1
|
Dunphy K, Bazou D, Henry M, Meleady P, Miettinen JJ, Heckman CA, Dowling P, O’Gorman P. Proteomic and Metabolomic Analysis of Bone Marrow and Plasma from Patients with Extramedullary Multiple Myeloma Identifies Distinct Protein and Metabolite Signatures. Cancers (Basel) 2023; 15:3764. [PMID: 37568580 PMCID: PMC10417544 DOI: 10.3390/cancers15153764] [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: 07/05/2023] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 08/13/2023] Open
Abstract
Multiple myeloma (MM) is an incurable haematological malignancy of plasma cells in the bone marrow. In rare cases, an aggressive form of MM called extramedullary multiple myeloma (EMM) develops, where myeloma cells enter the bloodstream and colonise distal organs or soft tissues. This variant is associated with refractoriness to conventional therapies and a short overall survival. The molecular mechanisms associated with EMM are not yet fully understood. Here, we analysed the proteome of bone marrow mononuclear cells and blood plasma from eight patients (one serial sample) with EMM and eight patients without extramedullary spread. The patients with EMM had a significantly reduced overall survival with a median survival of 19 months. Label-free mass spectrometry revealed 225 proteins with a significant differential abundance between bone marrow mononuclear cells (BMNCs) isolated from patients with MM and EMM. This plasma proteomics analysis identified 22 proteins with a significant differential abundance. Three proteins, namely vascular cell adhesion molecule 1 (VCAM1), pigment epithelium derived factor (PEDF), and hepatocyte growth factor activator (HGFA), were verified as the promising markers of EMM, with the combined protein panel showing excellent accuracy in distinguishing EMM patients from MM patients. Metabolomic analysis revealed a distinct metabolite signature in EMM patient plasma compared to MM patient plasma. The results provide much needed insight into the phenotypic profile of EMM and in identifying promising plasma-derived markers of EMM that may inform novel drug development strategies.
Collapse
Affiliation(s)
- Katie Dunphy
- Department of Biology, Maynooth University, W23 F2K8 Kildare, Ireland;
| | - Despina Bazou
- Department of Haematology, Mater Misericordiae University Hospital, D07 AX57 Dublin, Ireland; (D.B.); (P.O.)
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, D09 NR58 Dublin, Ireland; (M.H.); (P.M.)
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, D09 NR58 Dublin, Ireland; (M.H.); (P.M.)
| | - Juho J. Miettinen
- Institute for Molecular Medicine Finland-FIMM, HiLIFE–Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00290 Helsinki, Finland; (J.J.M.); (C.A.H.)
| | - Caroline A. Heckman
- Institute for Molecular Medicine Finland-FIMM, HiLIFE–Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00290 Helsinki, Finland; (J.J.M.); (C.A.H.)
| | - Paul Dowling
- Department of Biology, Maynooth University, W23 F2K8 Kildare, Ireland;
| | - Peter O’Gorman
- Department of Haematology, Mater Misericordiae University Hospital, D07 AX57 Dublin, Ireland; (D.B.); (P.O.)
| |
Collapse
|
2
|
Solimando AG, Da Vià MC, Bolli N, Steinbrunn T. The Route of the Malignant Plasma Cell in Its Survival Niche: Exploring “Multiple Myelomas”. Cancers (Basel) 2022; 14:cancers14133271. [PMID: 35805041 PMCID: PMC9265748 DOI: 10.3390/cancers14133271] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023] Open
Abstract
Growing evidence points to multiple myeloma (MM) and its stromal microenvironment using several mechanisms to subvert effective immune and anti-tumor responses. Recent advances have uncovered the tumor-stromal cell influence in regulating the immune-microenvironment and have envisioned targeting these suppressive pathways to improve therapeutic outcomes. Nevertheless, some subgroups of patients include those with particularly unfavorable prognoses. Biological stratification can be used to categorize patient-, disease- or therapy-related factors, or alternatively, these biological determinants can be included in a dynamic model that customizes a given treatment to a specific patient. Genetic heterogeneity and current knowledge enforce a systematic and comprehensive bench-to-bedside approach. Given the increasing role of cancer stem cells (CSCs) in better characterizing the pathogenesis of solid and hematological malignancies, disease relapse, and drug resistance, identifying and describing CSCs is of paramount importance in the management of MM. Even though the function of CSCs is well-known in other cancer types, their role in MM remains elusive. With this review, we aim to provide an update on MM homing and resilience in the bone marrow micro milieu. These data are particularly interesting for clinicians facing unmet medical needs while designing novel treatment approaches for MM.
Collapse
Affiliation(s)
- Antonio Giovanni Solimando
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine ‘G. Baccelli’, University of Bari Medical School, 70124 Bari, Italy
- Department of Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany
- Correspondence: (A.G.S.); (T.S.); Tel.: +39-3395626475 (A.G.S.)
| | - Matteo Claudio Da Vià
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.C.D.V.); (N.B.)
| | - Niccolò Bolli
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.C.D.V.); (N.B.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Torsten Steinbrunn
- Department of Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
- Correspondence: (A.G.S.); (T.S.); Tel.: +39-3395626475 (A.G.S.)
| |
Collapse
|
3
|
Górska A, Mazur AJ. Integrin-linked kinase (ILK): the known vs. the unknown and perspectives. Cell Mol Life Sci 2022; 79:100. [PMID: 35089438 PMCID: PMC8799556 DOI: 10.1007/s00018-021-04104-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/29/2021] [Accepted: 12/17/2021] [Indexed: 02/08/2023]
Abstract
Integrin-linked kinase (ILK) is a multifunctional molecular actor in cell-matrix interactions, cell adhesion, and anchorage-dependent cell growth. It combines functions of a signal transductor and a scaffold protein through its interaction with integrins, then facilitating further protein recruitment within the ILK-PINCH-Parvin complex. ILK is involved in crucial cellular processes including proliferation, survival, differentiation, migration, invasion, and angiogenesis, which reflects on systemic changes in the kidney, heart, muscle, skin, and vascular system, also during the embryonal development. Dysfunction of ILK underlies the pathogenesis of various diseases, including the pro-oncogenic activity in tumorigenesis. ILK localizes mostly to the cell membrane and remains an important component of focal adhesion. We do know much about ILK but a lot still remains either uncovered or unclear. Although it was initially classified as a serine/threonine-protein kinase, its catalytical activity is now questioned due to structural and functional issues, leaving the exact molecular mechanism of signal transduction by ILK unsolved. While it is known that the three isoforms of ILK vary in length, the presence of crucial domains, and modification sites, most of the research tends to focus on the main isoform of this protein while the issue of functional differences of ILK2 and ILK3 still awaits clarification. The activity of ILK is regulated on the transcriptional, protein, and post-transcriptional levels. The crucial role of phosphorylation and ubiquitylation has been investigated, but the functions of the vast majority of modifications are still unknown. In the light of all those open issues, here we present an extensive literature survey covering a wide spectrum of latest findings as well as a past-to-present view on controversies regarding ILK, finishing with pointing out some open questions to be resolved by further research.
Collapse
Affiliation(s)
- Agata Górska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wrocław, Poland.
| | - Antonina Joanna Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wrocław, Poland.
| |
Collapse
|
4
|
Integrin-Linked Kinase (ILK) Regulates Urinary Stem Cells Differentiation into Smooth Muscle via NF- κB Signal Pathway. Stem Cells Int 2021; 2021:6633111. [PMID: 33854551 PMCID: PMC8019365 DOI: 10.1155/2021/6633111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/28/2021] [Accepted: 03/09/2021] [Indexed: 11/17/2022] Open
Abstract
Objectives Urinary stem cells (USCs) have the capacity for unlimited growth and are promising tools for the investigations of cell differentiation and urinary regeneration. However, the limited life span significantly restricts their usefulness. This study is aimed at exploring the effect of integrin-linked kinase (ILK) on the smooth muscle cells (SMCs) differentiation of the dog USCs and investigating its molecular mechanism. Methods An immortalized USCs cell line with the molecular markers and biological functions was prepared. After successfully inducing the differentiation of USCs into SMCs, the expression level of the unique key factor and its mechanisms in this process was determined through real-time polymerase chain reaction, Western blot, or Immunofluorescence staining. Results We found that high cell density promoted USCs differentiation SMCs, and ILK was necessary for USCs differentiation into SMCs. Knocking down ILK decreased the expression of SMCs specific-marker, while using a selective ILK agonist increased the expression of SMCs specific-marker. Furthermore, ILK regulated SMCs differentiation in part through the activation of NF-κB pathway in USCs. A NF-κB activity assay showed overexpression of ILK could significantly upregulate NF-κB p50 expression, and NF-κB p50 acts as downstream signal molecular of ILK. Conclusion High cell density induces the differentiation of USCs into SMCs, and ILK is a key regulator of myogenesis. Furthermore, NF-κB signaling pathway might play a crucial role in this process.
Collapse
|
5
|
Zhao W, Zhang X, Zang L, Zhao P, Chen Y, Wang X. ILK promotes angiogenic activity of mesenchymal stem cells in multiple myeloma. Oncol Lett 2018; 16:1101-1106. [PMID: 29963187 DOI: 10.3892/ol.2018.8711] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 02/06/2018] [Indexed: 01/19/2023] Open
Abstract
Angiogenic activity in solid tumors has been demonstrated to promote metastasis through the activation of certain proteins involved in the epithelial-mesenchymal transition-associated process. The molecular mechanism underlying multiple myeloma-induced angiogenesis involves angiogenic cytokines by plasma cells as well as their induction within the microenvironment. Integrin-linked kinase (ILK) is a highly evolutionarily conserved intracellular protein that was originally identified as an integrin-interacting protein, and extensive genetic and biochemical studies have identified ILK expression to be vital during tumor-driven angiogenesis. In the present study, it was identified that angiogenic factors were upregulated in mesenchymal stem cells (MSCs) that were co-cultured with multiple myeloma cell lines. It was also revealed that upregulated ILK expression significantly promoted the capillary-formation ability of MSCs. The concentrations of angiogenic factors were significantly decreased compared with non-targeting siRNA-transfected and control MSCs. MSCs may participate in inducing the angiogenic response in multiple myeloma depending on ILK expression.
Collapse
Affiliation(s)
- Weipeng Zhao
- Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China.,Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Xiaoying Zhang
- Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Li Zang
- Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Pan Zhao
- Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Yafang Chen
- Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Xiaofang Wang
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| |
Collapse
|
6
|
de la Puente P, Weisberg E, Muz B, Nonami A, Luderer M, Stone RM, Melo JV, Griffin JD, Azab AK. Identification of ILK as a novel therapeutic target for acute and chronic myeloid leukemia. Leuk Res 2015; 39:S0145-2126(15)30377-5. [PMID: 26413753 PMCID: PMC5016250 DOI: 10.1016/j.leukres.2015.09.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 08/12/2015] [Accepted: 09/06/2015] [Indexed: 11/16/2022]
Abstract
Current treatment options as well as clinical efficacy are limited for chronic myelogenous leukemia (CML), Ph+ acute lymphoblastic leukemia (ALL), and acute myeloid leukemia (AML). In response to the pressing need for more efficacious treatment approaches and strategies to override drug resistance in advanced stage CML, Ph+ ALL, and AML, we investigated the effects of inhibition of ILK as a potentially novel and effective approach to treatment of these challenging malignancies. Using the small molecule ILK inhibitor, Cpd22, and ILK knockdown, we investigated the importance of ILK in the growth and viability of leukemia. Our results suggest that the ILK inhibition may be an effective treatment for CML, Ph+ ALL, and AML as a single therapy, with ILK expression levels positively correlating with the efficacy of ILK inhibition. The identification of ILK as a novel target for leukemia therapy warrants further investigation as a therapeutic approach that could be of potential clinical benefit in both acute and chronic myeloid leukemias.
Collapse
Affiliation(s)
- Pilar de la Puente
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, St. Louis, MO, USA
| | - Ellen Weisberg
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Barbara Muz
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, St. Louis, MO, USA
| | - Atsushi Nonami
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Micah Luderer
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, St. Louis, MO, USA
| | - Richard M Stone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - James D Griffin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
7
|
Kalra J, Dragowska WH, Bally MB. Using Pharmacokinetic Profiles and Digital Quantification of Stained Tissue Microarrays as a Medium-Throughput, Quantitative Method for Measuring the Kinetics of Early Signaling Changes Following Integrin-Linked Kinase Inhibition in an In Vivo Model of Cancer. J Histochem Cytochem 2015; 63:691-709. [PMID: 25940338 PMCID: PMC4804727 DOI: 10.1369/0022155415587978] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 04/27/2015] [Indexed: 12/24/2022] Open
Abstract
A small molecule inhibitor (QLT0267) targeting integrin-linked kinase is able to slow breast tumor growth in vivo; however, the mechanism of action remains unknown. Understanding how targeting molecules involved in intersecting signaling pathways impact disease is challenging. To facilitate this understanding, we used tumor tissue microarrays (TMA) and digital image analysis for quantification of immunohistochemistry (IHC) in order to investigate how QLT0267 affects signaling pathways in an orthotopic model of breast cancer over time. Female NCR nude mice were inoculated with luciferase-positive human breast tumor cells (LCC6Luc) and tumor growth was assessed by bioluminescent imaging (BLI). The plasma levels of QLT0267 were determined by LC-MS/MS methods following oral dosing of QLT0267 (200 mg/kg). A TMA was constructed using tumor tissue collected at 2, 4, 6, 24, 78 and 168 hr after treatment. IHC methods were used to assess changes in ILK-related signaling. The TMA was digitized, and Aperio ScanScope and ImageScope software were used to provide semi-quantitative assessments of staining levels. Using medium-throughput IHC quantitation, we show that ILK targeting by QLT0267 in vivo influences tumor physiology through transient changes in pathways involving AKT, GSK-3 and TWIST accompanied by the translocation of the pro-apoptotic protein BAD and an increase in Caspase-3 activity.
Collapse
Affiliation(s)
- Jessica Kalra
- Experimental Therapeutics BC Cancer Agency, British Columbia, Canada (JK,WHD,MBB),Langara College, Vancouver, British Columbia, Canada (JK)
| | - Weislawa H Dragowska
- Experimental Therapeutics BC Cancer Agency, British Columbia, Canada (JK,WHD,MBB)
| | - Marcel B Bally
- Experimental Therapeutics BC Cancer Agency, British Columbia, Canada (JK,WHD,MBB),Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia (MBB),Department of Biochemistry, University of British Columbia, Vancouver, British Columbia (MBB),Faculty of Pharm. Sciences, University of British Columbia, Vancouver, British Columbia (MBB),Center for Drug Research and Development Vancouver, British Columbia, Canada (MBB)
| |
Collapse
|
8
|
Efficient transient transfection of human multiple myeloma cells by electroporation--an appraisal. PLoS One 2014; 9:e97443. [PMID: 24901949 PMCID: PMC4047019 DOI: 10.1371/journal.pone.0097443] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 04/17/2014] [Indexed: 01/26/2023] Open
Abstract
Cell lines represent the everyday workhorses for in vitro research on multiple myeloma (MM) and are regularly employed in all aspects of molecular and pharmacological investigations. Although loss-of-function studies using RNA interference in MM cell lines depend on successful knockdown, no well-established and widely applied protocol for efficient transient transfection has so far emerged. Here, we provide an appraisal of electroporation as a means to introduce either short-hairpin RNA expression vectors or synthesised siRNAs into MM cells. We found that electroporation using siRNAs was much more efficient than previously anticipated on the basis of transfection efficiencies deduced from EGFP-expression off protein expression vectors. Such knowledge can even confidently be exploited in “hard-to-transfect” MM cell lines to generate large numbers of transient knockdown phenotype MM cells. In addition, special attention was given to developing a protocol that provides easy implementation, good reproducibility and manageable experimental costs.
Collapse
|