1
|
Anloague A, Sabol HM, Kaur J, Khan S, Ashby C, Schinke C, Barnes CL, Alturkmani F, Ambrogini E, Gundesen MT, Lund T, Amstrup AK, Andersen TL, Diaz-delCastillo M, Roodman GD, Bellido T, Delgado-Calle J. A novel CCL3-HMGB1 signaling axis regulating osteocyte RANKL expression in multiple myeloma. Haematologica 2025; 110:952-966. [PMID: 39605211 PMCID: PMC11959238 DOI: 10.3324/haematol.2024.286484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Multiple myeloma (MM) is a clonal plasma cell proliferative malignancy characterized by a debilitating bone disease. Osteolytic destruction, a hallmark of MM, is driven by increased osteoclast number and exacerbated bone resorption, primarily fueled by the excessive production of RANKL, the master regulator of osteoclast formation, within the tumor niche. We previously reported that osteocytes, the most abundant cells in the bone niche, promote tumor progression and support MM bone disease by overproducing RANKL. However, the molecular mechanisms underlying RANKL dysregulation in osteocytes in the context of MM bone disease are not entirely understood. Here, we present evidence that MM-derived CCL3 induces upregulation of RANKL expression in both human and murine osteocytes. Through a combination of in vitro, ex vivo, and in vivo models and clinical data, we demonstrate that genetic or pharmacologic inhibition of CCL3 prevents RANKL upregulation in osteocytes and attenuates the bone loss induced by MM cells. Mechanistic studies revealed that MM-derived CCL3 triggers the secretion of HMGB1 by osteocytes, a process required for osteocytic RANKL upregulation by MM cells. These findings identify a previously unknown CCL3-HMGB1 signaling axis in the MM tumor niche that drives bone resorption by promoting RANKL overproduction in osteocytes.
Collapse
Affiliation(s)
- Aric Anloague
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Hayley M Sabol
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Japneet Kaur
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US; Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock
| | - Sharmin Khan
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Cody Ashby
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, US; Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Carolina Schinke
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, US; Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR
| | - C Lowry Barnes
- Department of Orthopedic Surgery; University of Arkansas for Medical Sciences, Little Rock, AR
| | - Farah Alturkmani
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US; Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences Little Rock, AR
| | - Elena Ambrogini
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US; Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences Little Rock, AR, US; Central Arkansas Veterans Healthcare System, Little Rock, AR
| | - Michael Tveden Gundesen
- Department of Hematology, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Thomas Lund
- Department of Hematology, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Centre for Innovative Medical Technology, Odense University Hospital, Odense, Denmark
| | - Anne Kristine Amstrup
- Department of Endocrinology and Internal Medicine (MEA), THG, Aarhus University Hospital, Aarhus, Denmark
| | - Thomas Levin Andersen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Pathology, Odense University Hospital, Odense, Denmark; Department of Forensic Medicine, University of Aarhus, Aarhus, Denmark
| | | | - G David Roodman
- Division of Hematology and Oncology, Department of Medicine, Indiana University, Indianapolis, IN
| | - Teresita Bellido
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US; Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, US; Department of Orthopedic Surgery; University of Arkansas for Medical Sciences, Little Rock, AR, US; Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences Little Rock, AR, US; Central Arkansas Veterans Healthcare System, Little Rock, AR
| | - Jesus Delgado-Calle
- Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US; Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, US; Department of Orthopedic Surgery; University of Arkansas for Medical Sciences, Little Rock, AR, US; Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences Little Rock, AR.
| |
Collapse
|
2
|
Luyckx B, Van Trimpont M, Declerck F, Staessens E, Verhee A, T'Sas S, Eyckerman S, Offner F, Van Vlierberghe P, Goossens S, Clarisse D, De Bosscher K. CCR1 inhibition sensitizes multiple myeloma cells to glucocorticoid therapy. Pharmacol Res 2025; 215:107709. [PMID: 40132675 DOI: 10.1016/j.phrs.2025.107709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/12/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025]
Abstract
Glucocorticoids (GC) are cornerstone drugs in the treatment of multiple myeloma (MM). Because MM cells exploit the bone marrow microenvironment to obtain growth and survival signals, resistance to glucocorticoid-induced apoptosis emerges, yet the underlying mechanisms remain poorly characterized. Here, we identify that the chemokine receptor CCR1, together with its main ligand CCL3, plays a pivotal role in reducing the glucocorticoid sensitivity of MM cells. We show that blocking CCR1 signaling with the antagonist BX471 enhances the anti-MM effects of the glucocorticoid dexamethasone in MM cell lines, primary patient material and a myeloma xenograft mouse model. Mechanistically, the drug combination shifts the balance between pro- and antiapoptotic proteins towards apoptosis and deregulates lysosomal proteins. Our findings suggest that CCR1 may play a role in glucocorticoid resistance, as the GC-induced downregulation of CCR1 mRNA and protein is blunted in a GC-resistance onset model. Moreover, we demonstrate that inhibiting CCR1 partially reverses this resistance, providing a promising strategy for resensitizing MM cells to GC treatment.
Collapse
Affiliation(s)
- Bert Luyckx
- VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, Gent 9052, Belgium; Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium; Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Maaike Van Trimpont
- Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium; Department of Diagnostic Sciences, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Fien Declerck
- VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, Gent 9052, Belgium; Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Eleni Staessens
- VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, Gent 9052, Belgium; Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium; Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Annick Verhee
- VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, Gent 9052, Belgium; Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Sara T'Sas
- Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium; Department of Diagnostic Sciences, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Sven Eyckerman
- VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, Gent 9052, Belgium; Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium; Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Fritz Offner
- Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium; Department of Internal Medicine and Pediatrics, Ghent University Hospital, Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Pieter Van Vlierberghe
- Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium; Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Steven Goossens
- Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium; Department of Diagnostic Sciences, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Dorien Clarisse
- VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, Gent 9052, Belgium; Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium; Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium
| | - Karolien De Bosscher
- VIB-UGent Center for Medical Biotechnology, Technologiepark-Zwijnaarde 75, Gent 9052, Belgium; Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Gent 9000, Belgium; Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, Gent 9000, Belgium.
| |
Collapse
|
3
|
Yang H, He Y, Qu F, Zhu J, Deng L, Jiang F, Wu X, Chen Y, Kashif A, Wang X. Maraviroc enhances Bortezomib sensitivity in multiple myeloma by inhibiting M2 macrophage polarization via PI3K/AKT/RhoA signaling pathway in macrophages. Cell Div 2025; 20:5. [PMID: 39953613 PMCID: PMC11829472 DOI: 10.1186/s13008-025-00145-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 01/21/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a malignancy where drug resistance often leads to relapse or refractory disease. Chemokine receptor 5 (CCR5) has emerged as a novel therapeutic target. However, the role of CCR5-antagonist Maraviroc (MVC) in M2 macrophage polarization and its potential to enhance Bortezomib sensitivity in MM has not been fully explored. METHODS We used human bone marrow samples, RPMI 8226 cells, and THP-1 monocytes to investigate CCL3/CCR5 axis. ELISA measured CCL3/CCR5 levels. Knockdown/overexpression vectors modulated expression. Cell proliferation, apoptosis, and macrophage polarization were assessed using CCK8, flow cytometry, and transwell assays. QRT-PCR analyzed CCL3 expression, and western blotting examined PI3K/AKT/RhoA signaling. CCR5 was targeted via siRNAs or MVC. NOD/SCID mouse model evaluated CCL3/CCR5 effects on macrophage polarization and MVC's impact on Bortezomib efficacy. RESULTS CCL3, CCR5, and M2 macrophage markers are upregulated in MM patients, with CCL3/CCR5 expression correlating with M2 macrophage polarization. Myeloma-secreted CCL3 and paracrine CCR5 significantly promoted M2 macrophage polarization by activating PI3K/AKT/RhoA signaling, which in turn enhanced myeloma proliferation, inhibited apoptosis, and reduced Bortezomib sensitivity. MVC inhibited M2 macrophage polarization and improved Bortezomib sensitivity in vitro and xenograft mouse myeloma models. CONCLUSIONS MVC reduced macrophage polarization and enhanced Bortezomib sensitivity in MM cells.
Collapse
Affiliation(s)
- Huiye Yang
- Department of Hematology, Affiliated Hospital of Guilin Medicial University, Lequn Road 15#, Guilin City, Guangx, 541000, China
| | - Yuchan He
- Department of Hematology, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
| | - Fujun Qu
- Department of Hematology, Affiliated Hospital of Guilin Medicial University, Lequn Road 15#, Guilin City, Guangx, 541000, China
| | - Jie Zhu
- Department of Hematology, Affiliated Hospital of Guilin Medicial University, Lequn Road 15#, Guilin City, Guangx, 541000, China
| | - Liyuan Deng
- Department of Hematology, Affiliated Hospital of Guilin Medicial University, Lequn Road 15#, Guilin City, Guangx, 541000, China
| | - Fang Jiang
- Department of Hematology, Affiliated Hospital of Guilin Medicial University, Lequn Road 15#, Guilin City, Guangx, 541000, China
| | - Xianyi Wu
- Department of Hematology, Affiliated Hospital of Guilin Medicial University, Lequn Road 15#, Guilin City, Guangx, 541000, China
| | - Yixuan Chen
- Department of Hematology, Affiliated Hospital of Guilin Medicial University, Lequn Road 15#, Guilin City, Guangx, 541000, China
| | - Ali Kashif
- Department of Hematology, Affiliated Hospital of Guilin Medicial University, Lequn Road 15#, Guilin City, Guangx, 541000, China
| | - Xiaotao Wang
- Department of Hematology, Affiliated Hospital of Guilin Medicial University, Lequn Road 15#, Guilin City, Guangx, 541000, China.
| |
Collapse
|
4
|
Chen Y, Zhao H, Zhang H, Wang B, Ma J. Cytokine profile of cerebrospinal fluid in pediatric patients with metastatic medulloblastoma. Heliyon 2024; 10:e38504. [PMID: 39524698 PMCID: PMC11546137 DOI: 10.1016/j.heliyon.2024.e38504] [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: 06/10/2024] [Revised: 08/20/2024] [Accepted: 09/25/2024] [Indexed: 11/16/2024] Open
Abstract
Background Medulloblastoma (MB) is a malignant pediatric central nervous system tumor that is prone to leptomeningeal metastasis. Currently, apart from magnetic resonance imaging and cerebrospinal fluid (CSF) cytology, there are no reliable biomarkers for MB progression. Cytokines are key proteins in signaling pathways in the tumor microenvironment and are closely related to tumor recurrence and progression. This study aimed to investigate the CSF cytokine profile in pediatric patients with MB to identify biomarkers of tumor progression and metastasis. Methods In total, 10 patients were recruited for this study. Five patients had nonmetastatic MB and five had metastatic MB. A cytokine antibody array was used to detect the expression of 120 cytokines in the CSF, and differentially expressed cytokines were screened by integrated bioinformatics analysis. Results Twenty-seven cytokines were upregulated in patients with MB compared to control individuals. Of these, eight were upregulated by > 1.5-fold (CCL2, BMP-4, beta-NGF, FGF-7, IL-12p40, eotaxin-2, M-CSF, and NT-4). Twelve cytokines were differentially expressed between patients with metastatic MB and nonmetastatic (nine cytokines were upregulated and three were downregulated). Among them, NAP-2, MIP-1α, MIP-1β, IGFBP-1, IGFBP-2 and IGFBP-3 were upregulated by more than two-fold. Gene Ontology analysis revealed that the upregulated cytokines were enriched mainly in "epithelial cell proliferation" and "chemotaxis," and the Kyoto Encyclopedia of Genes and Genomes analysis indicated the enrichment of the "MAPK," "PI3K-Akt," and "Ras" signaling pathways. Conclusions The present study investigated cytokine profiles in the CSF of pediatric patients with MB. Our results suggest that these differentially expressed cytokines may serve as novel markers for detecting MB, especially for assessing the risk of progression and metastasis.
Collapse
Affiliation(s)
| | | | | | - Baocheng Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Ma
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Zeissig MN, Hewett DR, Mrozik KM, Panagopoulos V, Wallington-Gates CT, Spencer A, Dold SM, Engelhardt M, Vandyke K, Zannettino ACW. Expression of the chemokine receptor CCR1 decreases sensitivity to bortezomib in multiple myeloma cell lines. Leuk Res 2024; 139:107469. [PMID: 38479337 DOI: 10.1016/j.leukres.2024.107469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND The proteasome inhibitor bortezomib is one of the primary therapies used for the haematological malignancy multiple myeloma (MM). However, intrinsic or acquired resistance to bortezomib, via mechanisms that are not fully elucidated, is a barrier to successful treatment in many patients. Our previous studies have shown that elevated expression of the chemokine receptor CCR1 in MM plasma cells in newly diagnosed MM patients is associated with poor prognosis. Here, we hypothesised that the poor prognosis conferred by CCR1 expression is, in part, due to a CCR1-mediated decrease in MM plasma cell sensitivity to bortezomib. METHODS In order to investigate the role of CCR1 in MM cells, CCR1 was knocked out in human myeloma cell lines OPM2 and U266 using CRISPR-Cas9. Additionally, CCR1 was overexpressed in the mouse MM cell line 5TGM1. The effect of bortezomib on CCR1 knockout or CCR1-overexpressing cells was then assessed by WST-1 assay, with or without CCL3 siRNA knockdown or addition of recombinant human CCL3. NSG mice were inoculated intratibially with OPM2-CCR1KO cells and were treated with 0.7 mg/kg bortezomib or vehicle twice per week for 3 weeks and GFP+ tumour cells in the bone marrow were quantitated by flow cytometry. The effect of CCR1 overexpression or knockout on unfolded protein response pathways was assessed using qPCR for ATF4, HSPA5, XBP1, ERN1 and CHOP and Western blot for IRE1α and p-Jnk. RESULTS Using CCR1 overexpression or CRIPSR-Cas9-mediated CCR1 knockout in MM cell lines, we found that CCR1 expression significantly decreases sensitivity to bortezomib in vitro, independent of the CCR1 ligand CCL3. In addition, CCR1 knockout rendered the human MM cell line OPM2 more sensitive to bortezomib in an intratibial MM model in NSG mice in vivo. Moreover, CCR1 expression negatively regulated the expression of the unfolded protein response receptor IRE1 and downstream target gene XBP1, suggesting this pathway may be responsible for the decreased bortezomib sensitivity of CCR1-expressing cells. CONCLUSIONS Taken together, these studies suggest that CCR1 expression may be associated with decreased response to bortezomib in MM cell lines.
Collapse
Affiliation(s)
- Mara N Zeissig
- Myeloma Research Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia; Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Duncan R Hewett
- Myeloma Research Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia; Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Krzysztof M Mrozik
- Myeloma Research Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia; Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Vasilios Panagopoulos
- Myeloma Research Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia; Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Craig T Wallington-Gates
- College of Medicine and Public Health, Flinders University, Adelaide, Australia; Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia
| | - Andrew Spencer
- Department of Haematology, Alfred Health-Monash University, Melbourne, Australia
| | - Sandra M Dold
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Monika Engelhardt
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kate Vandyke
- Myeloma Research Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia; Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia.
| | - Andrew C W Zannettino
- Myeloma Research Laboratory, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia; Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia; Central Adelaide Local Health Network, Adelaide, Australia
| |
Collapse
|
6
|
Du J, Lin Z, Fu XH, Gu XR, Lu G, Hou J. Research progress of the chemokine/chemokine receptor axes in the oncobiology of multiple myeloma (MM). Cell Commun Signal 2024; 22:177. [PMID: 38475811 PMCID: PMC10935833 DOI: 10.1186/s12964-024-01544-7] [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: 08/29/2023] [Accepted: 02/25/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND The incidence of multiple myeloma (MM), a type of blood cancer affecting monoclonal plasma cells, is rising. Although new drugs and therapies have improved patient outcomes, MM remains incurable. Recent studies have highlighted the crucial role of the chemokine network in MM's pathological mechanism. Gaining a better understanding of this network and creating an overview of chemokines in MM could aid in identifying potential biomarkers and developing new therapeutic strategies and targets. PURPOSE To summarize the complicated role of chemokines in MM, discuss their potential as biomarkers, and introduce several treatments based on chemokines. METHODS Pubmed, Web of Science, ICTRP, and Clinical Trials were searched for articles and research related to chemokines. Publications published within the last 5 years are selected. RESULTS Malignant cells can utilize chemokines, including CCL2, CCL3, CCL5, CXCL7, CXCL8, CXCL12, and CXCL13 to evade apoptosis triggered by immune cells or medication, escape from bone marrow and escalate bone lesions. Other chemokines, including CXCL4, CCL19, and CXCL10, may aid in recruiting immune cells, increasing their cytotoxicity against cancer cells, and inducing apoptosis of malignant cells. CONCLUSION Utilizing anti-tumor chemokines or blocking pro-tumor chemokines may provide new therapeutic strategies for managing MM. Inspired by developed CXCR4 antagonists, including plerixafor, ulocuplumab, and motixafortide, more small molecular antagonists or antibodies for pro-tumor chemokine ligands and their receptors can be developed and used in clinical practice. Along with inhibiting pro-tumor chemokines, studies suggest combining chemokines with chimeric antigen receptor (CAR)-T therapy is promising and efficient.
Collapse
Affiliation(s)
- Jun Du
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zheng Lin
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xue-Hang Fu
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xiao-Ran Gu
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Guang Lu
- Department of Hematology, Shengli Oilfield Central Hospital, Dongying, 257099, China.
| | - Jian Hou
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
7
|
Tsubaki M, Takeda T, Matsuda T, Kishimoto K, Tanaka R, Tsurushima K, Ishizaka T, Nishida S. Bim downregulation by activation of NF-κB p65, Akt, and ERK1/2 is associated with adriamycin and dexamethasone resistance in multiple myeloma cells. Clin Exp Med 2023; 23:1597-1607. [PMID: 36451049 DOI: 10.1007/s10238-022-00951-9] [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: 06/11/2022] [Accepted: 11/14/2022] [Indexed: 12/05/2022]
Abstract
Multiple myeloma (MM) frequently acquires multidrug resistance (MDR), which is due to poor prognosis. Our previous study indicated that high expression of Survivin and multidrug resistance protein 1 (MDR1) and decreased expression of Bim are associated with MDR in adriamycin- and dexamethasone-resistant cells. However, the fundamental mechanism of MDR in adriamycin- and dexamethasone-resistant MM cells is still unidentified. In this study, we examined the MDR mechanism in adriamycin- and dexamethasone-resistant cells. RPMI8226/ADM, ARH-77/ADM, RPMI8226/DEX, and ARH-77/DEX cells exhibited enhanced nuclear factor κB (NF-κB) p65, Akt, and extracellular signal-regulated kinase 1/2 (ERK1/2) activation. Combination treatment with NF-κB p65, phosphoinositide 3-kinase (PI3K), and mitogen-activated protein kinase 1/2 (MEK1/2) inhibitors resensitized to adriamycin and dexamethasone via increased Bim expression. Although treatment with MDR1 or Survivin siRNA did not overcome adriamycin and dexamethasone resistance in RPMI8226/ADM and RPMI8226/DEX cells, administration of Bim siRNA induced adriamycin and dexamethasone resistance in RPMI8226 cells. Moreover, low expression of Bim was related to poor prognosis in MM patients. These results indicate that activation of NF-κB p65, Akt, and ERK1/2 is associated with adriamycin and dexamethasone resistance via decreasing Bim expression, and these signal inhibitor combinations overcome drug resistance in MM. These findings suggest that combination treatment with these inhibitors and adriamycin or dexamethasone may be a promising therapy for adriamycin- and dexamethasone-resistant MM.
Collapse
Affiliation(s)
- Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Tomoya Takeda
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Takuya Matsuda
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Kana Kishimoto
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Remi Tanaka
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Katsumasa Tsurushima
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka, 577-8502, Japan
- Department of Pharmacy, Sakai City Medical Center, Sakai, 593-8304, Japan
| | - Toshihiko Ishizaka
- Department of Pharmacy, Sakai City Medical Center, Sakai, 593-8304, Japan
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka, 577-8502, Japan.
| |
Collapse
|
8
|
Tsubaki M, Takeda T, Matsuda T, Kimura A, Tanaka R, Nagayoshi S, Hoshida T, Tanabe K, Nishida S. Hypoxia-inducible factor 1α inhibitor induces cell death via suppression of BCR-ABL1 and Met expression in BCR-ABL1 tyrosine kinase inhibitor sensitive and resistant chronic myeloid leukemia cells. BMB Rep 2023; 56:78-83. [PMID: 36195570 PMCID: PMC9978365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Indexed: 02/24/2023] Open
Abstract
Chronic myeloid leukemia (CML) has a markedly improved prognosis with the use of breakpoint cluster region-abelson 1 (BCR-ABL1) tyrosine kinase inhibitors (BCR-ABL1 TKIs). However, approximately 40% of patients are resistant or intolerant to BCR-ABL1 TKIs. Hypoxia-inducible factor 1α (HIF-1α) is a hypoxia response factor that has been reported to be highly expressed in CML patients, making it a therapeutic target for BCR-ABL1 TKI-sensitive CML and BCR-ABL1 TKI-resistant CML. In this study, we examined whether HIF-1α inhibitors induce cell death in CML cells and BCR-ABL1 TKI-resistant CML cells. We found that echinomycin and PX-478 induced cell death in BCR-ABL1 TKIs sensitive and resistant CML cells at similar concentrations while the cell sensitivity was not affected with imatinib or dasatinib in BCR-ABL1 TKIs resistant CML cells. In addition, echinomycin and PX-478 inhibited the c-Jun N-terminal kinase (JNK), Akt, and extracellular-regulated protein kinase 1/2 (ERK1/2) activation via suppression of BCR-ABL1 and Met expression in BCR-ABL1 sensitive and resistant CML cells. Moreover, treatment with HIF-1α siRNA induced cell death by inhibiting BCR-ABL1 and Met expression and activation of JNK, Akt, and ERK1/2 in BCR-ABL1 TKIs sensitive and resistant CML cells. These results indicated that HIF-1α regulates BCR-ABL and Met expression and is involved in cell survival in CML cells, suggesting that HIF-1α inhibitors induce cell death in BCR-ABL1 TKIs sensitive and resistant CML cells and therefore HIF-1α inhibitors are potential candidates for CML treatment. [BMB Reports 2023; 56(2): 78-83].
Collapse
Affiliation(s)
- Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae 577-8502, Higashi-Osaka, Wakayama 640-8558, Japan
| | - Tomoya Takeda
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae 577-8502, Higashi-Osaka, Wakayama 640-8558, Japan
| | - Takuya Matsuda
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae 577-8502, Higashi-Osaka, Wakayama 640-8558, Japan
| | - Akihiro Kimura
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae 577-8502, Higashi-Osaka, Wakayama 640-8558, Japan
| | - Remi Tanaka
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae 577-8502, Higashi-Osaka, Wakayama 640-8558, Japan
| | - Sakiko Nagayoshi
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae 577-8502, Higashi-Osaka, Wakayama 640-8558, Japan
| | - Tadafumi Hoshida
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae 577-8502, Higashi-Osaka, Wakayama 640-8558, Japan,Department of Pharmacy, Japanese Red Cross Society Wakayama Medical Center, Wakayama 640-8558, Japan
| | - Kazufumi Tanabe
- Department of Pharmacy, Japanese Red Cross Society Wakayama Medical Center, Wakayama 640-8558, Japan
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae 577-8502, Higashi-Osaka, Wakayama 640-8558, Japan,Corresponding author. Tel: +81-6-6721-2332; Fax: +81-6-6730-1394; E-mail:
| |
Collapse
|
9
|
Tsubaki M, Takeda T, Koumoto Y, Usami T, Matsuda T, Seki S, Sakai K, Nishio K, Nishida S. Activation of ERK1/2 by MOS and TPL2 leads to dasatinib resistance in chronic myeloid leukaemia cells. Cell Prolif 2023:e13420. [PMID: 36847709 DOI: 10.1111/cpr.13420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/21/2023] [Accepted: 01/27/2023] [Indexed: 03/01/2023] Open
Abstract
The development of BCR::ABL1 tyrosine kinase inhibitors (TKIs), such as dasatinib, has dramatically improved survival in cases of chronic myeloid leukaemia (CML). However, the development of resistance to BCR::ABL1 TKIs is a clinical problem. BCR::ABL1 TKI resistance is known to have BCR::ABL1-dependent or BCR::ABL1-independent mechanisms, but the mechanism of BCR::ABL1 independence is not well understood. In the present study, we investigated the mechanism of BCR::ABL1-independent dasatinib resistance. The expression and activation level of genes or proteins were evaluated using array CGH, real time PCR, or western blot analysis. Gene expression was modulated using siRNA-mediated knockdown. Cell survival was assessed by using trypan blue dye method. We found that dasatinib-resistant K562/DR and KU812/DR cells did not harbour a BCR::ABL1 mutation but had elevated expression and/or activation of MOS, TPL2 and ERK1/2. In addition, MOS siRNA, TPL2 siRNA and trametinib resensitized dasatinib-resistant cells to dasatinib. Moreover, expression levels of MOS in dasatinib non-responder patients with CML were higher than those in dasatinib responders, and the expression of TPL2 tended to increase in dasatinib non-responder patients compared with that in responder patients. Our results indicate that activation of ERK1/2 by elevated MOS and TPL2 expression is involved in dasatinib resistance, and inhibition of these proteins overcomes dasatinib resistance. Therefore, MOS, TPL2 and ERK1/2 inhibitors may be therapeutically useful for treating BCR::ABL1-independent dasatinib-resistant CML.
Collapse
Affiliation(s)
- Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| | - Tomoya Takeda
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| | - Yuuichi Koumoto
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| | - Takehiro Usami
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| | - Takuya Matsuda
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| | - Shiori Seki
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| | - Kazuko Sakai
- Department of Genome Biology, Kindai University School of Medicine, Osakasayama, Osaka, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University School of Medicine, Osakasayama, Osaka, Japan
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka, Japan
| |
Collapse
|
10
|
Tsubaki M, Takeda T, Matsuda T, Kimura A, Tanaka R, Nagayoshi S, Hoshida T, Tanabe K, Nishida S. Hypoxia-inducible factor 1α inhibitor induces cell death via suppression of BCR-ABL1 and Met expression in BCR-ABL1 tyrosine kinase inhibitor sensitive and resistant chronic myeloid leukemia cells. BMB Rep 2023; 56:78-83. [PMID: 36195570 PMCID: PMC9978365 DOI: 10.5483/bmbrep.2022-0095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/21/2022] [Accepted: 09/16/2022] [Indexed: 08/18/2023] Open
Abstract
Chronic myeloid leukemia (CML) has a markedly improved prognosis with the use of breakpoint cluster region-abelson 1 (BCR-ABL1) tyrosine kinase inhibitors (BCR-ABL1 TKIs). However, approximately 40% of patients are resistant or intolerant to BCR-ABL1 TKIs. Hypoxia-inducible factor 1α (HIF-1α) is a hypoxia response factor that has been reported to be highly expressed in CML patients, making it a therapeutic target for BCR-ABL1 TKI-sensitive CML and BCR-ABL1 TKI-resistant CML. In this study, we examined whether HIF-1α inhibitors induce cell death in CML cells and BCR-ABL1 TKI-resistant CML cells. We found that echinomycin and PX-478 induced cell death in BCR-ABL1 TKIs sensitive and resistant CML cells at similar concentrations while the cell sensitivity was not affected with imatinib or dasatinib in BCR-ABL1 TKIs resistant CML cells. In addition, echinomycin and PX-478 inhibited the c-Jun N-terminal kinase (JNK), Akt, and extracellular-regulated protein kinase 1/2 (ERK1/2) activation via suppression of BCR-ABL1 and Met expression in BCR-ABL1 sensitive and resistant CML cells. Moreover, treatment with HIF-1α siRNA induced cell death by inhibiting BCR-ABL1 and Met expression and activation of JNK, Akt, and ERK1/2 in BCR-ABL1 TKIs sensitive and resistant CML cells. These results indicated that HIF-1α regulates BCR-ABL and Met expression and is involved in cell survival in CML cells, suggesting that HIF-1α inhibitors induce cell death in BCR-ABL1 TKIs sensitive and resistant CML cells and therefore HIF-1α inhibitors are potential candidates for CML treatment. [BMB Reports 2023; 56(2): 78-83].
Collapse
MESH Headings
- Humans
- Tyrosine Protein Kinase Inhibitors
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Echinomycin/therapeutic use
- Proto-Oncogene Proteins c-akt
- Drug Resistance, Neoplasm
- Apoptosis
- Protein Kinase Inhibitors/pharmacology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Cell Death
Collapse
Affiliation(s)
- Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae 577-8502, Higashi-Osaka, Wakayama 640-8558, Japan
| | - Tomoya Takeda
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae 577-8502, Higashi-Osaka, Wakayama 640-8558, Japan
| | - Takuya Matsuda
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae 577-8502, Higashi-Osaka, Wakayama 640-8558, Japan
| | - Akihiro Kimura
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae 577-8502, Higashi-Osaka, Wakayama 640-8558, Japan
| | - Remi Tanaka
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae 577-8502, Higashi-Osaka, Wakayama 640-8558, Japan
| | - Sakiko Nagayoshi
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae 577-8502, Higashi-Osaka, Wakayama 640-8558, Japan
| | - Tadafumi Hoshida
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae 577-8502, Higashi-Osaka, Wakayama 640-8558, Japan
- Department of Pharmacy, Japanese Red Cross Society Wakayama Medical Center, Wakayama 640-8558, Japan
| | - Kazufumi Tanabe
- Department of Pharmacy, Japanese Red Cross Society Wakayama Medical Center, Wakayama 640-8558, Japan
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae 577-8502, Higashi-Osaka, Wakayama 640-8558, Japan
| |
Collapse
|
11
|
Dimethyl Fumarate Induces Apoptosis via Inhibition of NF-κB and Enhances the Effect of Paclitaxel and Adriamycin in Human TNBC Cells. Int J Mol Sci 2022; 23:ijms23158681. [PMID: 35955813 PMCID: PMC9369077 DOI: 10.3390/ijms23158681] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/29/2022] [Accepted: 08/03/2022] [Indexed: 02/05/2023] Open
Abstract
Triple-negative breast cancer (TNBC) has the poorest prognosis of all breast cancer subtypes. Recently, the activation of NF-κB, which is involved in the growth and survival of malignant tumors, has been demonstrated in TNBC, suggesting that NF-κB may serve as a new therapeutic target. In the present study, we examined whether dimethyl fumarate (DMF), an NF-κB inhibitor, induces apoptosis in TNBC cells and enhances the apoptosis-inducing effect of paclitaxel and adriamycin. Cell survival was analyzed by the trypan blue assay and apoptosis assay. Protein detection was examined by immunoblotting. The activation of NF-κB p65 was correlated with poor prognosis in patients with TNBC. DMF induced apoptosis in MDA-MB-231 and BT-549 cells at concentrations that were non-cytotoxic to the normal mammary cell line MCF-10A. Furthermore, DMF inhibited NF-κB nuclear translocation and Survivin, XIAP, Bcl-xL, and Bcl-2 expression in MDA-MB-231 and BT-549 cells. Moreover, DMF enhanced the apoptosis-inducing effect of paclitaxel and adriamycin in MDA-MB-231 cells. These findings suggest that DMF may be an effective therapeutic agent for the treatment of TNBC, in which NF-κB is constitutively active. DMF may also be useful as an adjuvant therapy to conventional anticancer drugs.
Collapse
|
12
|
Cheng Q, Zhao F, Zhang B, Zhang Y, Cai L, Qiao B, Hu Y, Sun C. Prognostic nomogram incorporating cytokines for overall survival in patients with newly diagnosed multiple myeloma. Int Immunopharmacol 2021; 99:108016. [PMID: 34385029 DOI: 10.1016/j.intimp.2021.108016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 02/01/2023]
Abstract
OBJECTIVE The purpose of this study was to explore the relationship between pretreatment cytokine status and overall survival and establish a prognostic nomogram incorporating cytokines in newly diagnosed multiple myeloma (NDMM) patients. METHODS A total of 121 patients with NDMM from the Wuhan Union Hospital were included in our study. Patient serum levels of cytokines, including macrophage inflammatory protein 1 alpha (MIP-1α), migration inhibitory factor (MIF), tumor necrosis factor-α (TNF-α), vascular endothelial growth factor-α (VEGF-α), monocyte chemoattractant protein-1 (MCP-1) and soluble interleukins IL-17A, IL-6, IL-21 and IL-10 were assessed before treatment. Based on the results of the multivariate Cox proportional hazards model, we developed a prognostic nomogram. We used the concordance index (C-index) and a calibration curve to measure the predictive performance of the nomogram. RESULTS Three important variables (lactate dehydrogenase, MIP-1α and creatinine) were incorporated in the nomogram using multivariate Cox analysis. The 3-year overall survival (OS) rate and progression-free survival (PFS) rate were 83.8% and 21.8% in the low-risk group of the nomogram and 17.4% and 8.4% in the high-risk group, respectively. The C-index of the nomogram for OS prediction was 0.80 (95% CI: 0.68-0.92), showing superiority over the predictive power of the Durie-Salmon staging system (C-index = 0.58; 95% CI: 0.49-0.67), International Staging System (C-index = 0.70; 95% CI: 0.61-0.79) and Revised-International Staging System (C-index = 0.71; 95% CI: 0.63-0.80). The calibration curve showed that the nomogram accurately predicted the 1-year, 2-year and 3-year OS of NDMM patients. CONCLUSION The established nomogram provides accurate and individualized OS risk estimation for NDMM patients.
Collapse
Affiliation(s)
- Qianwen Cheng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Fei Zhao
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Bo Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Yuyang Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Li Cai
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Bing Qiao
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China.
| | - Chunyan Sun
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China.
| |
Collapse
|
13
|
Abstract
Secretory proteins in tumor tissues are important components of the tumor microenvironment. Secretory proteins act on tumor cells or stromal cells or mediate interactions between tumor cells and stromal cells, thereby affecting tumor progression and clinical treatment efficacy. In this paper, recent research advances in secretory proteins in malignant tumors are reviewed.
Collapse
Affiliation(s)
- Na Zhang
- State Key Laboratory of Molecular Oncology, Center for Cancer Precision Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jiajie Hao
- State Key Laboratory of Molecular Oncology, Center for Cancer Precision Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yan Cai
- State Key Laboratory of Molecular Oncology, Center for Cancer Precision Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Mingrong Wang
- State Key Laboratory of Molecular Oncology, Center for Cancer Precision Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
14
|
Activation of Serum/Glucocorticoid Regulated Kinase 1/Nuclear Factor-κB Pathway Are Correlated with Low Sensitivity to Bortezomib and Ixazomib in Resistant Multiple Myeloma Cells. Biomedicines 2021; 9:biomedicines9010033. [PMID: 33406639 PMCID: PMC7823718 DOI: 10.3390/biomedicines9010033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/14/2020] [Accepted: 12/30/2020] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is an incurable malignancy often associated with primary and acquired resistance to therapeutic agents, such as proteasome inhibitors. However, the mechanisms underlying the proteasome inhibitor resistance are poorly understood. Here, we elucidate the mechanism of primary resistance to bortezomib and ixazomib in the MM cell lines, KMS-20, KMS-26, and KMS-28BM. We find that low bortezomib and ixazomib concentrations induce cell death in KMS-26 and KMS-28BM cells. However, high bortezomib and ixazomib concentrations induce cell death only in KMS-20 cells. During Gene Expression Omnibus analysis, KMS-20 cells exhibit high levels of expression of various genes, including anti-phospho-fibroblast growth factor receptor 1 (FGFR1), chemokine receptor type (CCR2), and serum and glucocorticoid regulated kinase (SGK)1. The SGK1 inhibitor enhances the cytotoxic effects of bortezomib and ixazomib; however, FGFR1 and CCR2 inhibitors do not show such effect in KMS-20 cells. Moreover, SGK1 activation induces the phosphorylation of NF-κB p65, and an NF-κB inhibitor enhances the sensitivity of KMS-20 cells to bortezomib and ixazomib. Additionally, high levels of expression of SGK1 and NF-κB p65 is associated with a low sensitivity to bortezomib and a poor prognosis in MM patients. These results indicate that the activation of the SGK1/NF-κB pathway correlates with a low sensitivity to bortezomib and ixazomib, and a combination of bortezomib and ixazomib with an SGK1 or NF-κB inhibitor may be involved in the treatment of MM via activation of the SGK1/NF-κB pathway.
Collapse
|
15
|
Tsubaki M, Seki S, Takeda T, Chihara A, Arai Y, Morii Y, Imano M, Satou T, Shimomura K, Nishida S. The HGF/Met/NF-κB Pathway Regulates RANKL Expression in Osteoblasts and Bone Marrow Stromal Cells. Int J Mol Sci 2020; 21:ijms21217905. [PMID: 33114380 PMCID: PMC7663721 DOI: 10.3390/ijms21217905] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/16/2020] [Accepted: 10/22/2020] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma (MM)-induced bone disease occurs through hyperactivation of osteoclasts by several factors secreted by MM cells. MM cell-secreted factors induce osteoclast differentiation and activation via direct and indirect actions including enhanced expression of receptor activator of nuclear factor κB ligand (RANKL) in osteoblasts and bone marrow stromal cells (BMSCs). Hepatocyte growth factor (HGF) is elevated in MM patients and is associated with MM-induced bone disease, although the mechanism by which HGF promotes bone disease remains unclear. In the present study, we demonstrated that HGF induces RANKL expression in osteoblasts and BMSCs, and investigated the mechanism of induction. We found that HGF and MM cell supernatants induced RANKL expression in ST2 cells, MC3T3-E1 cells, and mouse BMSCs. In addition, HGF increased phosphorylation of Met and nuclear factor κB (NF-κB) in ST2 cells, MC3T3-E1 cells, or mouse BMSCs. Moreover, Met and NF-κB inhibitors suppressed HGF-induced RANKL expression in ST2 cells, MC3T3-E1 cells, and mouse BMSCs. These results indicated that HGF promotes RANKL expression in osteoblasts and BMSCs via the Met/NF-κB signaling pathway, and Met and NF-κB inhibitors suppressed HGF-induced RANKL expression. Our findings suggest that Met and NF-κB inhibitors are potentially useful in mitigating MM-induced bone disease in patients expressing high levels of HGF.
Collapse
Affiliation(s)
- Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (S.S.); (T.T.); (A.C.); (Y.A.); (Y.M.)
| | - Shiori Seki
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (S.S.); (T.T.); (A.C.); (Y.A.); (Y.M.)
| | - Tomoya Takeda
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (S.S.); (T.T.); (A.C.); (Y.A.); (Y.M.)
| | - Akiko Chihara
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (S.S.); (T.T.); (A.C.); (Y.A.); (Y.M.)
| | - Yuuko Arai
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (S.S.); (T.T.); (A.C.); (Y.A.); (Y.M.)
| | - Yuusuke Morii
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (S.S.); (T.T.); (A.C.); (Y.A.); (Y.M.)
- Department of Pharmacy, Municipal Ikeda Hospital, Ikeda 563-0025, Japan;
| | - Motohiro Imano
- Department of Surgery, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-0014, Japan;
| | - Takao Satou
- Department of Pathology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-0014, Japan;
| | - Kazunori Shimomura
- Department of Pharmacy, Municipal Ikeda Hospital, Ikeda 563-0025, Japan;
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (S.S.); (T.T.); (A.C.); (Y.A.); (Y.M.)
- Correspondence: ; Tel.: +81-6-6721-2332
| |
Collapse
|
16
|
Korbecki J, Grochans S, Gutowska I, Barczak K, Baranowska-Bosiacka I. CC Chemokines in a Tumor: A Review of Pro-Cancer and Anti-Cancer Properties of Receptors CCR5, CCR6, CCR7, CCR8, CCR9, and CCR10 Ligands. Int J Mol Sci 2020; 21:ijms21207619. [PMID: 33076281 PMCID: PMC7590012 DOI: 10.3390/ijms21207619] [Citation(s) in RCA: 225] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/05/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
CC chemokines (or β-chemokines) are 28 chemotactic cytokines with an N-terminal CC domain that play an important role in immune system cells, such as CD4+ and CD8+ lymphocytes, dendritic cells, eosinophils, macrophages, monocytes, and NK cells, as well in neoplasia. In this review, we discuss human CC motif chemokine ligands: CCL1, CCL3, CCL4, CCL5, CCL18, CCL19, CCL20, CCL21, CCL25, CCL27, and CCL28 (CC motif chemokine receptor CCR5, CCR6, CCR7, CCR8, CCR9, and CCR10 ligands). We present their functioning in human physiology and in neoplasia, including their role in the proliferation, apoptosis resistance, drug resistance, migration, and invasion of cancer cells. We discuss the significance of chemokine receptors in organ-specific metastasis, as well as the influence of each chemokine on the recruitment of various cells to the tumor niche, such as cancer-associated fibroblasts (CAF), Kupffer cells, myeloid-derived suppressor cells (MDSC), osteoclasts, tumor-associated macrophages (TAM), tumor-infiltrating lymphocytes (TIL), and regulatory T cells (Treg). Finally, we show how the effect of the chemokines on vascular endothelial cells and lymphatic endothelial cells leads to angiogenesis and lymphangiogenesis.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (S.G.)
| | - Szymon Grochans
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (S.G.)
| | - Izabela Gutowska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Katarzyna Barczak
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University, Powstańców Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (S.G.)
- Correspondence: ; Tel.: +48-914661515
| |
Collapse
|
17
|
Korbecki J, Kojder K, Barczak K, Simińska D, Gutowska I, Chlubek D, Baranowska-Bosiacka I. Hypoxia Alters the Expression of CC Chemokines and CC Chemokine Receptors in a Tumor-A Literature Review. Int J Mol Sci 2020; 21:ijms21165647. [PMID: 32781743 PMCID: PMC7460668 DOI: 10.3390/ijms21165647] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 02/06/2023] Open
Abstract
Hypoxia, i.e., oxygen deficiency condition, is one of the most important factors promoting the growth of tumors. Since its effect on the chemokine system is crucial in understanding the changes in the recruitment of cells to a tumor niche, in this review we have gathered all the available data about the impact of hypoxia on β chemokines. In the introduction, we present the chronic (continuous, non-interrupted) and cycling (intermittent, transient) hypoxia together with the mechanisms of activation of hypoxia inducible factors (HIF-1 and HIF-2) and NF-κB. Then we describe the effect of hypoxia on the expression of chemokines with the CC motif: CCL1, CCL2, CCL3, CCL4, CCL5, CCL7, CCL8, CCL11, CCL13, CCL15, CCL16, CCL17, CCL18, CCL19, CCL20, CCL21, CCL22, CCL24, CCL25, CCL26, CCL27, CCL28 together with CC chemokine receptors: CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, and CCR10. To better understand the effect of hypoxia on neoplastic processes and changes in the expression of the described proteins, we summarize the available data in a table which shows the effect of individual chemokines on angiogenesis, lymphangiogenesis, and recruitment of eosinophils, myeloid-derived suppressor cells (MDSC), regulatory T cells (Treg), and tumor-associated macrophages (TAM) to a tumor niche.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (J.K.); (D.S.); (D.C.)
| | - Klaudyna Kojder
- Department of Anaesthesiology and Intensive Care, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-281 Szczecin, Poland;
| | - Katarzyna Barczak
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Donata Simińska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (J.K.); (D.S.); (D.C.)
| | - Izabela Gutowska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (J.K.); (D.S.); (D.C.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (J.K.); (D.S.); (D.C.)
- Correspondence: ; Tel.: +48-914661515; Fax: +48-914661516
| |
Collapse
|
18
|
Galland S, Martin P, Fregni G, Letovanec I, Stamenkovic I. Attenuation of the pro-inflammatory signature of lung cancer-derived mesenchymal stromal cells by statins. Cancer Lett 2020; 484:50-64. [PMID: 32418888 DOI: 10.1016/j.canlet.2020.05.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/20/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023]
Abstract
Solid tumor growth triggers a dynamic host response, which recapitulates wound healing and defines the tumor microenvironment (TME). In addition to the action of the tumor cells themselves, the TME is maintained by a myriad of immune and stromal cell-derived soluble mediators and extracellular matrix components whose combined action supports tumor progression. However, therapeutic targeting of the TME has proven challenging because of incomplete understanding of the tumor-host crosstalk at the molecular level. Here, we investigated the crosstalk between mesenchymal stromal cells (MSCs) and primary cancer cells (PCCs) from human squamous cell lung carcinoma (SCC). We discovered that PCCs secrete CCL3 and stimulate IL-6, CCL2, ICAM-1 and VCAM-1 expression in MSCs and that the MSC-PCC crosstalk can be disrupted by the lipid-lowering drug simvastatin, which displays pleiotropic effects on cell metabolism and suppresses IL-6 and CCL2 production by MSCs and CCL3 secretion by PCCs. In addition, simvastatin inhibited spheroid formation by PCCs and negatively affected PCC survival. Our observations demonstrate that commonly used statins may be repurposed to target the TME in lung carcinoma.
Collapse
Affiliation(s)
- Sabine Galland
- Experimental Pathology Service, Institute of Pathology, CHUV, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 25, 1011, Lausanne, Switzerland.
| | - Patricia Martin
- Experimental Pathology Service, Institute of Pathology, CHUV, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 25, 1011, Lausanne, Switzerland
| | - Giulia Fregni
- Experimental Pathology Service, Institute of Pathology, CHUV, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 25, 1011, Lausanne, Switzerland
| | - Igor Letovanec
- Clinical Pathology Service, Institute of Pathology, CHUV, Rue du Bugnon 25, 1011, Lausanne, Switzerland
| | - Ivan Stamenkovic
- Experimental Pathology Service, Institute of Pathology, CHUV, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 25, 1011, Lausanne, Switzerland
| |
Collapse
|
19
|
Dasatinib reverses drug resistance by downregulating MDR1 and Survivin in Burkitt lymphoma cells. BMC Complement Med Ther 2020; 20:84. [PMID: 32171300 PMCID: PMC7076888 DOI: 10.1186/s12906-020-2879-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 03/04/2020] [Indexed: 12/17/2022] Open
Abstract
Background Current chemotherapies for Burkitt lymphoma (BL) have dramatically improved its clinical outcome. However, chemoresistance can lead to chemotherapy failure and very poor prognosis; thus, novel strategies are urgently required for patients with drug-resistant BL. To investigate the mechanisms underlying drug resistance in BL, we established drug-resistant BL cell lines: HS-Sultan/ADM (adriamycin-resistant), HS-Sultan/VCR (vincristine-resistant), HS-Sultan/DEX (dexamethasone-resistant), and HS-Sultan/L-PAM (melphalan-resistant). Methods Drug transporter and survival factor expression were investigated the using western blotting and real time polymerase chain reaction. Cell survival was analyzed by trypan blue dye exclusion method. Results The established cell lines acquired cross-resistance to adriamycin, vincristine, dexamethasone, and melphalan and exhibited 50% inhibitory concentration values 106-, 40-, 81-, and 45-fold higher than the parental cell lines, respectively. We found that protein and mRNA expression of MDR1 and Survivin were higher in drug-resistant BL cells than in the parent cells. Treatment with verapamil, an MDR1 inhibitor, or Survivin siRNA alongside each anti-cancer drug suppressed the proliferation of all drug-resistant BL cells. Src kinase activity was higher in all resistant cell lines than the parental cells; suppressing Src with dasatinib restored drug sensitivity by reducing MDR1 and Survivin expression. Conclusions MDR1 and Survivin upregulation are responsible for resistance to conventional drugs and dasatinib can restore drug sensitivity by reducing MDR1 and Survivin expression in drug-resistant BL cells. Src inhibitors could therefore be a novel treatment strategy for patients with drug resistant BL.
Collapse
|
20
|
Tsubaki M, Takeda T, Noguchi M, Jinushi M, Seki S, Morii Y, Shimomura K, Imano M, Satou T, Nishida S. Overactivation of Akt Contributes to MEK Inhibitor Primary and Acquired Resistance in Colorectal Cancer Cells. Cancers (Basel) 2019; 11:cancers11121866. [PMID: 31769426 PMCID: PMC6966459 DOI: 10.3390/cancers11121866] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/21/2019] [Accepted: 11/21/2019] [Indexed: 12/21/2022] Open
Abstract
RAS and BRAF-mutated colorectal cancers are associated with resistance to chemotherapy and poor prognosis, highlighting the need for new therapeutic strategies. Although these cancers sometimes respond to mitogen activated protein kinase kinase (MEK) inhibitor treatment, they often acquire resistance via mechanisms, which are poorly understood. Here, we investigated the mechanism of MEK inhibitor resistance in primary- and acquired-resistant cells. Cell viability was examined using the trypan blue dye exclusion assay. Protein expression was analyzed by western blotting. Somatic mutations in colorectal cancer cells were investigated using the polymerase chain reaction array. PD0325901 and trametinib induced cell death in LoVo and Colo-205 cells but not in DLD-1 and HT-29 cells, which have a PIK3CA mutation constitutively activating Akt and NF-κB. Treatment with PD0325901 and trametinib suppressed ERK1/2 activation in all four cell lines but only induced Akt and NF-κB activation in DLD-1 and HT-29 cells. Inhibition of Akt but not NF-κB, overcame MEK inhibitor resistance in DLD-1 and HT-29 cells. Acquired-resistant LoVo/PR, Colo-205/PR and LoVo/TR cells have constitutively active Akt due to a M1043V mutation in the kinase activation loop of PIK3CA and Akt inhibitor resensitized these cells to MEK inhibitor. These results demonstrate that the overactivation of Akt plays a critical role in MEK inhibitor primary and acquired resistance and implicate combined Akt/MEK inhibition as a potentially useful treatment for RAS/BRAF-mutated colorectal cancer.
Collapse
Affiliation(s)
- Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
| | - Tomoya Takeda
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
| | - Masaki Noguchi
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
| | - Minami Jinushi
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
| | - Shiori Seki
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
| | - Yuusuke Morii
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
- Department of Phamacy, Municipal Ikeda Hospital, Ikeda, Osaka 563-8510, Japan;
| | - Kazunori Shimomura
- Department of Phamacy, Municipal Ikeda Hospital, Ikeda, Osaka 563-8510, Japan;
| | - Motohiro Imano
- Department of Surgery, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-0014, Japan;
| | - Takao Satou
- Department of Pathology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-0014, Japan.;
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University Faculty of Pharmacy, Kowakae, Higashi-Osaka 577-8502, Japan; (M.T.); (T.T.); (M.N.); (M.J.); (S.S.); (Y.M.)
- Correspondence:
| |
Collapse
|
21
|
Tsubaki M, Takeda T, Tomonari Y, Koumoto YI, Imano M, Satou T, Nishida S. Overexpression of HIF-1α contributes to melphalan resistance in multiple myeloma cells by activation of ERK1/2, Akt, and NF-κB. J Transl Med 2019; 99:72-84. [PMID: 30353128 DOI: 10.1038/s41374-018-0114-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 07/07/2018] [Accepted: 07/13/2018] [Indexed: 12/20/2022] Open
Abstract
Multiple myeloma (MM) commonly displays multidrug resistance and is associated with poor prognosis. Therefore, it is important to identify the mechanisms by which MM cells develop multidrug resistance. Our previous study showed that multidrug resistance is correlated with overexpression of multidrug resistance protein 1 (MDR1) and Survivin, and downregulation of Bim expression in melphalan-resistant RPMI8226/L-PAM cells; however, the underlying mechanism of multidrug resistance remains unclear. In the present study, we investigated the mechanism of multidrug resistance in melphalan-resistant cells. We found that RPMI8226/L-PAM and ARH-77/L-PAM cells showed increased phosphorylation of extracellular signal-regulated protein kinase 1/2 (ERK1/2) and Akt, and nuclear localization of nuclear factor κB (NF-κB). The combination of ERK1/2, Akt, and NF-κB inhibitors with melphalan reversed melphalan resistance via suppression of Survivin expression and enhanced Bim expression in melphalan-resistant cells. In addition, RPMI8226/L-PAM and ARH-77/L-PAM cells overexpressed hypoxia-inducible factor 1α (HIF-1α) via activation of ERK1/2, Akt, and NF-κB. Moreover, suppression of HIF-1α by echinomycin or HIF-1α siRNA resensitized RPMI8226/L-PAM cells to melphalan through downregulation of Survivin expression and upregulation of Bim expression. These results indicate that enhanced Survivin expression and decreased Bim expression by HIF-1α via activation of ERK1/2, Akt, and NF-κB play a critical role in melphalan resistance. Our findings suggest that HIF-1α, ERK1/2, Akt, and NF-κB inhibitors are potentially useful as anti-MDR agents for the treatment of melphalan-resistant MM.
Collapse
Affiliation(s)
- Masanobu Tsubaki
- Division of Pharmacotherapy, Faculty of Pharmacy, Kindai University, Kowakae, Higashi-Osaka, Japan
| | - Tomoya Takeda
- Division of Pharmacotherapy, Faculty of Pharmacy, Kindai University, Kowakae, Higashi-Osaka, Japan
| | - Yoshika Tomonari
- Division of Pharmacotherapy, Faculty of Pharmacy, Kindai University, Kowakae, Higashi-Osaka, Japan
| | - Yu-Ichi Koumoto
- Division of Pharmacotherapy, Faculty of Pharmacy, Kindai University, Kowakae, Higashi-Osaka, Japan
| | - Motohiro Imano
- Department of Surgery, Faculty of Medicine, Kindai University, Osakasayama, Osaka, Japan
| | - Takao Satou
- Department of Pathology, Faculty of Medicine, Kindai University, Osakasayama, Osaka, Japan
| | - Shozo Nishida
- Division of Pharmacotherapy, Faculty of Pharmacy, Kindai University, Kowakae, Higashi-Osaka, Japan.
| |
Collapse
|
22
|
Mashimo K, Tsubaki M, Takeda T, Asano R, Jinushi M, Imano M, Satou T, Sakaguchi K, Nishida S. RANKL-induced c-Src activation contributes to conventional anti-cancer drug resistance and dasatinib overcomes this resistance in RANK-expressing multiple myeloma cells. Clin Exp Med 2018; 19:133-141. [DOI: 10.1007/s10238-018-0531-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/27/2018] [Indexed: 12/23/2022]
|
23
|
Farrell ML, Reagan MR. Soluble and Cell-Cell-Mediated Drivers of Proteasome Inhibitor Resistance in Multiple Myeloma. Front Endocrinol (Lausanne) 2018; 9:218. [PMID: 29765356 PMCID: PMC5938346 DOI: 10.3389/fendo.2018.00218] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 04/17/2018] [Indexed: 12/17/2022] Open
Abstract
It is becoming clear that myeloma cell-induced disruption of the highly organized bone marrow components (both cellular and extracellular) results in destruction of the marrow and support for multiple myeloma (MM) cell proliferation, survival, migration, and drug resistance. Since the first phase I clinical trial on bortezomib was published 15 years ago, proteasome inhibitors (PIs) have become increasingly common for treatment of MM and are currently an essential part of any anti-myeloma combination therapy. PIs, either the first generation (bortezomib), second generation (carfilzomib) or oral agent (ixazomib), all take advantage of the heavy reliance of myeloma cells on the 26S proteasome for their degradation of excessive or misfolded proteins. Inhibiting the proteasome can create a crisis specifically for myeloma cells due to their rapid production of immunoglobulins. PIs have relatively few side effects and can be very effective, especially in combination therapy. If PI resistance can be overcome, these drugs may prove even more useful to a greater range of patients. Both soluble and insoluble (contact mediated) signals drive PI-resistance via activation of various intracellular signaling pathways. This review discusses the currently known mechanisms of non-autonomous (microenvironment dependent) mechanisms of PI resistance in myeloma cells. We also introduce briefly cell-autonomous and stress-mediated mechanisms of PI resistance. Our goal is to help researchers design better ways to study and overcome PI resistance, to ultimately design better combination therapies.
Collapse
Affiliation(s)
- Mariah L. Farrell
- Reagan Laboratory, Maine Medical Center Research Institute, Scarborough, ME, United States
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States
- School of Medicine, Tufts University, Boston, MA, United States
- Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, United States
| | - Michaela R. Reagan
- Reagan Laboratory, Maine Medical Center Research Institute, Scarborough, ME, United States
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States
- School of Medicine, Tufts University, Boston, MA, United States
- Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, United States
- *Correspondence: Michaela R. Reagan,
| |
Collapse
|