1186/s13045-016-0247-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Arnold Bolomsky
- Wilhelminen Cancer Research Institute, Department of Medicine I, Wilhelminenspital, Montleartstraße 37, 1160, Vienna, Austria.
| | - Karin Schlangen
- Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, Vienna, Austria.
| | - Wolfgang Schreiner
- Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, Vienna, Austria.
| | - Niklas Zojer
- Wilhelminen Cancer Research Institute, Department of Medicine I, Wilhelminenspital, Montleartstraße 37, 1160, Vienna, Austria.
| | - Heinz Ludwig
- Wilhelminen Cancer Research Institute, Department of Medicine I, Wilhelminenspital, Montleartstraße 37, 1160, Vienna, Austria.
| |
Collapse
|
24
|
Chhabra S, Jain S, Wallace C, Hong F, Liu B. High expression of endoplasmic reticulum chaperone grp94 is a novel molecular hallmark of malignant plasma cells in multiple myeloma. J Hematol Oncol 2015; 8:77. [PMID: 26108343 PMCID: PMC4483199 DOI: 10.1186/s13045-015-0177-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 06/17/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a hematologic malignancy that is characterized by the proliferation of abnormal bone marrow plasma cells (BMPC) and overproduction of immunoglobulin or light chains with evidence of end-organ damage such as bone damage, anemia, hypercalcemia, and renal dysfunction. The pathogenesis of MM is closely linked to dysregulated unfolded protein response (UPR) in the endoplasmic reticulum (ER). Constitutive activation of UPR in mice, as demonstrated by transgenic expression of a master UPR transcription factor XBP1s (a UPR-specific splice variant of X-box binding protein 1), causes myeloma. grp94 (gp96) is a key downstream chaperone in the ER that mediates the UPR as a part of the protein quality control mechanism in the secretory pathway. Our recent study has shown that the persistence of plasma cells as well as the development of myeloma in XBP1s-transgenic mice is critically dependent on grp94. However, the role of grp94 in the initiation and progression of human MM is still unknown. METHODS The expression level of grp94 in BMPCs was measured by flow cytometry, real-time RT-PCR, and Western blot analysis. We compared the expression levels of grp94 in BMPCs in a spectrum of patients including MM, monoclonal gammopathy of undetermined significance (MGUS), smoldering MM (SMM), as well as non-plasma cell disorders (NPC). RESULTS We found that grp94 was highly expressed in malignant plasma cells in patients with MM, but not in BMPCs in patients with MGUS/SMM and NPC. The expression level of grp94 correlated significantly with CD138 expression level. We also found that the grp94 expression level in BMPCs from International Staging System (ISS) stage III MM patients is higher than those in ISS stage I/II MM patients. CONCLUSIONS grp94 is highly expressed in BMPCs in MM, which correlates with the advanced stage of this disease. Our data demonstrated that grp94 is a novel diagnostic and prognostic biomarker. It also positioned grp94 as a promising therapeutic target for MM.
Collapse
Affiliation(s)
- Saurabh Chhabra
- Hollings Cancer Center, 86 Jonathan Lucas Street, Charleston, SC, 29425, USA. .,Division of Hematology and Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA.
| | - Sandeep Jain
- Hollings Cancer Center, 86 Jonathan Lucas Street, Charleston, SC, 29425, USA. .,Division of Hematology and Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA.
| | - Caroline Wallace
- Hollings Cancer Center, 86 Jonathan Lucas Street, Charleston, SC, 29425, USA. .,Department of Microbiology and Immunology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC, 29425, USA.
| | - Feng Hong
- Hollings Cancer Center, 86 Jonathan Lucas Street, Charleston, SC, 29425, USA. .,Department of Microbiology and Immunology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC, 29425, USA.
| | - Bei Liu
- Hollings Cancer Center, 86 Jonathan Lucas Street, Charleston, SC, 29425, USA. .,Department of Microbiology and Immunology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC, 29425, USA.
| |
Collapse
|