1
|
Tachita T, Ri M, Aoki S, Asano A, Kanamori T, Totani H, Kinoshita S, Asao Y, Narita T, Masaki A, Ito A, Kusumoto S, Komatsu H, Iida S. Comprehensive analysis of serum cytokines in patients with multiple myeloma before and after lenalidomide and dexamethasone. Cancer Med 2024; 13:e70019. [PMID: 39031503 PMCID: PMC11259000 DOI: 10.1002/cam4.70019] [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: 01/31/2024] [Revised: 06/19/2024] [Accepted: 07/05/2024] [Indexed: 07/22/2024] Open
Abstract
Multiple myeloma (MM) is an incurable B-cell malignancy often accompanied by profound immunodeficiency. Lenalidomide (Len) is an immunomodulatory drug that exerts promising therapeutic effects on MM through the immune system. However, predictive markers related to the effects of Len treatment are not fully understood. This study aimed to identify candidate biomarkers for predicting the clinical efficacy of Len and dexamethasone (Ld) therapy through a comprehensive analysis of serum cytokines. The levels of 48 cytokines in the serum of patients with MM just before Ld therapy (n = 77), at the time of best response (n = 56), and at disease progression (n = 49) were measured and evaluated. Patients with high IL-18 and M-CSF levels showed significantly shorter progression-free survival and overall survival (OS). In contrast, patients with high PDGF-BB levels had longer survival. Moreover, low levels of G-CSF, IL-7, IL-8, and SDF-1α were associated with shorter OS after Ld therapy. During Ld therapy, pro-inflammatory cytokines such as IL-2Rα, IL-18, and TNF-α were decreased, while IFN-γ was increased. IL-4 and IL-6 levels increased during disease progression. In conclusion, this study provides a better understanding of the association between cytokines and the efficacy of Ld therapy as well as the unique changes in cytokines related to inflammatory and immune responses during Ld therapy.
Collapse
Affiliation(s)
- Takuto Tachita
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
- Department of Gastroenterology and HematologyHirosaki University Graduate School of MedicineHirosakiJapan
| | - Masaki Ri
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Sho Aoki
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Arisa Asano
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Takashi Kanamori
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Haruhito Totani
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Shiori Kinoshita
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Yu Asao
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Tomoko Narita
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Ayako Masaki
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Asahi Ito
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Shigeru Kusumoto
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Hirokazu Komatsu
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Shinsuke Iida
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| |
Collapse
|
2
|
Korbecki J, Bosiacki M, Szatkowska I, Kupnicka P, Chlubek D, Baranowska-Bosiacka I. The Clinical Significance and Involvement in Molecular Cancer Processes of Chemokine CXCL1 in Selected Tumors. Int J Mol Sci 2024; 25:4365. [PMID: 38673949 PMCID: PMC11050300 DOI: 10.3390/ijms25084365] [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: 01/29/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Chemokines play a key role in cancer processes, with CXCL1 being a well-studied example. Due to the lack of a complete summary of CXCL1's role in cancer in the literature, in this study, we examine the significance of CXCL1 in various cancers such as bladder, glioblastoma, hemangioendothelioma, leukemias, Kaposi's sarcoma, lung, osteosarcoma, renal, and skin cancers (malignant melanoma, basal cell carcinoma, and squamous cell carcinoma), along with thyroid cancer. We focus on understanding how CXCL1 is involved in the cancer processes of these specific types of tumors. We look at how CXCL1 affects cancer cells, including their proliferation, migration, EMT, and metastasis. We also explore how CXCL1 influences other cells connected to tumors, like promoting angiogenesis, recruiting neutrophils, and affecting immune cell functions. Additionally, we discuss the clinical aspects by exploring how CXCL1 levels relate to cancer staging, lymph node metastasis, patient outcomes, chemoresistance, and radioresistance.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Góra, Zyty 28, 65-046 Zielona Góra, Poland
| | - Mateusz Bosiacki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
| | - Iwona Szatkowska
- Department of Ruminants Science, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology, Klemensa Janickiego 29 St., 71-270 Szczecin, Poland;
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (M.B.); (D.C.)
| |
Collapse
|
3
|
Mazurek M, Szudy-Szczyrek A, Homa-Mlak I, Hus M, Małecka-Massalska T, Mlak R. IL1B Polymorphism (rs1143634) and IL-1β Plasma Concentration as Predictors of Nutritional Disorders and Prognostic Factors in Multiple Myeloma Patients. Cancers (Basel) 2024; 16:1263. [PMID: 38610941 PMCID: PMC11011170 DOI: 10.3390/cancers16071263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a hematological neoplasm of the early precursor of B-cells. The most characteristic symptoms observed during MM include hypocalcemia, anemia, bacterial infections, and renal damage. Nutritional disorders, especially malnutrition, are noted in about 35-71% of MM patients. Interleukin 1 beta (IL-1β) is a proinflammatory cytokine responsible for muscle atrophy and lipolysis during malnutrition and cachexia. This study aimed to evaluate the usefulness of the IL1B single-nucleotide polymorphism (SNP) (rs1143634) and plasma concentration of IL-1β in the assessment of the risk of nutritional disorders and prognosis in patients with MM. METHODS In our study, 93 patients with the de novo MM were enrolled. The real-time PCR with specific TaqMan probes method was used in genotyping. The IL-1β ELISA kit was used to determine IL-1β concentration in plasma samples. RESULTS Patients with the CC genotype, compared to the carriers of the other variants of the IL1B, demonstrated significantly higher concentrations of IL-1β in plasma (7.56 vs. 4.97 pg/mL), a significantly higher risk of cachexia (OR = 5.11), and a significantly higher risk of death (HR = 2.03). Moreover, high IL-1β plasma level was related to a significantly higher risk of cachexia (OR = 7.76); however, it was not significantly associated with progression-free survival (PFS) or overall survival (OS). CONCLUSIONS Determination of the IL1B SNP (rs1143634) and plasma concentration of IL-1β may be useful in the assessment of the risk of cachexia and prognosis in patients with MM.
Collapse
Affiliation(s)
- Marcin Mazurek
- Department of Human Physiology of Chair of Preclinical Sciences, Medical University of Lublin, 20-080 Lublin, Poland; (I.H.-M.); (T.M.-M.)
| | - Aneta Szudy-Szczyrek
- Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-080 Lublin, Poland; (A.S.-S.); (M.H.)
| | - Iwona Homa-Mlak
- Department of Human Physiology of Chair of Preclinical Sciences, Medical University of Lublin, 20-080 Lublin, Poland; (I.H.-M.); (T.M.-M.)
| | - Marek Hus
- Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-080 Lublin, Poland; (A.S.-S.); (M.H.)
| | - Teresa Małecka-Massalska
- Department of Human Physiology of Chair of Preclinical Sciences, Medical University of Lublin, 20-080 Lublin, Poland; (I.H.-M.); (T.M.-M.)
| | - Radosław Mlak
- Department of Laboratory Diagnostics, Medical University of Lublin, 20-080 Lublin, Poland;
| |
Collapse
|
4
|
Li Z, Wang Y, Hou X, Guo L, Li Y, Ma Y, Ma Y. High expression of HOXC6 predicts a poor prognosis and induces proliferation and inflammation in multiple myeloma cells. Genes Genomics 2023; 45:945-955. [PMID: 37202556 DOI: 10.1007/s13258-023-01397-3] [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: 12/30/2022] [Accepted: 05/07/2023] [Indexed: 05/20/2023]
Abstract
BACKGROUND Multiple myeloma (MM) is a common blood system malignance accompanied by monoclonal plasma cell hyperplasia. Homeobox C6 (HOXC6) acts as an oncogene in various cancers, but its function on MM is elusive. OBJECTIVE The role of HOXC6 on MM development was clarified in this study. METHODS HOXC6 expression and its clinical significance were determined in the peripheral blood samples collected from forty MM patients and thirty healthy adult volunteers. The overall survival was evaluated by Kaplan-Meier analysis with the log-rank test. Cell viability, proliferation and apoptosis were measured by CCK-8, EdU assay and Flow cytometry in U266 and MM.1R cells. Tumor growth was estimated by a xenograft assay. The apoptosis of tumor tissues was evaluated using TUNEL staining. The protein level in tissues was tested by immunohistochemistry. RESULTS The HOXC6 expression was elevated in MM and high HOXC6 level was associated with the poor overall survival of MM. Besides, the HOXC6 expression was associated with hemoglobin level and ISS stage. Furthermore, silencing HOXC6 suppressed cell proliferation, induced cell apoptosis, and restrained the secretion of inflammatory factors (TNF-α, IL-6, and IL-8) in MM cells through inactivating the NF-κB pathway. Moreover, silencing HOXC6 suppressed the tumor growth of MM, the inflammatory factors levels, and the activation of NF-κB pathway but enhanced apoptosis in vivo. CONCLUSION HOXC6 was elevated in MM and associated with poor survival. Knockdown of HOXC6 suppressed proliferation, inflammation and tumorigenicity of MM cells via inactivating the NF-κB pathway. HOXC6 may be a meaningful target for MM therapy.
Collapse
Affiliation(s)
- Zhihua Li
- Department of hematology, Second Hospital of Shanxi Medical University, 0300001, NO382,Wuyi Road, Taiyuan, shanxi, province, Taiyuan, 0300001, shanxi, province, China
| | - Yaru Wang
- Department of hematology, Second Hospital of Shanxi Medical University, 0300001, NO382,Wuyi Road, Taiyuan, shanxi, province, Taiyuan, 0300001, shanxi, province, China
| | - Xiaoxu Hou
- Department of hematology, Second Hospital of Shanxi Medical University, 0300001, NO382,Wuyi Road, Taiyuan, shanxi, province, Taiyuan, 0300001, shanxi, province, China
| | - Luyao Guo
- Department of hematology, Second Hospital of Shanxi Medical University, 0300001, NO382,Wuyi Road, Taiyuan, shanxi, province, Taiyuan, 0300001, shanxi, province, China
| | - Yanling Li
- Department of hematology, Second Hospital of Shanxi Medical University, 0300001, NO382,Wuyi Road, Taiyuan, shanxi, province, Taiyuan, 0300001, shanxi, province, China
| | - Yanping Ma
- Department of hematology, Second Hospital of Shanxi Medical University, 0300001, NO382,Wuyi Road, Taiyuan, shanxi, province, Taiyuan, 0300001, shanxi, province, China
| | - Yanping Ma
- Department of hematology, Second Hospital of Shanxi Medical University, 0300001, NO382,Wuyi Road, Taiyuan, shanxi, province, Taiyuan, 0300001, shanxi, province, China.
| |
Collapse
|
5
|
Circulating cytokines present in multiple myeloma patients inhibit the osteoblastic differentiation of adipose stem cells. Leukemia 2022; 36:540-548. [PMID: 34556797 DOI: 10.1038/s41375-021-01428-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 09/02/2021] [Accepted: 09/14/2021] [Indexed: 02/08/2023]
Abstract
Myeloma is characterized by bone lesions, which are related to both an increased osteoclast activity and a defect in the differentiation of medullary mesenchymal stem cells (MSCs) into osteoblasts. Outside the medullary environment, adipocyte-derived MSCs (ASCs) could represent a source of functional osteoblasts. However, we recently found a defect in the osteoblastic differentiation of ASCs from myeloma patients (MM-ASCs). We examined the effects of plasma from myeloma patients at diagnosis (MM-plasmas) and in complete remission (CR-plasmas) and from healthy donors on the osteoblastic differentiation of healthy donor-derived ASCs (HD-ASCs). Osteoblastogenesis in HD-ASCs was suppressed by MM-plasmas. Seven cytokines (ANG1, ENA-78, EGF, PDGF-AA/AB/BB, and TARC) were increased in MM-plasmas and separately inhibited the osteoblastic differentiation of HD-ASCs. Comparison of MM-ASCs and HD-ASCs by RNA sequencing showed that two master genes characterizing adipocyte differentiation, CD36 and PPARγ, were upregulated in MM-ASCs as compared to HD-ASCs. Finally, we demonstrated a significant increase in CD36 and PPARγ expression in HD-ASCs in the presence of MM-plasmas or the seven cytokines individually, similarly as in MM-ASCs. We conclude that specific cytokines in MM-plasmas, besides the well-known DKK1, inhibit the osteoblastic differentiation of MM- and HD-ASCs with a skewing towards adipocyte differentiation.
Collapse
|
6
|
Visram A, Kourelis TV. Aging-associated immune system changes in multiple myeloma: The dark side of the moon. Cancer Treat Res Commun 2021; 29:100494. [PMID: 34837796 DOI: 10.1016/j.ctarc.2021.100494] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 11/18/2021] [Indexed: 02/08/2023]
Abstract
Multiple myeloma (MM) is a disease of the elderly. Changes that occur in the immune system with aging, also known as immunosenescence, have been associated with decreased tumor immunosurveillance and are thought to contribute to the development of MM and other cancers in the elderly. Once MM establishes itself in the bone marrow, immunosenescence related changes have been observed in the immune tumor microenvironment (iTME) and are driven by the malignant cells. The efficacy of novel immunotherapies used to treat MM has been blunted by detrimental iTME changes that occur at later disease stages and are, to some extent, driven by prior therapies. In this review, we discuss general changes that occur in the immune system with aging as well as our current knowledge of immunosenescence in MM. We discuss the differences and overlap between T cell senescence and exhaustion as well as potential methods to prevent or reverse immunosenescence. We focus predominantly on T cell immunosenescence which has been better evaluated in this disease and is more pertinent to novel MM immunotherapies. Our lack of understanding of the drivers of immunosenescence at each stage of the disease, from precursor stages to heavily pretreated MM, represents a major barrier to improving the efficacy of novel and existing therapies.
Collapse
Affiliation(s)
- Alissa Visram
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN United States; Department of Medicine, Division of Hematology, University of Ottawa, Ottawa Hospital Research Institute, Ontario, Canada
| | - Taxiarchis V Kourelis
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN United States.
| |
Collapse
|
7
|
Takemori N, Ooi HK, Imai G, Hoshino K, Saio M. Possible mechanisms of action of clarithromycin and its clinical application as a repurposing drug for treating multiple myeloma. Ecancermedicalscience 2020; 14:1088. [PMID: 33014130 PMCID: PMC7498274 DOI: 10.3332/ecancer.2020.1088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Indexed: 12/15/2022] Open
Abstract
Clarithromycin (CAM), a semisynthetic macrolide antibiotic, is a widely used antibacterial drug. Recently, the efficacy of CAM as an add-on drug for treating multiple myeloma (MM) has been noted. Its effect on treating MM has been confirmed in combination chemotherapies that include CAM. However, a single treatment of CAM has no efficacy for treating MM. Many myeloma growth factors (MGFs) including interleukin (IL)-6 are known to be closely involved in the development of MM. CAM has been shown to suppress many MGFs, particularly IL-6. The possible mechanisms of action of CAM in treating MM have been suggested to include its immunomodulatory effect, autophagy inhibition, reversibility of drug resistance, steroid-sparing/enhancing effect and suppression of MGFs. In addition, MM is characterised by uncontrolled cell growth of monoclonal immunoglobulin (Ig)-producing neoplastic plasma cells. Large quantities of unfolded or misfolded Ig production may trigger considerable endoplasmic reticulum stress. Thus, MM is originally a fragile neoplasm particularly susceptible to autophagy-, proteasome- and histone deacetylase 6-inhibitors. Taken together, CAM plays an important role in MM treatments through its synergistic mechanisms. In addition, CAM with its pleiotropic effects on cytokines including IL-6 and indirect antiviral effects might be worth a try for treating COVID-19.
Collapse
Affiliation(s)
- Nobuo Takemori
- Department of Internal Medicine, Division of Hematology, Imai Hospital, Tanaka-cho 100, Ashikaga, Tochigi 326-0822, Japan
- https://orcid.org/0000-0001-9742-8385
| | - Hong-Kean Ooi
- Department of Veterinary Medicine, Azabu University, Fuchinobe 1-17-71, Sagamihara, Kanagawa 252-5201, Japan
| | - Goro Imai
- Department of Internal Medicine, Imai Hospital, Tanaka-cho 100, Ashikaga, Tochigi 326-0822, Japan
| | - Kazuo Hoshino
- Department of Surgery, Imai Hospital, Tanaka-cho 100, Ashikaga, Tochigi 326-0822, Japan
| | - Masanao Saio
- Laboratory of Histopathology & Cytopathology, Department of Laboratory Sciences, Gunma University, Graduate School of Health Sciences, 39-22, 3-chome, Showa-machi, Maebashi, Gunma 371-8514, Japan
| |
Collapse
|
8
|
Liu X, Chen Z, Lan T, Liang P, Tao Q. Upregulation of interleukin-8 and activin A induces osteoclastogenesis in ameloblastoma. Int J Mol Med 2019; 43:2329-2340. [PMID: 31017256 PMCID: PMC6488175 DOI: 10.3892/ijmm.2019.4171] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 04/10/2019] [Indexed: 02/07/2023] Open
Abstract
Ameloblastoma is a common odontogenic benign tumor located in the jaws and is characterized by severe local bone destruction. The current study aimed to investigate the effect of interactions between tumor cells and bone marrow stromal cells (BMSCs) on osteoclast formation in ameloblastoma. The impact of ameloblastoma/BMSC interactions on cytokine production, gene expression and osteoclastogenesis was examined using an immortalized ameloblastoma cell line that the authors' previously established. The results demonstrated that interactions between ameloblastoma cells and BMSCs increased interleukin (IL)‑8 and activin A secretion by BMSCs. IL‑8 expression in BMSCs was modulated by tumor‑derived tumor necrosis factor‑α and IL‑8 contributed to osteoclast formation not only directly but also by stimulating receptor activator of NF‑κB ligand (RANKL) expression in BMSCs. Activin A secretion in BMSCs was stimulated by ameloblastoma cells via cell‑to‑cell‑mediated activation of c‑Jun N‑terminal kinase activation, acting as a cofactor of RANKL to induce osteoclast formation and function. The present study highlights the critical role of communication between BMSCs and ameloblastoma cells in bone resorption in ameloblastoma.
Collapse
Affiliation(s)
- Xin Liu
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Zhifeng Chen
- Department of Oral and Maxillofacial Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Tianjun Lan
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Peisheng Liang
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Qian Tao
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| |
Collapse
|
9
|
Piddock RE, Marlein CR, Abdul-Aziz A, Shafat MS, Auger MJ, Bowles KM, Rushworth SA. Myeloma-derived macrophage inhibitory factor regulates bone marrow stromal cell-derived IL-6 via c-MYC. J Hematol Oncol 2018; 11:66. [PMID: 29769142 PMCID: PMC5956761 DOI: 10.1186/s13045-018-0614-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/06/2018] [Indexed: 01/19/2023] Open
Abstract
Abstract Multiple myeloma (MM) remains an incurable malignancy despite the recent advancements in its treatment. The protective effects of the niche in which it develops has been well documented; however, little has been done to investigate the MM cell’s ability to ‘re-program’ cells within its environment to benefit disease progression. Here, we show that MM-derived macrophage migratory inhibitory factor (MIF) stimulates bone marrow stromal cells to produce the disease critical cytokines IL-6 and IL-8, prior to any cell-cell contact. Furthermore, we provide evidence that this IL-6/8 production is mediated by the transcription factor cMYC. Pharmacological inhibition of cMYC in vivo using JQ1 led to significantly decreased levels of serum IL-6—a highly positive prognostic marker in MM patients. Conclusions Our presented findings show that MM-derived MIF causes BMSC secretion of IL-6 and IL-8 via BMSC cMYC. Furthermore, we show that the cMYC inhibitor JQ1 can reduce BMSC secreted IL-6 in vivo, irrespective of tumor burden. These data provide evidence for the clinical evaluation of both MIF and cMYC inhibitors in the treatment of MM. Electronic supplementary material The online version of this article (10.1186/s13045-018-0614-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rachel E Piddock
- Department of Molecular Haematology, Norwich Medical School, The University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Christopher R Marlein
- Department of Molecular Haematology, Norwich Medical School, The University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Amina Abdul-Aziz
- Department of Molecular Haematology, Norwich Medical School, The University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Manar S Shafat
- Department of Molecular Haematology, Norwich Medical School, The University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Martin J Auger
- Department of Haematology, Norfolk and Norwich University Hospitals NHS Trust, Colney Lane, Norwich, NR4 7UY, UK
| | - Kristian M Bowles
- Department of Molecular Haematology, Norwich Medical School, The University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK. .,Department of Haematology, Norfolk and Norwich University Hospitals NHS Trust, Colney Lane, Norwich, NR4 7UY, UK.
| | - Stuart A Rushworth
- Department of Molecular Haematology, Norwich Medical School, The University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| |
Collapse
|
10
|
Cosemans C, Oben B, Arijs I, Daniëls A, Declercq J, Vanhees K, Froyen G, Maes B, Mebis J, Rummens JL. Prognostic Biomarkers in the Progression From MGUS to Multiple Myeloma: A Systematic Review. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 18:235-248. [PMID: 29506935 DOI: 10.1016/j.clml.2018.02.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 01/24/2018] [Accepted: 02/13/2018] [Indexed: 12/17/2022]
Abstract
Multiple myeloma (MM), characterized by malignant plasma cells in the bone marrow, is consistently preceded by asymptomatic premalignant stage monoclonal gammopathy of undetermined significance (MGUS). These MGUS patients have an annual risk of 1% to progress to MM. Clinical, imaging, and genomic (genetic and epigenetic) factors were identified, whose presence increased the risk of progression from MGUS to MM. In this systematic review we summarize the currently identified clinical, imaging, and genomic biomarkers suggested to increase the progression risk or shown to be differentially expressed/present between both cohorts of patients. Despite the wide range of proposed markers, there are still no reliable biomarkers to individually predict which MGUS patient will progress to MM and which will not. Research on biomarkers in the progression from MGUS to MM will give more insight in the unknown pathogenesis of this hematological malignancy. This would improve research by elucidating new pathways and potential therapeutic targets as well as clinical management by closer follow-up and earlier treatment of high-risk MGUS patients.
Collapse
Affiliation(s)
- Charlotte Cosemans
- Department of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Bénedith Oben
- Department of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium.
| | - Ingrid Arijs
- Department of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Annick Daniëls
- Department of Experimental Hematology, Jessa Hospital, Hasselt, Belgium
| | - Jeroen Declercq
- Department of Experimental Hematology, Jessa Hospital, Hasselt, Belgium
| | - Kimberly Vanhees
- Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; University Biobank Limburg (UBiLim) and Biobank Jessa, Hasselt, Belgium
| | - Guy Froyen
- Department of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Department of Clinical Biology, Jessa Hospital, Hasselt, Belgium
| | - Brigitte Maes
- Department of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Department of Clinical Biology, Jessa Hospital, Hasselt, Belgium
| | - Jeroen Mebis
- Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Division of Medical Oncology, Jessa Hospital, Hasselt, Belgium
| | - Jean-Luc Rummens
- Department of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; University Biobank Limburg (UBiLim) and Biobank Jessa, Hasselt, Belgium; Department of Clinical Biology, Jessa Hospital, Hasselt, Belgium
| |
Collapse
|
11
|
Guo Y, Zang Y, Lv L, Cai F, Qian T, Zhang G, Feng Q. IL‑8 promotes proliferation and inhibition of apoptosis via STAT3/AKT/NF‑κB pathway in prostate cancer. Mol Med Rep 2017; 16:9035-9042. [PMID: 29039490 DOI: 10.3892/mmr.2017.7747] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 09/12/2017] [Indexed: 11/06/2022] Open
Abstract
Interleukin-8 (IL-8) possesses tumorigenic and proangiogenic properties, and is overexpressed in many human cancer types. However, only few studies have demonstrated the mechanisms of action of IL‑8 regarding the ability to promote proliferation and to inhibit apoptosis in prostate cancer. Here, the aim of the present study was to investigate the effects of IL‑8 on the prostate cancer cell line and determine possible mechanisms underlying its effect. In this study, IL‑8 was shown to be significantly upregulated in prostate cancer compared with paired normal control tissues. The data showed that IL‑8 exhibits direct oncogenicity, which significantly induced cell proliferation, invasion and attenuated apoptosis in prostate cancer cells via signal transducer and activator of transcription 3/protein kinase B/nuclear factor‑κB signaling pathways. In conclusion, modulation of IL‑8 expression or its associated signaling pathway may provide a novel working mechanism of IL‑8 in prostate cancer, and a promising strategy for controlling the progression and metastasis of prostate cancer.
Collapse
Affiliation(s)
- Yidi Guo
- Department of Clinical Laboratory, Nanjing Integrated Traditional Chinese and Western Medicine Hospital Affiliated with Nanjing University of Chinese Medicine, Nanjing 210000, P.R. China
| | - Ying Zang
- Department of Clinical Laboratory, Nanjing Integrated Traditional Chinese and Western Medicine Hospital Affiliated with Nanjing University of Chinese Medicine, Nanjing 210000, P.R. China
| | - Lianzheng Lv
- Department of Clinical Laboratory, Nanjing Integrated Traditional Chinese and Western Medicine Hospital Affiliated with Nanjing University of Chinese Medicine, Nanjing 210000, P.R. China
| | - Feng Cai
- Department of Clinical Laboratory, Nanjing Integrated Traditional Chinese and Western Medicine Hospital Affiliated with Nanjing University of Chinese Medicine, Nanjing 210000, P.R. China
| | - Tingting Qian
- Department of Clinical Laboratory, Nanjing Integrated Traditional Chinese and Western Medicine Hospital Affiliated with Nanjing University of Chinese Medicine, Nanjing 210000, P.R. China
| | - Guoying Zhang
- Department of Clinical Laboratory, Nanjing Integrated Traditional Chinese and Western Medicine Hospital Affiliated with Nanjing University of Chinese Medicine, Nanjing 210000, P.R. China
| | - Quancheng Feng
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing 210000, P.R. China
| |
Collapse
|
12
|
Liu C, Gu X, Jiang Z. Identification of novel targets for multiple myeloma through integrative approach with Monte Carlo cross-validation analysis. J Bone Oncol 2017; 8:8-12. [PMID: 28856086 PMCID: PMC5565744 DOI: 10.1016/j.jbo.2017.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 08/01/2017] [Accepted: 08/10/2017] [Indexed: 11/20/2022] Open
Abstract
More than one pathway is involved in disease development and progression, and two or more pathways may be interconnected to further affect the disease onset, as functional proteins participate in multiple pathways. Thus, identifying cross-talk among pathways is necessary to understand the molecular mechanisms of multiple myeloma (MM). Based on this, this paper looked at extracting potential pathway cross-talk in MM through an integrative approach using Monte Carlo cross-validation analysis. The gene expression library of MM (accession number: GSE6477) was downloaded from the Gene Expression Omnibus (GEO) database. The integrative approach was then used to identify potential pathway cross-talk, and included four steps: Firstly, differential expression analysis was conducted to identify differentially expressed genes (DEGs). Secondly, the DEGs obtained were mapped to the pathways downloaded from an ingenuity pathways analysis (IPA), to reveal the underlying relationship between the DEGs and pathways enriched by these DEGs. A subset of pathways enriched by the DEGs was then obtained. Thirdly, a discriminating score (DS) value for each paired pathway was computed. Lastly, random forest (RF) classification was used to identify the paired pathways based on area under the curve (AUC) and Monte Carlo cross-validation, which was repeated 50 times to explore the best paired pathways. These paired pathways were tested with another independently published MM microarray data (GSE85837), using in silico validation. Overall, 60 DEGs and 19 differential pathways enriched by DEGs were extracted. Each pathway was sorted based on their AUC values. The paired pathways, inhibition of matrix metalloproteases and EIF2 signaling pathway, indicated the best AUC value of 1.000. Paired pathways consisting of IL-8 and EIF2 signaling pathways with higher AUC of 0.975, were involved in 7 runs. Furthermore, it was validated consistently in separate microarray data sets (GSE85837). Paired pathways (inhibition of matrix metalloproteases and EIF2 signaling, IL-8 signaling and EIF2 signaling) exhibited the best AUC values and higher frequency of validation. Two paired pathways (inhibition of matrix metalloproteases and EIF2 signaling, IL-8 signaling and EIF2 signaling) were used to accurately classify MM and control samples. These paired pathways may be potential bio-signatures for diagnosis and management of MM.
Collapse
|
13
|
Immune signatures associated with improved progression-free and overall survival for myeloma patients treated with AHSCT. Blood Adv 2017; 1:1056-1066. [PMID: 29296748 DOI: 10.1182/bloodadvances.2017005447] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/12/2017] [Indexed: 11/20/2022] Open
Abstract
Multiple therapeutic options exist for multiple myeloma (MM), including autologous hematopoietic stem cell transplantation (AHSCT). Measurement of minimal residual disease (MRD) and immune reconstitution is rapidly becoming an integral part of the care of MM patients. We investigated comprehensive immune profiling (IP) associated with progression-free survival (PFS) and overall survival (OS). From August 2007 to January 2014, 101 consecutive MM patients underwent peripheral blood IP and marrow MRD testing before and approximately 100 days after AHSCT. Higher pre-AHSCT CD19+ B-cell counts correlated with improved 2-year PFS (83% [highest quartile] vs 53% [lowest quartile]; P = .01) and OS (93% [highest quartile] vs 63% [lowest quartile]; P = .0003). This effect was seen primarily in patients with MRD-positive marrow tests. Higher γδ T-cell counts post-AHSCT correlated with improved 2-year PFS (65% [highest quartile] vs 45% [lowest quartile]; P = .02) and OS (89% [highest quartile] vs 65% [lowest quartile]; P = .01). Higher CD4+ central memory (CM) cell counts post-AHSCT were associated with improved 2-year OS (95% [upper quartile] vs 47% [lowest quartile]; P = .0003) but not PFS. The higher γδ T-cell and CD4+ CM-cell count associations were primarily observed in MRD-negative patients post-AHSCT and in patients not receiving maintenance therapy. This proof-of-concept study demonstrates that IP before and after AHSCT can be of complementary prognostic value for depth of response. Maintenance therapy seems to overcome negative IP. IP and MRD should be measured in clinical trials of maintenance therapy with novel agents post-AHSCT for MM to confirm their utility for prognosis and management.
Collapse
|
14
|
Al-Hujaily EM, Oldham RAA, Hari P, Medin JA. Development of Novel Immunotherapies for Multiple Myeloma. Int J Mol Sci 2016; 17:E1506. [PMID: 27618026 PMCID: PMC5037783 DOI: 10.3390/ijms17091506] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/24/2016] [Accepted: 09/01/2016] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is a disorder of terminally differentiated plasma cells characterized by clonal expansion in the bone marrow (BM). It is the second-most common hematologic malignancy. Despite significant advances in therapeutic strategies, MM remains a predominantly incurable disease emphasizing the need for the development of new treatment regimens. Immunotherapy is a promising treatment modality to circumvent challenges in the management of MM. Many novel immunotherapy strategies, such as adoptive cell therapy and monoclonal antibodies, are currently under investigation in clinical trials, with some already demonstrating a positive impact on patient survival. In this review, we will summarize the current standards of care and discuss major new approaches in immunotherapy for MM.
Collapse
Affiliation(s)
- Ensaf M Al-Hujaily
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Robyn A A Oldham
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
| | - Parameswaran Hari
- Department of Medicine, Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Jeffrey A Medin
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
- The Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada.
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
15
|
Effects of IL-8 Up-Regulation on Cell Survival and Osteoclastogenesis in Multiple Myeloma. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2171-2182. [PMID: 27301357 DOI: 10.1016/j.ajpath.2016.04.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 03/14/2016] [Accepted: 04/11/2016] [Indexed: 02/07/2023]
Abstract
IL-8 promotes cancer cell growth, survival, angiogenesis, and metastasis in several tumors. Herein, we investigated the sources of IL-8 production in multiple myeloma (MM) and its potential roles in MM pathogenesis. We found that bone marrow cells from patients with MM secreted higher amounts of IL-8 than healthy donors. IL-8 production was detected in cultures of CD138(+) plasma cells and CD138(-) cells isolated from bone marrows of MM patients, and in three of seven human myeloma cell lines (HMCLs) analyzed. Interactions between MM and stromal cells increased IL-8 secretion by stromal cells through cell-cell adhesion and soluble factors. Interestingly, IL8 expression also increased in HMCLs, stromal cells, and osteoclasts after treatment with the antimyeloma drugs melphalan and bortezomib. In fact, the effect of bortezomib on IL-8 production was higher than that exerted by stromal-MM cell interactions. Addition of exogenous IL-8 did not affect growth of HMCLs, although it protected cells from death induced by serum starvation through a caspase-independent mechanism. Furthermore, IL-8 induced by stromal-MM cell interactions strongly contributed to osteoclast formation in vitro, because osteoclastogenesis was markedly reduced by IL-8-specific neutralizing antibodies. In conclusion, our results implicate IL-8 in myeloma bone disease and point to the potential utility of an anti-IL-8 therapy to prevent unwanted effects of IL-8 up-regulation on survival, angiogenesis, and osteolysis in MM.
Collapse
|
16
|
Garcia-Gomez A, De Las Rivas J, Ocio EM, Díaz-Rodríguez E, Montero JC, Martín M, Blanco JF, Sanchez-Guijo FM, Pandiella A, San Miguel JF, Garayoa M. Transcriptomic profile induced in bone marrow mesenchymal stromal cells after interaction with multiple myeloma cells: implications in myeloma progression and myeloma bone disease. Oncotarget 2015; 5:8284-305. [PMID: 25268740 PMCID: PMC4226683 DOI: 10.18632/oncotarget.2058] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Despite evidence about the implication of the bone marrow (BM) stromal microenvironment in multiple myeloma (MM) cell growth and survival, little is known about the effects of myelomatous cells on BM stromal cells. Mesenchymal stromal cells (MSCs) from healthy donors (dMSCs) or myeloma patients (pMSCs) were co-cultured with the myeloma cell line MM.1S, and the transcriptomic profile of MSCs induced by this interaction was analyzed. Deregulated genes after co-culture common to both d/pMSCs revealed functional involvement in tumor microenvironment cross-talk, myeloma growth induction and drug resistance, angiogenesis and signals for osteoclast activation and osteoblast inhibition. Additional genes induced by co-culture were exclusively deregulated in pMSCs and predominantly associated to RNA processing, the ubiquitine-proteasome pathway, cell cycle regulation, cellular stress and non-canonical Wnt signaling. The upregulated expression of five genes after co-culture (CXCL1, CXCL5 and CXCL6 in d/pMSCs, and Neuregulin 3 and Norrie disease protein exclusively in pMSCs) was confirmed, and functional in vitro assays revealed putative roles in MM pathophysiology. The transcriptomic profile of pMSCs co-cultured with myeloma cells may better reflect that of MSCs in the BM of myeloma patients, and provides new molecular insights to the contribution of these cells to MM pathophysiology and to myeloma bone disease.
Collapse
Affiliation(s)
- Antonio Garcia-Gomez
- Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain. Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain. Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
| | - Javier De Las Rivas
- Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain
| | - Enrique M Ocio
- Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain. Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain
| | - Elena Díaz-Rodríguez
- Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain
| | - Juan C Montero
- Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain
| | - Montserrat Martín
- Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain. Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
| | - Juan F Blanco
- Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain
| | - Fermín M Sanchez-Guijo
- Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain. Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
| | - Atanasio Pandiella
- Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain. Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain
| | - Jesús F San Miguel
- Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain. Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain. Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
| | - Mercedes Garayoa
- Centro de Investigación del Cáncer, IBMCC (Universidad de Salamanca-CSIC), Salamanca, Spain. Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain. Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
| |
Collapse
|
17
|
Live imaging and gene expression analysis in zebrafish identifies a link between neutrophils and epithelial to mesenchymal transition. PLoS One 2014; 9:e112183. [PMID: 25372289 PMCID: PMC4221567 DOI: 10.1371/journal.pone.0112183] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 10/06/2014] [Indexed: 12/18/2022] Open
Abstract
Chronic inflammation is associated with epithelial to mesenchymal transition (EMT) and cancer progression however the relationship between inflammation and EMT remains unclear. Here, we have exploited zebrafish to visualize and quantify the earliest events during epithelial cell transformation induced by oncogenic HRasV12. Live imaging revealed that expression of HRasV12 in the epidermis results in EMT and chronic neutrophil and macrophage infiltration. We have developed an in vivo system to probe and quantify gene expression changes specifically in transformed cells from chimeric zebrafish expressing oncogenic HRasV12 using translating ribosomal affinity purification (TRAP). We found that the expression of genes associated with EMT, including slug, vimentin and mmp9, are enriched in HRasV12 transformed epithelial cells and that this enrichment requires the presence of neutrophils. An early signal induced by HRasV12 in epithelial cells is the expression of il-8 (cxcl8) and we found that the chemokine receptor, Cxcr2, mediates neutrophil but not macrophage recruitment to the transformed cells. Surprisingly, we also found a cell autonomous role for Cxcr2 signaling in transformed cells for both neutrophil recruitment and EMT related gene expression associated with Ras transformation. Taken together, these findings implicate both autocrine and paracrine signaling through Cxcr2 in the regulation of inflammation and gene expression in transformed epithelial cells.
Collapse
|
18
|
Zingone A, Wang W, Corrigan-Cummins M, Wu SP, Plyler R, Korde N, Kwok M, Manasanch EE, Tageja N, Bhutani M, Mulquin M, Zuchlinski D, Yancey MA, Roschewski M, Zhang Y, Roccaro AM, Ghobrial IM, Calvo KR, Landgren O. Altered cytokine and chemokine profiles in multiple myeloma and its precursor disease. Cytokine 2014; 69:294-7. [PMID: 25043675 PMCID: PMC4148078 DOI: 10.1016/j.cyto.2014.05.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 04/20/2014] [Accepted: 05/16/2014] [Indexed: 02/02/2023]
Abstract
Currently, no reliable biomarkers are available to predict transformation from smoldering myeloma (SMM) to multiple myeloma (MM). Using an ultrasensitive enzyme-linked immunosorbent assay (ELISA) we assessed the levels of a broad range of cytokines and chemokines in the peripheral blood (PB) and bone marrow (BM) supernatant collected from 14 SMM and 38 MM patients and compared to healthy donors. We found significantly increased levels of key cytokines, in particular CXCL8 (IL-8), associated with progressive disease state (controls→SMM→MM). Cytokine profiles were found similar in PB and BM. Five of fourteen SMM patients (36%) progressed to MM. Our findings, although based on a limited number of patients, suggest that serum-based cytokines may have a future role as biomarkers for disease progression and could potentially be assessed as novel targets for treatment.
Collapse
Affiliation(s)
- Adriana Zingone
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Weixin Wang
- Hematology Service, Department of Laboratory Medicine, Clinical Center, NIH, Bethesda, MD, USA
| | - Meghan Corrigan-Cummins
- Hematology Service, Department of Laboratory Medicine, Clinical Center, NIH, Bethesda, MD, USA
| | - S Peter Wu
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ryan Plyler
- Hematology Service, Department of Laboratory Medicine, Clinical Center, NIH, Bethesda, MD, USA
| | - Neha Korde
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mary Kwok
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elisabet E Manasanch
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nishant Tageja
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Manisha Bhutani
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marcia Mulquin
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Diamond Zuchlinski
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mary Ann Yancey
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mark Roschewski
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yong Zhang
- Dana-Faber Cancer Institute, Department of Medical Oncology, Harvard Medical School, Boston, MA, USA
| | - Aldo M Roccaro
- Dana-Faber Cancer Institute, Department of Medical Oncology, Harvard Medical School, Boston, MA, USA
| | - Irene M Ghobrial
- Dana-Faber Cancer Institute, Department of Medical Oncology, Harvard Medical School, Boston, MA, USA
| | - Katherine R Calvo
- Hematology Service, Department of Laboratory Medicine, Clinical Center, NIH, Bethesda, MD, USA
| | - Ola Landgren
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Myeloma Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
19
|
Zhou J, Xiang Y, Yoshimura T, Chen K, Gong W, Huang J, Zhou Y, Yao X, Bian X, Wang JM. The role of chemoattractant receptors in shaping the tumor microenvironment. BIOMED RESEARCH INTERNATIONAL 2014; 2014:751392. [PMID: 25110692 PMCID: PMC4119707 DOI: 10.1155/2014/751392] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/17/2014] [Indexed: 12/13/2022]
Abstract
Chemoattractant receptors are a family of seven transmembrane G protein coupled receptors (GPCRs) initially found to mediate the chemotaxis and activation of immune cells. During the past decades, the functions of these GPCRs have been discovered to not only regulate leukocyte trafficking and promote immune responses, but also play important roles in homeostasis, development, angiogenesis, and tumor progression. Accumulating evidence indicates that chemoattractant GPCRs and their ligands promote the progression of malignant tumors based on their capacity to orchestrate the infiltration of the tumor microenvironment by immune cells, endothelial cells, fibroblasts, and mesenchymal cells. This facilitates the interaction of tumor cells with host cells, tumor cells with tumor cells, and host cells with host cells to provide a basis for the expansion of established tumors and development of distant metastasis. In addition, many malignant tumors of the nonhematopoietic origin express multiple chemoattractant GPCRs that increase the invasiveness and metastasis of tumor cells. Therefore, GPCRs and their ligands constitute targets for the development of novel antitumor therapeutics.
Collapse
Affiliation(s)
- Jiamin Zhou
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
- Endoscopic Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yi Xiang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
- Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Teizo Yoshimura
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Keqiang Chen
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Jian Huang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ye Zhou
- Department of Gastric Cancer and Soft Tissue Surgery, Fudan University Cancer Center, Shanghai 200032, China
| | - Xiaohong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ji Ming Wang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
20
|
Pappa CA, Tsirakis G, Devetzoglou M, Zafeiri M, Vyzoukaki R, Androvitsanea A, Xekalou A, Sfiridaki K, Alexandrakis MG. Bone marrow mast cell density correlates with serum levels of VEGF and CXC chemokines ENA-78 and GRO-α in multiple myeloma. Tumour Biol 2014; 35:5647-51. [PMID: 24563338 DOI: 10.1007/s13277-014-1747-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 02/13/2014] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is a crucial process in growth and progression of multiple myeloma (MM). Mast cells (MCs) play an important role in MM angiogenesis. Various angiogenic mediators secreted by MCs regulate endothelial cell proliferation and function. Among them, ELR(+) CXC chemokines, such as growth-related oncogen-alpha (GRO-α) and epithelial neutrophil activating protein-78 (ENA-78), have been described as potential mediators in regulation of angiogenesis. The purpose of the study was to quantify MCs in bone marrow (BM) biopsies of MM patients, expressed as MC density (MCD), and correlate it with serum concentrations of vascular endothelial factor (VEGF), GRO-α, ENA-78. Fifty-four newly diagnosed MM patients and 22 healthy controls were studied. Tryptase was used for the immunohistochemical stain of MCs. VEGF, GRO-α, and ENA-78 were measured in sera by ELISA. MCD and serum levels of GRO-α, ENA-78, and VEGF were significantly higher in MM patients compared to controls (p<0.001 in all cases). MCD was significantly increasing with increased stage of the disease (p<0.001). Furthermore, significant correlations were found between MCD with VEGF, GRO-α, and ENA-78. These findings support that MCs participate in the pathophysiology of MM and is implicated in the angiogenic process and disease progression.
Collapse
Affiliation(s)
- C A Pappa
- Hematology Department, Venizelion General Hospital of Heraklion, Heraklion, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Slany A, Haudek-Prinz V, Meshcheryakova A, Bileck A, Lamm W, Zielinski C, Gerner C, Drach J. Extracellular matrix remodeling by bone marrow fibroblast-like cells correlates with disease progression in multiple myeloma. J Proteome Res 2013; 13:844-54. [PMID: 24256566 DOI: 10.1021/pr400881p] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The pathogenesis of multiple myeloma (MM) is regarded as a multistep process, in which an asymptomatic stage of monoclonal gammopathy of undetermined significance (MGUS) precedes virtually all cases of MM. Molecular events characteristic for the transition from MGUS to MM are still poorly defined. We hypothesized that fibroblast-like cells in the tumor microenvironment are critically involved in the pathogenesis of MM. Therefore, we performed a comparative proteome profiling study, analyzing primary human fibroblast-like cells isolated from the bone marrow of MM, of MGUS, as well as of non-neoplastic control patients. Thereby, a group of extracellular matrix (ECM) proteins, ECM receptors, and ECM-modulating enzymes turned out to be progressively up-regulated in MGUS and MM. These proteins include laminin α4, lysyl-hydroxylase 2, prolyl 4-hydroxylase 1, nidogen-2, integrin α5β5, c-type mannose receptor 2, PAI-1, basigin, and MMP-2, in addition to PDGF-receptor β and the growth factor periostin, which are likewise involved in ECM activities. Our results indicate that ECM remodeling by fibroblast-like cells may take place already at the level of MGUS and may become even more pronounced in MM. The identified proteins which indicate the stepwise progression from MGUS to MM may offer new tools for therapeutic strategies.
Collapse
Affiliation(s)
- Astrid Slany
- Faculty of Chemistry, Institute of Analytical Chemistry, University of Vienna , Währingerstraße 38, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Cohen Y, Gutwein O, Garach-Jehoshua O, Bar-Haim A, Kornberg A. The proliferation arrest of primary tumor cells out-of-niche is associated with widespread downregulation of mitotic and transcriptional genes. ACTA ACUST UNITED AC 2013; 19:286-92. [PMID: 24074379 DOI: 10.1179/1607845413y.0000000125] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
In recording the changes acquired in gene expression profile during culture of fresh bone marrow samples from patients with multiple myeloma or acute myeloid leukemia, the most remarkable finding in both instances was widespread downregulation of mitotic and transcriptional genes (e.g. MKI67, CCNB1, ASPM, SGOL1, DLGAP5, CENPF, BUB1, KIF23, KIF18a, KIF11, KIF14, KIF4, NUF2, KIF1, AE2FB, TOP2A, NCAPG, TTK, CDC20, and AURKB), which could account for the ensuing proliferation arrest. Many of these genes were also underexpressed in leukemic cells from the blood or myeloma cells from an extramedullary site compared with their expression in the aspirates. Taken together, our results exhibited mitotic and transcriptional gene subsets where their expression appears to be coordinated and niche dependent. In addition, the genes induced during culture specified a variety of angiogenic factors (e.g. interleukin-8 and CXCL-5) and extracellular matrix proteins (e.g. osteopontin and fibronectin) probably released by the tumor cells while generating their favored microenvironment.
Collapse
|
23
|
Amodio N, Bellizzi D, Leotta M, Raimondi L, Biamonte L, D'Aquila P, Di Martino MT, Calimeri T, Rossi M, Lionetti M, Leone E, Passarino G, Neri A, Giordano A, Tagliaferri P, Tassone P. miR-29b induces SOCS-1 expression by promoter demethylation and negatively regulates migration of multiple myeloma and endothelial cells. Cell Cycle 2013; 12:3650-62. [PMID: 24091729 DOI: 10.4161/cc.26585] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Epigenetic silencing of tumor suppressor genes frequently occurs and may account for their inactivation in cancer cells. We previously demonstrated that miR-29b is a tumor suppressor microRNA (miRNA) that targets de novo DNA methyltransferases and reduces the global DNA methylation of multiple myeloma (MM) cells. Here, we provide evidence that epigenetic activity of miR-29b leads to promoter demethylation of suppressor of cytokine signaling-1 (SOCS-1), a hypermethylated tumor suppressor gene. Enforced expression of synthetic miR-29b mimics in MM cell lines resulted in SOCS-1 gene promoter demethylation, as assessed by Sequenom MassARRAY EpiTYPER analysis, and SOCS-1 protein upregulation. miR-29b-induced SOCS-1 demethylation was associated with reduced STAT3 phosphorylation and impaired NFκB activity. Downregulation of VEGF-A and IL-8 mRNAs could be detected in MM cells transfected with miR-29b mimics as well as in endothelial (HUVEC) or stromal (HS-5) cells treated with conditioned medium from miR-29b-transfected MM cells. Notably, enforced expression of miR-29b mimics increased adhesion of MM cells to HS-5 and reduced migration of both MM and HUVEC cells. These findings suggest that miR-29b is a negative regulator of either MM or endothelial cell migration. Finally, the proteasome inhibitor bortezomib, which induces the expression of miR-29b, decreased global DNA methylation by a miR-29b-dependent mechanism and induced SOCS-1 promoter demethylation and protein upregulation. In conclusion, our data indicate that miR-29b is endowed with epigenetic activity and mediates previously unknown functions of bortezomib in MM cells.
Collapse
Affiliation(s)
- Nicola Amodio
- Department of Experimental and Clinical Medicine; Magna Graecia University and Medical Oncology Unit; T. Campanella Cancer Center; Salvatore Venuta University Campus; Catanzaro, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Pappa CA, Tsirakis G, Samiotakis P, Tsigaridaki M, Alegakis A, Goulidaki N, Alexandrakis MG. Serum levels of angiopoietin-2 are associated with the growth of multiple myeloma. Cancer Invest 2013; 31:385-9. [PMID: 23758184 DOI: 10.3109/07357907.2013.800093] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Angiopoietins and their receptor, Tie-2, have crucial role in angiogenesis. We measured serum levels of angiopoietin-2 (Ang-2), soluble Tie-2, and factors of burden and prognosis in myeloma (LDH, CRP, beta-2 microglobulin, and interleukin-6) in 55 newly diagnosed patients, with 30 of them in plateau phase, in order to note correlations among them. Levels of Ang-2 were higher in patients in advanced stage of disease, decreased in plateau phase, and correlated with all other factors. Circulating Ang-2 in myeloma patients significantly correlated to factors of disease burden and prognosis, and therefore measuring its levels may be important for the valuation of the disease.
Collapse
Affiliation(s)
- Constantina A Pappa
- Hematology Department, Venizelion Hospital of Heraklion, Heraklion, Crete, Greece
| | | | | | | | | | | | | |
Collapse
|
25
|
Asimakopoulos F, Kim J, Denu RA, Hope C, Jensen JL, Ollar SJ, Hebron E, Flanagan C, Callander N, Hematti P. Macrophages in multiple myeloma: emerging concepts and therapeutic implications. Leuk Lymphoma 2013; 54:2112-21. [PMID: 23432691 DOI: 10.3109/10428194.2013.778409] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Multiple myeloma, a clonal plasma cell malignancy, has long provided a prototypic model to study regulatory interactions between malignant cells and their microenvironment. Myeloma-associated macrophages have historically received limited scrutiny, but recent work points to central and non-redundant roles in myeloma niche homeostasis. The evidence supports a paradigm of complex, dynamic and often mutable interactions between macrophages and other cellular constituents of the niche. We and others have shown that macrophages support myeloma cell growth, viability and drug resistance through both contact-mediated and non-contact-mediated mechanisms. These tumor-beneficial roles have evolved in opposition to, or in parallel with, intrinsic pro-inflammatory and tumoricidal properties. Thus, simple blockade of protective "don't eat me" signals on the surface of myeloma cells leads to macrophage-mediated myeloma cell killing. Macrophages also enhance the tumor-supportive role of mesenchymal stem/stromal cells (MSCs) in the niche: importantly, this interaction is bidirectional, producing a distinct state of macrophage polarization that we termed "MSC-educated macrophages." The intriguing pattern of cross-talk between macrophages, MSCs and tumor cells highlights the myeloma niche as a dynamic multi-cellular structure. Targeted reprogramming of these interactions harbors significant untapped therapeutic potential, particularly in the setting of minimal residual disease, the main obstacle toward a cure.
Collapse
Affiliation(s)
- Fotis Asimakopoulos
- Division of Hematology/Oncology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health , Madison, WI , USA and University of Wisconsin Carbone Cancer Center , Madison, WI , USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Tijono SM, Guo K, Henare K, Palmer BD, Wang LCS, Albelda SM, Ching LM. Identification of human-selective analogues of the vascular-disrupting agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA). Br J Cancer 2013; 108:1306-15. [PMID: 23481185 PMCID: PMC3619269 DOI: 10.1038/bjc.2013.101] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Background: Species selectivity of DMXAA (5,6-dimethylxanthenone-4-acetic acid, Vadimezan) for murine cells over human cells could explain in part the recent disappointing phase III trials clinical results when preclinical studies were so promising. To identify analogues with greater human clinical potential, we compared the activity of xanthenone-4-acetic acid (XAA) analogues in murine or human cellular models. Methods: Analogues with a methyl group systematically substituted at different positions of the XAA backbone were evaluated for cytokine induction in cultured murine or human leukocytes; and for anti-vascular effects on endothelial cells on matrigel. In vivo antitumour activity and cytokine production by stromal or cancer cells was measured in human A375 and HCT116 xenografts. Results: Mono-methyl XAA analogues with substitutions at the seventh and eighth positions were the most active in stimulating human leukocytes to produce IL-6 and IL-8; and for inhibition of tube formation by ECV304 human endothelial-like cells, while 5- and 6-substituted analogues were the most active in murine cell systems. Conclusion: Xanthenone-4-acetic acid analogues exhibit extreme species selectivity. Analogues that are the most active in human systems are inactive in murine models, highlighting the need for the use of appropriate in vivo animal models in selecting clinical candidates for this class of compounds.
Collapse
Affiliation(s)
- S M Tijono
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, School of Medicine, University of Auckland, Auckland, New Zealand
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Bruton’s tyrosine kinase (Btk) is intimately involved in multiple signal-transduction pathways regulating survival, activation, proliferation, and differentiation of B-lineage lymphoid cells. Btk is overexpressed and constitutively active in several B-lineage lymphoid malignancies. Btk has emerged as a new antiapoptotic molecular target for treatment of B-lineage leukemias and lymphomas. Preclinical and early clinical results indicate that Btk inhibitors may be useful in the treatment of leukemias and lymphomas.
Collapse
Affiliation(s)
- Osmond J D'Cruz
- Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | | |
Collapse
|
28
|
Pappa CA, Tsirakis G, Boula A, Sfiridaki A, Psarakis FE, Alexandrakis MG, Stathopoulos EN. The Significance of non Correlation Between Interleukin-8 Serum Levels with Bone Marrow Microvascular Density in Patients with Myeloma Multiple. Pathol Oncol Res 2013; 19:539-43. [DOI: 10.1007/s12253-013-9614-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 02/11/2013] [Indexed: 02/07/2023]
|
29
|
Siegel DS. Relapsed/Refractory multiple myeloma: defining refractory disease and identifying strategies to overcome resistance. Semin Hematol 2012; 49 Suppl 1:S3-15. [PMID: 22727390 DOI: 10.1053/j.seminhematol.2012.05.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Despite the development of more effective therapies for multiple myeloma (MM) over the past decade, nearly all patients will eventually experience disease relapse and require further therapy. Designing the next generation of therapies for relapsed and refractory disease will depend on understanding the complex molecular pathogenesis of MM and mechanisms of resistance. Oncogenomic studies have identified many potential therapeutic targets and have led to emerging models of the multistep molecular pathogenesis of MM. The key to overcoming resistance may depend on interrupting the complex interactions between MM cells and the bone microenvironment. Direct interaction between MM cells and bone marrow cells activates pleiotropic signaling pathways that mediate growth, survival, and migration of MM cells as well as resistance to chemotherapy (known as cell adhesion-mediated drug resistance). The bone marrow also secretes growth factors and cytokines that maintain MM cells and inhibit apoptosis. Therefore, successful therapeutic strategies must target not only the MM plasma cell but also the bone microenvironment. The benefit of immunomodulatory drugs such as thalidomide and lenalidomide and the proteasome inhibitor bortezomib in relapsed/refractory MM is related to their ability to target both. Novel agents and combination strategies are building on the success of these agents and targeting synergistic pathways.
Collapse
Affiliation(s)
- David S Siegel
- Hackensack University Medical Center, Hackensack, NJ 07601, USA.
| |
Collapse
|
30
|
Liang P, Cheng SH, Cheng CK, Lau KM, Lin SY, Chow EYD, Chan NPH, Ip RKL, Wong RSM, Ng MHL. Platelet factor 4 induces cell apoptosis by inhibition of STAT3 via up-regulation of SOCS3 expression in multiple myeloma. Haematologica 2012; 98:288-95. [PMID: 22929979 DOI: 10.3324/haematol.2012.065607] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Platelet factor 4 (PF4) is an angiostatic chemokine that suppresses tumor growth and metastasis. We previously revealed frequent transcriptional silencing of PF4 in multiple myeloma, but the functional roles of this chemokine are still unknown. We studied the apoptotic effects of PF4 on myeloma cell lines and primary myeloma in vitro, and investigated the involved signaling pathway. The in vivo effects were also studied using a mouse model. PF4 not only suppressed myeloma-associated angiogenesis, but also inhibited growth and induced apoptosis in myeloma cells. We found that PF4 negatively regulated STAT3 and concordantly inhibited constitutive and interleukin-6-induced phosphorylation of STAT3, and down-regulated the expression of STAT3 target genes (Mcl-1, survivin and VEGF). Overexpression of constitutively activated STAT3 could rescue PF4-induced apoptotic effects. Furthermore, we found that PF4 induced the expression of SOCS3, a STAT3 inhibitor, and gene silencing of SOCS3 abolished its ability to inhibit STAT3 activation, suggesting a critical role of SOCS3 in PF4-induced STAT3 inhibition. Knockdown of LRP1, a putative PF4 receptor, could also abolish PF4-induced apoptosis and STAT3 inhibition. Finally, the tumor growth inhibitory effect of PF4 was confirmed by in vivo mouse models. Immunostaining of rabbit bone xenografts from PF4-treated mice showed induction of apoptosis of myeloma cells and inhibition of angiogenesis, which was associated with suppression of STAT3 activity. Together, our preclinical data indicate that PF4 may be a potential new targeting agent for the treatment of myeloma.
Collapse
Affiliation(s)
- Pei Liang
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, the Chinese University of Hong Kong, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Bruton tyrosine kinase inhibition is a novel therapeutic strategy targeting tumor in the bone marrow microenvironment in multiple myeloma. Blood 2012; 120:1877-87. [PMID: 22689860 DOI: 10.1182/blood-2011-12-396853] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Bruton tyrosine kinase (Btk) has a well-defined role in B-cell development, whereas its expression in osteoclasts (OCs) further suggests a role in osteoclastogenesis. Here we investigated effects of PCI-32765, an oral and selective Btk inhibitor, on osteoclastogenesis as well as on multiple myeloma (MM) growth within the BM microenvironment. PCI-32765 blocked RANKL/M-CSF-induced phosphorylation of Btk and downstream PLC-γ2 in OCs, resulting in diminished TRAP5b (ED50 = 17 nM) and bone resorption activity. PCI-32765 also inhibited secretion of multiple cytokines and chemokines from OC and BM stromal cell cultures from both normal donors (ED50 = 0.5 nM) and MM patients. It decreased SDF-1-induced migration of MM cells, and down-regulated MIP1-α/CCL3 in MM cells. It also blocked MM cell growth and survival triggered by IL-6 or coculture with BM stromal cells or OCs in vitro. Importantly, PCI-32765 treatment significantly inhibits in vivo MM cell growth (P < .03) and MM cell-induced osteolysis of implanted human bone chips in SCID mice. Moreover, PCI-32765 prevents in vitro colony formation by stem-like cells from MM patients. Together, these results delineate functional sequelae of Btk activation mediating osteolysis and growth of MM cells, supporting evaluation of PCI-32765 as a novel therapeutic in MM.
Collapse
|
32
|
Vangsted A, Klausen TW, Vogel U. Genetic variations in multiple myeloma I: effect on risk of multiple myeloma. Eur J Haematol 2011; 88:8-30. [PMID: 21883480 DOI: 10.1111/j.1600-0609.2011.01700.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Few risk factors have been established for the plasma cell disorder multiple myeloma, but some of these like African American ethnicity and a family history of B-cell lymphoproliferative diseases suggest a genetic component for the disease. Genetic variation represents the genetic basis of variability in a population. The complex interplay between environment and genes for the development of cancer may therefore be influenced by genetic variations. A genetic variation may change the function of the gene, and if the genetic variation is associated with the risk of disease, that particular gene may be involved in the pathogenesis of disease. Genes of interest are genes involved in the normal development and function of the plasma cell and genes that protect us against exposures from the environment, for example, genes involved in the metabolism of xenobiotics, metabolism of folate and methionine, as well as genes involved in inflammation and DNA repair. Identification of genes with potential influence on cancer risk may help us to establish relevant laboratory studies on exposure and dose-response assessment and may help us to test the hypothesis in epidemiological studies. Knowledge of individual at high risk of cancer may offer promising insight for the prevention of cancer.
Collapse
Affiliation(s)
- Annette Vangsted
- Department of Haematology, Roskilde Hospital, Copenhagen University, Roskilde, Denmark.
| | | | | |
Collapse
|
33
|
Alhamdani MSS, Hoheisel JD. Antibody Microarrays in Proteome Profiling. MOLECULAR ANALYSIS AND GENOME DISCOVERY 2011:219-243. [DOI: 10.1002/9781119977438.ch10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
34
|
Pappa CA, Tsirakis G, Kanellou P, Kaparou M, Stratinaki M, Xekalou A, Alegakis A, Boula A, Stathopoulos EN, Alexandrakis MG. Monitoring serum levels ELR+ CXC chemokines and the relationship between microvessel density and angiogenic growth factors in multiple myeloma. Cytokine 2011; 56:616-20. [PMID: 21940178 DOI: 10.1016/j.cyto.2011.08.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Revised: 07/23/2011] [Accepted: 08/23/2011] [Indexed: 01/12/2023]
Abstract
BACKGROUND The ELR+ CXC chemokines are important mediators of tumorigenesis, related to their angiogenic properties. Angiogenesis appears to be a prominent feature in the progression of multiple myeloma (MM). CXC chemokines have four highly conserved cysteine amino acid residues, with the first two cysteine molecules separated by a single amino acid. The angiogenic potential of this group is determined by the presence of three amino acid residues (Glu-Leu-Arg: the ELR motif) preceding the first cysteine amino acid, in the NH2 terminus. AIMS The purpose of this study was to determine serum concentrations of angiogenesis-related chemokines ELR+ motif, such as interleukin-8 (IL-8), epithelial neutrophil activating protein-78 (ENA-78) and growth-related gene alpha (GRO-α), as well the bone marrow microvascular density (MVD) in patients with MM at diagnosis and after treatment, in plateau phase. We also evaluated the relationship among them with other known growth factors involved in angiogenesis. METHODS Serum levels of the ELR+ CXC chemokines: IL-8, ENA-78 and GRO-α as well as of the angiogenic factors: hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF) and tumor necrosis factor-α (TNF-α) were determined in 63 newly diagnosed MM patients, in 30 in plateau phase and in 20 healthy controls. Serum measurements of them were performed with commercially available kits for ELISA. Bone marrow biopsies were performed before and after treatment, in plateau phase, in order to determine MVD by staining vessels with anti-CD31. RESULTS Serum concentrations of IL-8, ENA-78, GRO-α and TNF-α were significantly higher in the group of MM patients (44.5±25.3, 765±572.1, 186.5±129.1 and 4.2±2.8 pg/ml, respectively) in comparison to control group (27.3±6.4, 335.1±268.6, 112.5±76.1 and 1.3±0.8 pg/ml) (p<0.02 for GRO-α, p<0.001 for other cases). We also found that untreated patients had higher levels of IL-8, ENA-78, GRO-α than post treatment patients, but statistical significant difference was found only for IL-8 (48.36±30.93 pg/ml vs. 35.05±19.77 pg/ml, p<0.001). Furthermore IL-8, GRO-α, TNF-α, HGF and VEGF were significantly higher with increasing disease stage (p<0.001 in all cases). ENA-78 serum levels were higher in stage III than in stage I and II, but without statistical significance. Additionally we correlated each proinflammatory cytokine with well known angiogenic factors such as HGF, VEGF and TNF-α. A positive correlation was found between serum HGF and IL-8 and GRO-α (r=0.316 p<0.01, r=0.297 p<0.02, respectively). Similarly serum VEGF correlated with ENA-78 and GRO-α (r=0.323 p<0.01, r=0.469 p<0.001, respectively). In the pretreatment group of patients a positive correlation between bone marrow MVD and serum levels of GRO-α was found (r=0.304 p<0.01). There was a difference in survival times between patients with higher than median versus low IL-8, ENA-78 and GRO-α levels, but the differences could not reach statistical significance in either case. CONCLUSIONS These findings support the hypothesis that ELR+ motif CXC chemokines, such as IL-8, ENA-78 and GRO-α correlate with angiogenic growth factors and may play a role in the progression of MM. Further studies are needed to determine their prognostic and predictive significance.
Collapse
Affiliation(s)
- C A Pappa
- Department of Hematology, Venizelion Hospital of Heraklion, and Crete, Greece
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Tsirakis G, Pappa CA, Kaparou M, Katsomitrou V, Hatzivasili A, Alegakis T, Xekalou A, Stathopoulos EN, Alexandrakis MG. Assessment of proliferating cell nuclear antigen and its relationship with proinflammatory cytokines and parameters of disease activity in multiple myeloma patients. Eur J Histochem 2011; 55:e21. [PMID: 22073368 PMCID: PMC3203474 DOI: 10.4081/ejh.2011.e21] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Accepted: 04/21/2011] [Indexed: 12/02/2022] Open
Abstract
Multiple myeloma (MM) is a malignant plasma cell disease. Several proinflammatory cytokines produced by malignant plasma cells and bone marrow (BM) stromal cells are involved in the pathogenesis of the disease. We evaluated serum levels of the proinflammatory cytokines Interleukin-1β (IL-1β), Interleukin-6 (IL-6), Interleukin-8 (IL-8), macrophage inflammatory protein-1α (MIP-1α), in MM patients before treatment, and determined its significance in tumor progression. We also analyzed the correlation between measured parameters with proliferating cell nuclear antigen (PCNA). Forty-four MM patients and 20 healthy controls were studied. Serum levels of the proinflammatory cytokines were measured using enzyme-linked immunosorbent assay (ELISA), whereas PCNA value in the BM was determined by immunohistochemistry staining. The mean concentrations of the measured cytokines were significantly different among the three stages of disease, with higher values in advanced disease stage. Furthermore, patients with MM had significantly higher serum levels of the measured cytokines than in controls. A positive correlation was found between IL-6 with IL-1β, IL-8 and MIP-1α. Similarly, IL-8 and MIP-1α were positively correlated with markers of disease activity such as β2 microglobulin and LDH. The proliferation index, determined by PCNA immunostaining, was higher in advanced disease stage. Furthermore PCNA value correlated significantly with β2 microglobulin, LDH and the levels of the measured cytokines. Our results showed that the proliferative activity, as measured with PCNA, increases in parallel with disease stage. The positive correlation between PCNA and other measured mediators supports the involvement of these factors in the biology of myeloma cell growth and can be used as markers of disease activity and as possible therapeutic targets.
Collapse
Affiliation(s)
- G Tsirakis
- Department of Haematology, University Hospital of Heraklion, Crete, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Angiogenesis and multiple myeloma. CANCER MICROENVIRONMENT 2011; 4:325-37. [PMID: 21735169 DOI: 10.1007/s12307-011-0072-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 06/23/2011] [Indexed: 01/13/2023]
Abstract
The bone marrow microenvironment in multiple myeloma is characterized by an increased microvessel density. The production of pro-angiogenic molecules is increased and the production of angiogenic inhibitors is suppressed, leading to an "angiogenic switch". Here we present an overview of the role of angiogenesis in multiple myeloma, the pro-angiogenic factors produced by myeloma cells and the microenvironment, and the mechanisms involved in the myeloma-induced angiogenic switch. Current data suggest that the increased bone marrow angiogenesis in multiple myeloma is due to the aberrant expression of angiogenic factors by myeloma cells, the subsequent increase in pro-angiogenic activity of normal plasma cells as a result of myeloma cell angiogenic activity, and the increased number of plasma cells overall. Hypoxia also contributes to the angiogenic properties of the myeloma marrow microenvironment. The transcription factor hypoxia-inducible factor-1α is overexpressed by myeloma cells and affects their transcriptional and angiogenic profiles. In addition, potential roles of the tumor suppressor gene inhibitor of growth family member 4 and homeobox B7 have also been recently highlighted as repressors of angiogenesis and pro-angiogenic related genes, respectively. This complex pathogenetic model of myeloma-induced angiogenesis suggests that several pro-angiogenic molecules and related genes in myeloma cells and the microenvironment are potential therapeutic targets.
Collapse
|
37
|
Anderson KC, Carrasco RD. Pathogenesis of myeloma. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2011; 6:249-74. [PMID: 21261519 DOI: 10.1146/annurev-pathol-011110-130249] [Citation(s) in RCA: 204] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Multiple myeloma (MM) is a neoplasm of post-germinal center, terminally differentiated B cells. It is characterized by a multifocal proliferation of clonal, long-lived plasma cells within the bone marrow (BM) and associated skeletal destruction, serum monoclonal gammopathy, immune suppression, and end-organ sequelae. MM is preceded by an age-progressive premalignant condition termed monoclonal gammopathy of undetermined significance. Unlike the genomes of most hematological malignancies, and similar to those of solid-tissue neoplasms, MM genomes are typified by numerous structural and numerical chromosomal aberrations as well as mutations in a number of oncogenes and tumor-suppressor genes, some of which have been linked to disease pathogenesis and clinical behavior. Recent studies have also defined the importance of interactions between the MM cells and their BM microenvironment, dysregulation in signaling pathways and in a specialized subpopulation of cells within the tumor (termed myeloma cancer stem cells) for tumor cell growth and survival, and the development of resistance to therapy.
Collapse
Affiliation(s)
- Kenneth C Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
38
|
Joshi S, Khan R, Sharma M, Kumar L, Sharma A. Angiopoietin-2: a potential novel diagnostic marker in multiple myeloma. Clin Biochem 2011; 44:590-5. [PMID: 21300047 DOI: 10.1016/j.clinbiochem.2011.01.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 01/17/2011] [Accepted: 01/24/2011] [Indexed: 11/17/2022]
Abstract
BACKGROUND Multiple myeloma (MM) was the first hematological malignancy in which a prognostic relevance of bone marrow (BM) angiogenesis was reported. Microenvironment formed by reactive oxygen species, growth factors and chemokines initiates the process of BM angiogenesis. Among the angiopoietins, angiopoietin-1 is involved in vessel stability, whereas angiopoietin-2 is involved in vessel sprouting. DESIGN AND METHODS Circulatory levels of angiopoietin-1, angiopoietin-2 and VEGF in 62 MM patients and 50 healthy controls were determined using ELISA. RESULTS Significant increase in VEGF and angiopoietin-2 level was observed in patients which correlated positively with the severity of the disease, whereas no alteration was observed in angiopoietin-1 levels. CONCLUSION Significant elevation in angiopoietin-2 and VEGF levels and their correlation with severity of the disease indicate their utility as potential tumor markers along with β(2)- microglobulin and might also suggest new therapeutic target for anticancer treatment.
Collapse
Affiliation(s)
- Suhasini Joshi
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi-110029, India
| | | | | | | | | |
Collapse
|
39
|
Xie JY, Li MX, Xiang DB, Mou JH, Qing Y, Zeng LL, Yang ZZ, Guan W, Wang D. Elevated Expression of APE1/Ref-1 and its Regulation on IL-6 and IL-8 in Bone Marrow Stromal Cells of Multiple Myeloma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2010; 10:385-93. [DOI: 10.3816/clml.2010.n.072] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
40
|
Bone marrow stromal cells from multiple myeloma patients uniquely induce bortezomib resistant NF-kappaB activity in myeloma cells. Mol Cancer 2010; 9:176. [PMID: 20604947 PMCID: PMC3095250 DOI: 10.1186/1476-4598-9-176] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Accepted: 07/06/2010] [Indexed: 01/10/2023] Open
Abstract
Background Components of the microenvironment such as bone marrow stromal cells (BMSCs) are well known to support multiple myeloma (MM) disease progression and resistance to chemotherapy including the proteasome inhibitor bortezomib. However, functional distinctions between BMSCs in MM patients and those in disease-free marrow are not completely understood. We and other investigators have recently reported that NF-κB activity in primary MM cells is largely resistant to the proteasome inhibitor bortezomib, and that further enhancement of NF-κB by BMSCs is similarly resistant to bortezomib and may mediate resistance to this therapy. The mediating factor(s) of this bortezomib-resistant NF-κB activity is induced by BMSCs is not currently understood. Results Here we report that BMSCs specifically derived from MM patients are capable of further activating bortezomib-resistant NF-κB activity in MM cells. This induced activity is mediated by soluble proteinaceous factors secreted by MM BMSCs. Among the multiple factors evaluated, interleukin-8 was secreted by BMSCs from MM patients at significantly higher levels compared to those from non-MM sources, and we found that IL-8 contributes to BMSC-induced NF-κB activity. Conclusions BMSCs from MM patients uniquely enhance constitutive NF-κB activity in MM cells via a proteinaceous secreted factor in part in conjunction with IL-8. Since NF-κB is known to potentiate MM cell survival and confer resistance to drugs including bortezomib, further identification of the NF-κB activating factors produced specifically by MM-derived BMSCs may provide a novel biomarker and/or drug target for the treatment of this commonly fatal disease.
Collapse
|
41
|
Gullo C, Koh LK, Pang WL, Ho KT, Tan SH, Schwarz H. Inhibition of proliferation and induction of apoptosis in multiple myeloma cell lines by CD137 ligand signaling. PLoS One 2010; 5:e10845. [PMID: 20520765 PMCID: PMC2877096 DOI: 10.1371/journal.pone.0010845] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 04/30/2010] [Indexed: 01/24/2023] Open
Abstract
Background Multiple myeloma (MM) is a malignancy of terminally-differentiated plasma cells, and the second most prevalent blood cancer. At present there is no cure for MM, and the average prognosis is only three to five years. Current treatments such as chemotherapy are able to prolong a patient's life but rarely prevent relapse of the disease. Even hematopoietic stem cell transplants and novel drug combinations are often not curative, underscoring the need for a continued search for novel therapeutics. CD137 and its ligand are members of the Tumor Necrosis Factor (TNF) receptor and TNF superfamilies, respectively. Since CD137 ligand cross-linking enhances proliferation and survival of healthy B cells we hypothesized that it would also act as a growth stimulus for B cell cancers. Methodology/Principal Findings Proliferation and survival of B cell lymphoma cell lines were not affected or slightly enhanced by CD137 ligand agonists in vitro. But surprisingly, they had the opposite effects on MM cells, where CD137 ligand signals inhibited proliferation and induced cell death by apoptosis. Furthermore, secretion of the pro-inflammatory cytokines, IL-6 and IL-8 were also enhanced in MM but not in non-MM cell lines in response to CD137 ligand agonists. The secretion of these cytokines in response to CD137 ligand signaling was consistent with the observed activation of the classical NF-κB pathway. We hypothesize that the induction of this pathway results in activation-induced cell death, and that this is the underlying mechanism of CD137-induced MM cell death and growth arrest. Conclusions/Significance These data point to a hitherto unrecognized role of CD137 and CD137 ligand in MM cell biology. The selective inhibition of proliferation and induction of cell death in MM cells by CD137 ligand agonists may also warrant a closer evaluation of their therapeutic potential.
Collapse
Affiliation(s)
- Charles Gullo
- Cancer Immunology Laboratory, Department of Clinical Research, Singapore General Hospital, Singapore, Singapore
| | - Liang Kai Koh
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wan Lu Pang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kian Tong Ho
- Cancer Immunology Laboratory, Department of Clinical Research, Singapore General Hospital, Singapore, Singapore
| | - Shi Hao Tan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Herbert Schwarz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
42
|
Singh S, Sadanandam A, Varney ML, Nannuru KC, Singh RK. Small interfering RNA-mediated CXCR1 or CXCR2 knock-down inhibits melanoma tumor growth and invasion. Int J Cancer 2010; 126:328-36. [PMID: 19585580 DOI: 10.1002/ijc.24714] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
CXCR1 and CXCR2 are receptors for CXCL-8 and are differentially expressed on melanoma and endothelial cells. In this study, we determined the functional role of these receptors in melanoma progression. We stably knock-down the expression of CXCR1 and/or CXCR2 in A375-SM (SM; high metastatic) human melanoma cells by short-hairpin RNA transfection. Cell proliferation, migration, invasion, ERK phosphorlyation and cytoskeletal rearrangements were carried out in vitro. In vivo growth was evaluated using murine subcutaneous xenograft model. Our data demonstrate that knock-down of CXCR1 and/or CXCR2 expression, inhibited melanoma cell proliferation, survival, migration and invasive potential in vitro. Moreover, we also observed inhibition of ERK phosphorylation and cytoskeltal rearrangement in SM-shCXCR1, SM-shCXCR2 and SM-shCXCR1/2 cells. Furthermore, when SM-shCXCR1 or SM-shCXCR2 cells implanted in nude mice, tumor growth, proliferation and microvessel density was significantly inhibited as compared to SM-control cells. In addition, we observed a significant increase in melanoma cell apoptosis in SM-shCXCR1 and SM-shCXCR2 tumors compared to SM-control tumors. Together, these data demonstrate that CXCR1 and CXCR2 expression play a critical role in human melanoma tumor progression and, functional blockade of CXCR1 and CXCR2 could be potentially used for future therapeutic intervention in malignant melanoma.
Collapse
Affiliation(s)
- Seema Singh
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, 68198-5900, USA
| | | | | | | | | |
Collapse
|
43
|
Abstract
The aggressiveness of malignant melanoma is associated with differential expression of CXCL-8 and its receptors, CXCR1 and CXCR2. However, the precise functional role of these receptors in melanoma progression remains unclear. In this study, we investigate the precise functional role of CXCR1 and CXCR2 in melanoma progression. CXCR1 or CXCR2 were stably overexpressed in human melanoma cell lines, SBC-2 (non-tumourigenic) and A375P (low-tumourigenic) exhibiting low endogenous expression of receptors. Functional assays were performed to study the resulting changes in cell proliferation, motility and invasion, and in vivo tumour growth using a mouse xenograft model. Our data demonstrated that CXCR1- or CXCR2-overexpressing SBC-2 and A375P melanoma cells had enhanced proliferation, chemotaxis and invasiveness in vitro. Interestingly, CXCR1 or CXCR2 overexpression in SBC-2 cells induced tumourigenicity, and A375P cells significantly enhanced tumour growth as examined in vivo. Immunohistochemical analyses showed significantly increased tumour cell proliferation and microvessel density and reduced apoptosis in tumours generated from CXCR1- or CXCR2-overexpressing melanoma cells. CXCR1- or CXCR2-induced modulation of melanoma cell proliferation and migration was observed to be mediated through the activation of ERK1/2 phosphorylation. Together, these studies demonstrate that CXCR1 and CXCR2 play essential role in growth, survival, motility and invasion of human melanoma.
Collapse
|
44
|
Guler N, Turgut M, Ozatli D, Turgut Y, Gokce AK, Koc S, Albayrak D. High ratio of Duffy (a+b+) phenotype in patients with multiple myeloma compared to healthy controls. Hematol Oncol 2009; 27:50-1. [PMID: 19206111 DOI: 10.1002/hon.887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
45
|
Yu X, Schneiderhan-Marra N, Hsu HY, Bachmann J, Joos TO. Protein microarrays: effective tools for the study of inflammatory diseases. Methods Mol Biol 2009; 577:199-214. [PMID: 19718518 DOI: 10.1007/978-1-60761-232-2_15] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Inflammation is a defense reaction of an organism against harmful stimuli such as tissue injury or infectious agents. The relationship between the infecting microorganism and the immune, inflammatory, and coagulation responses of the host is intricately intertwined. Due to its complex nature, the molecular mechanisms of inflammation are not yet understood in detail and additional diagnostic tools are required to clarify further aspects. In recent years, protein microarray-based research has moved from being technology-based to application-oriented. Protein microarrays are perfect tools for studying inflammatory diseases. High-density protein arrays enable new classes of autoantibodies, which cause autoimmune diseases, to be discovered. Protein arrays consisting of miniaturized multiplexed sandwich immunoassays allow the simultaneous expression analysis of dozens of signaling molecules such as the cytokines and chemokines involved in the regulation of the immune system. The data enable statements to be made on the status of the disease and its progression as well as support for the clinicians in choosing patient-specific treatment. This chapter reviews the technology and the applications of protein microarrays in diagnosing and monitoring inflammatory diseases.
Collapse
Affiliation(s)
- Xiaobo Yu
- NMI Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany
| | | | | | | | | |
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW This review aims to summarize recent advances in the mechanisms through which the activation of the transcription factor NF-kappaB contributes to the pathogenesis of multiple myeloma. RECENT FINDINGS This transcription factor regulates expression of numerous genes involved in multiple myeloma pathogenesis, including growth, survival, immortalization, angiogenesis and metastasis. Recently, mutations of NF-kappaB signaling molecules have been identified in multiple myeloma cells. In addition, interactions between multiple myeloma cells and the bone marrow environment play critical roles in NF-kappaB activation as well as in multiple myeloma pathogenesis. Moreover, several drugs that are effective against multiple myeloma, including bortezomib, thalidomide, lenalidomide and arsenic trioxide, have been found to block activation of NF-kappaB. The combination of conventional chemotherapeutic drugs and those that block NF-kappaB activation has now proven to be effective in the treatment of multiple myeloma. SUMMARY Recent studies further underscore the critical role of NF-kappaB in multiple myeloma pathogenesis and have provided the rationale for multiple myeloma therapy with NF-kappaB-specific inhibitors combined with conventional chemotherapeutic drugs.
Collapse
|
47
|
Lee HL, Eom HS, Yun T, Kim HJ, Park WS, Nam BH, Moon-Woo S, Lee DH, Kong SY. Serum and urine levels of interleukin-8 in patients with non-Hodgkin's lymphoma. Cytokine 2008; 43:71-5. [PMID: 18502145 DOI: 10.1016/j.cyto.2008.04.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2007] [Revised: 03/04/2008] [Accepted: 04/09/2008] [Indexed: 11/29/2022]
Abstract
Angiogenesis plays an important role in many types of cancer. Interleukin-8 (IL-8) is known to be a pro-inflammatory and pro-angiogenic cytokine, and IL-8 has been reported to be associated with tumor progression, prognosis and survival in several types of cancers. However, the role of IL-8 in non-Hodgkin's lymphoma (NHL) has not been fully determined. Here, we evaluated the usefulness of measuring serum and urine IL-8 levels in patients with NHL. We developed reference intervals for serum and urine IL-8 level in 131 control individuals. We measured serum IL-8 and urine IL-8 levels in patients with NHL, and we compared the concentrations with those of control individuals. The reference intervals for serum IL-8 and urine IL-8 corrected by creatinine (Cr) were 15.9-430.3 pg/mL and 0.0-28.4 pg/mg Cr, respectively. The concentrations of urine IL-8/Cr were significantly higher in patients than in controls (48.9+/-194.4 vs. 5.2+/-13.8 pg/mg Cr, P<0.001). However, there were no significant differences in serum IL-8 concentrations between NHL patients and controls (159.2+/-40.4 vs. 99.6+/-107.1 pg/mL; P=0.099). Receiver operating characteristic (ROC) analysis gave 0.83 and 0.43 ROC area values for urine IL-8/Cr and serum IL-8, respectively. There was no correlation between the serum and urine concentrations of IL-8 and clinical variables, the only exception being the international prognostic index (IPI), which showed a marginal correlation with urine IL-8/Cr levels (P=0.07). This study indicated that urine IL-8/Cr levels might be useful as a diagnostic marker of NHL.
Collapse
Affiliation(s)
- Hye Lin Lee
- Hematologic Malignancies Branch, Division of Translational & Clinical Research II, Research Institute & Hospital, National Cancer Center, 809 Madu-dong, Ilsan-gu, Goyang-si, Gyeonggi-do 410-769, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Colla S, Tagliaferri S, Morandi F, Lunghi P, Donofrio G, Martorana D, Mancini C, Lazzaretti M, Mazzera L, Ravanetti L, Bonomini S, Ferrari L, Miranda C, Ladetto M, Neri TM, Neri A, Greco A, Mangoni M, Bonati A, Rizzoli V, Giuliani N. The new tumor-suppressor gene inhibitor of growth family member 4 (ING4) regulates the production of proangiogenic molecules by myeloma cells and suppresses hypoxia-inducible factor-1 alpha (HIF-1alpha) activity: involvement in myeloma-induced angiogenesis. Blood 2007; 110:4464-75. [PMID: 17848618 DOI: 10.1182/blood-2007-02-074617] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Angiogenesis has a critical role in the pathophysiology of multiple myeloma (MM); however, the molecular mechanisms underlying this process are not completely elucidated. The new tumor-suppressor gene inhibitor of growth family member 4 (ING4) has been recently implicated in solid tumors as a repressor of angiogenesis. In this study, we found that ING4 expression in MM cells was correlated with the expression of the proangiogenic molecules interleukin-8 (IL-8) and osteopontin (OPN). Moreover, we demonstrate that ING4 suppression in MM cells up-regulated IL-8 and OPN, increasing the hypoxia inducible factor-1alpha (HIF-1alpha) activity and its target gene NIP-3 expression in hypoxic condition. In turn, we show that the inhibition of HIF-1alpha by siRNA suppressed IL-8 and OPN production by MM cells under hypoxia. A direct interaction between ING4 and the HIF prolyl hydroxylase 2 (HPH-2) was also demonstrated. Finally, we show that ING4 suppression in MM cells significantly increased vessel formation in vitro, blunted by blocking IL-8 or OPN. These in vitro observations were confirmed in vivo by finding that MM patients with high IL-8 production and microvascular density (MVD) have significantly lower ING4 levels compared with those with low IL-8 and MVD. Our data indicate that ING4 exerts an inhibitory effect on the production of proangiogenic molecules and consequently on MM-induced angiogenesis.
Collapse
Affiliation(s)
- Simona Colla
- Hematology and Bone Marrow Transplantation Center, University of Parma, Parma, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Hideshima T, Mitsiades C, Tonon G, Richardson PG, Anderson KC. Understanding multiple myeloma pathogenesis in the bone marrow to identify new therapeutic targets. Nat Rev Cancer 2007; 7:585-98. [PMID: 17646864 DOI: 10.1038/nrc2189] [Citation(s) in RCA: 708] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Multiple myeloma is a plasma cell malignancy characterized by complex heterogeneous cytogenetic abnormalities. The bone marrow microenvironment promotes multiple myeloma cell growth and resistance to conventional therapies. Although multiple myeloma remains incurable, novel targeted agents, used alone or in combination, have shown great promise to overcome conventional drug resistance and improve patient outcome. Recent oncogenomic studies have further advanced our understanding of the molecular pathogenesis of multiple myeloma, providing the framework for new prognostic classification and identifying new therapeutic targets.
Collapse
Affiliation(s)
- Teru Hideshima
- Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|