scholarly journals CCL2 promotes macrophages-associated chemoresistance via MCPIP1 dual catalytic activities in multiple myeloma

2019 ◽  
Vol 10 (10) ◽  
Author(s):  
Ruyi Xu ◽  
Yi Li ◽  
Haimeng Yan ◽  
Enfan Zhang ◽  
Xi Huang ◽  
...  

Abstract We previously showed that the chemokine CCL2 can recruit macrophages (Mφs) to the bone marrow (BM) in multiple myeloma (MM) and that myeloma-associated Mφs are important in drug resistance. Here, we explore the role of increased CCL2 expression in the BM microenvironment of MM and elucidate the underlying mechanism. Our results show that CCL2 expression is associated with the treatment status of MM patients. Mφs interact with MM cells and further upregulate their expression of CCL2. These increased level of CCL2 polarizes Mφs toward the M2-like phenotype and promotes Mφs to protect MM cells from drug-induced apoptosis. Mechanistically, CCL2 upregulated the expression of the immunosuppressive molecular MCP-1-induced protein (MCPIP1) in Mφs. MCPIP1 mediates Mφs’ polarization and protection via dual catalytic activities. Additionally, we found that CCL2 induces MCPIP1 expression via the JAK2-STAT3 signaling pathway. Taken together, our results indicate that increased CCL2 expression in MM patients’ BM polarizes Mφs toward the M2-like phenotype and promotes the protective effect of Mφs through MCPIP1, providing novel insight into the mechanism of Mφs-mediated drug resistance in MM.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3086-3086
Author(s):  
Ruyi Xu ◽  
Yi Li ◽  
Haimeng Yan ◽  
Xi Huang ◽  
He Huang ◽  
...  

Background: Despite with the introduction of novel chemotherapy agents to multiple myeloma (MM), chemoresistance remains the major problem in clinical management. The interaction of MM cells with different cell components in the tumor microenvironment is important for tumor growth and chemoresistance, in which macrophages (Mφs) represent the prominent components. Mφs, who provide a favorable microenvironment for MM cells, protect them from drug-induced apoptosis. Mφs have great plasticity and can differentiate into different functional states according to microenvironmental signals. We previously found that the chemokine CCL2 is overexpressed in MM patients and CCL2 promoted Mφs'infiltration in the MM-BM microenvironment. The present study aims to further investigate the clinical significance of CCL2 and elucidate the mechanism how it participates in Mφs-mediated MM chemoresistance. Methods: Clinical significance of CCL2 were investigated by immunohistochemistry and Elisa. Flow cytometry, quantitative real-time PCR and Western blotting were conducted to evaluated the apoptosis of MM cells and Mφs' polarization. RNA sequencing was performed to determine the key molecular induced by CCL2 in Mφs. Genetically modified cells (e.g., exhibiting siRNA knockdown, generating deletion mutants through lentiviruses) were employed to evaluate the functional significance of MCPIP1. Proteome Profiler Human phosphor-kinase antibody array was used to determine the changes of protein phosphorylation in Mφs caused by CCL2. MM xenograft models were used to evaluated the role of CCL2 and MCPIP1 in vivo. Results: We found that CCL2 expression is tightly associated with MM patients' treatment status. When newly diagnosed patients received 4 courses of PCD combined therapy, CCL2 expression in their BM significantly decreased. MM cell lines and PBMCs barely expressed CCL2, while Mφs highly expressed CCL2. Mφs interacted with MM cells further upregulated their expression of CCL2. These increased CCL2 in the MM' BM microenvironment had little effect on MM cells' proliferation and their drug responses, but CCL2-treated Mφs were more effective at protecting MM cells from bortezomib- and melphalan- induced apoptosis. CCL2 could also effectively polarize Mφs toward M2-like Mφs, which possess a stronger ability to protect MM cells from chemotherapy drug-induced apoptosis. RNA sequencing revealed that CCL2 significantly induced the immunosuppressive molecular MCP-1-induced protein (MCPIP1) in Mφs and the result was verified both in mRNA and protein level. MM cells cocultured with MCPIP1-knock down Mφs were more sensitive to drug-induced apoptosis than those cocultured with control Mφs. Mφs transfected with D141N mutant MCPIP1 lost their protective effects because the mutation resulted in impaired RNase and DUB activities of MCPIP1. Mφs isolated from NSG mice' tumor masses showed MCPIP1-knock down Mφs displayed a more M1-like phenotype and in vitro experiments showed MCPIP1-knock down Mφs were more difficult to be polarized toward M2-like Mφs by MM cells. CCL2 significantly activated JAK2/STAT3 pathway in Mφs and inhibition of STAT3 hindered CCL2-induced MCPIP1 protein expression. Conclusion: Our study showed that increased CCL2 in MM' BM microenvironment polarizes Mφs towards M2-like phenotype and promotes Mφs to protect MM cells from chemotherapy drug-induced apoptosis. Mechanistically, CCL2 activated JAK2/STAT3 pathway to induce the expression of MCPIP1. MCPIP1, the critical negative regulator of inflammation, mediated Mφs 'polarization and protection effect via its dual catalytic activities. Key words: Multiple Myeloma, Macrophages-Associated Chemoresistance, Polarization, CCL2, MCPIP1 Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3929-3929
Author(s):  
Liang Zhang ◽  
Jing Yang ◽  
Jianfei Qian ◽  
Sattva Neelapu ◽  
Larry Kwak ◽  
...  

Abstract Abstract 3929 Mantle cell lymphoma (MCL) is a type of aggressive B-cell non-Hodgkin lymphoma characterized by frequent resistance to conventional chemotherapy. Currently, there is no standard of care for the treatment of MCL, and patient prognosis is poor. It is well known that the tumor microenvironment plays an important role in tumor cell growth and resistance to chemotherapy. However, little is known about the microenvironment and its influence in MCL. In this study we investigated the role of IL-6 and its signaling in the growth, survival, and development of drug resistance in MCL, as IL-6 is an important cytokine for B cells and multiple myeloma. We found that the membrane IL-6 receptor gp130 is generally expressed in established MCL cell lines and in primary lymphoma cells from patients, which can be upregulated by stress such as serum starvation or low-dose chemotherapy drug treatment. Some but all not MCL cells also secrete IL-6 and/or IL-6 soluble receptor gp80. Although IL-6 and its signaling pathway do not affect MCL growth in vitro, they play an important role in MCL survival and resistance to chemotherapy drugs. Neutralizing IL-6 and/or blocking IL-6 receptors in IL-6- or gp80-secreting MCL cells increased their sensitivity to chemotherapy drug- and serum starvation-induced apoptosis. For MCL cells that do not secrete IL-6 or gp80, low doses of exogenous IL-6 or gp80 protected them from chemotherapy drug-induced apoptosis. Because T cells, macrophages, and bone marrow stromal cells (BMSCs) secrete IL-6 and/or gp80, coculture of MCL cells with peripheral blood mononuclear cells or BMSCs protected MCL cells from chemotherapy drug-induced growth inhibition and apoptosis, which can be abrogated by anti-IL-6, anti-gp80, and/or anti-gp130 antibodies. Knocking down gp80 in gp80high MCL cells rendered the cells more sensitive to chemotherapy drug-induced apoptosis, even in the presence of exogenous IL-6. Overexpression of gp80 in gp80low IL-6+ MCL cells provided protection of MCL cells from chemotherapy drug-induced apoptosis. Next, Jak2 or STAT3 inhibitors completely abrogated IL-6-mediated protection of MCL cells from apoptosis, whereas PI3K or MEK inhibitors partially abrogated IL-6-mediated protection of MCL cells from apoptosis. Furthermore, gp80-overexpressing, gp80low IL-6+ MCL cells grew faster than vector control or parental cells in the SCID mouse model, and immunohistochemistry staining showed strong surface gp80 and nuclear phosphorylated STAT3 in gp80-overexpressing MCL tumor cells. Thus, these results clearly show that IL-6 and gp80, derived from MCL cells themselves or from cells in the microenvironment, play a pivotal role in MCL cell survival and drug resistance. Although IL-6 activates Jak2/STAT3, PI3K/AKT, and MEK/Erk signaling pathways, STAT3 signaling may play an important role in mediating IL-6-induced protection of MCL against chemotherapy drug-induced apoptosis. This study suggests that targeting IL-6 and its signaling pathway may improve the efficacy of chemotherapy in MCL patients. Disclosures: Wang: Celgene: Honoraria, Research Funding; Onyx: Research Funding; Millenium: Research Funding; Novartis: Research Funding.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1915-1915
Author(s):  
Jing Yang ◽  
Jingda Xu ◽  
Zhiqiang Liu ◽  
Jin He ◽  
Huan Liu ◽  
...  

Abstract Currently, chemotherapy is the most effective treatment for multiple myeloma (MM). Although some new drugs have been shown to prolong survival in MM patients, these patients are prone to rapid relapse after high-dose treatment. Recent studies show that several bone marrow (BM) stromal cells are potentially involved in drug resistance. However, the role of other stromal cells is unclear. Adipocytes (ADs) are a major component of BM stromal cells. ADs have been shown to be involved in tumor rapid growth, metastasis, and apoptosis. Clinical studies suggest that BM ADs are associated with an increased risk of MM. Moreover, ADs isolated from patient BM biopsies were shown to support MM proliferation and migration. However, no published study has examined the importance of ADs in MM drug resistance. In addition, autophagy activation has been shown to induce drug resistance in cancer patients. We hypothesized that BM ADs protect MM cells from chemotherapy drug-induced apoptosis by autophagy activation. To examine the role of ADs in MM drug resistance, MM cells were cocultured with ADs at a ratio of 1:5 for 24 hours in medium with melphalan, dexamethasone, or bortezomib, the commonly used drugs for the treatment of MM. MM cells included primary MM cells isolated from BM aspirates of 5 MM patients and 6 MM cell lines. Human ADs were generated from mesenchymal stem cells derived from the BM mononuclear cells of healthy human fetal bones or BM aspirates of MM patients or healthy adult donors, cultured in AD medium for 2 weeks. ADs generated in vitro contained cytoplasmic Oil red O+ lipid droplets and produced triglycerol. Our results showed less drug-induced MM apoptosis in cocultures of MM cells and ADs compared with cultures of MM cells alone. Western blot analysis showed that treatment with melphalan upregulated the levels of cleaved caspase-9 and -3, but not -8, and PARP in MM cells. Compared with cultures alone, cocultures with ADs showed significantly lower levels of cleaved caspase-9, -3, and PARP in melphalan-treated MM cells. Mechanistic studies further showed that cocultures of ADs, compared with cultures alone, significantly upregulated the expression of autophagy proteins LC3B, Atg3, Atg5, and LAMP-1, but not Beclin-1. The addition of autophagy inhibitors 3-methyl adenine and chloroquine diphosphate to the cocultures remarkably enhanced apoptosis and caspase activation. Furthermore, we observed that cocultures of MM cells and ADs with either cell-cell contact or those separated by transwell inserts conferred similar protection from drug-induced apoptosis. We identified that AD-produced adipokines such as adiponection, leptin, adipsin, IL-6, MCP-1, TNF-a, and IGF-1, but not VEGF and CRP, were abundant in all examined ADs. Among these adipokines, adiponection, leptin, and adipsin were mainly produced from ADs and not from BM stromal cells, whereas other adipokines were produced from both cells. The addition of antibodies against these adipokines to the cocultures enhanced apoptosis and reduced autophagy, whereas addition of these adipokines to the cultures alone inhibited apoptosis and enhanced autophagy. In vivo studies validated these findings that injection of BM-derived ADs into the implanted human bones of SCID-hu mice bearing primary MM cells reduced response to treatment with melphalan and induced autophagy activation. Taken together, our findings elucidate a novel mechanism of MM drug resistance, through BM ADs. Our studies also provide evidence that targeting BM ADs may be a new approach to improve the efficacy of chemotherapy for the treatment of MM. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 111 (4) ◽  
pp. 2220-2229 ◽  
Author(s):  
Yulia Nefedova ◽  
Daniel M. Sullivan ◽  
Sophia C. Bolick ◽  
William S. Dalton ◽  
Dmitry I. Gabrilovich

Drug resistance remains a critical problem in the treatment of patients with multiple myeloma. Recent studies have de-termined that Notch signaling plays a major role in bone marrow (BM) stroma-mediated protection of myeloma cells from de novo drug-induced apoptosis. Here, we investigated whether pharmacologic inhibition of Notch signaling could affect the viability of myeloma cells and their sensitivity to chemotherapy. Treatment with a γ-secretase inhibitor (GSI) alone induced apoptosis of myeloma cells via specific inhibition of Notch signaling. At concentrations toxic for myeloma cell lines and primary myeloma cells, GSI did not affect normal BM or peripheral blood mononuclear cells. Treatment with GSI prevented BM stroma-mediated protection of myeloma cells from drug-induced apoptosis. The cytotoxic effect of GSI was mediated via Hes-1 and up-regulation of the proapoptotic protein Noxa. In vivo experiments using xenograft and SCID-hu models of multiple myeloma demonstrated substantial antitumor effect of GSI. In addition, GSI significantly improved the cytotoxicity of the chemotherapeutic drugs doxorubicin and melphalan. Thus, this study demonstrates that inhibition of Notch signaling prevents BM-mediated drug resistance and sensitizes myeloma cells to chemotherapy. This may represent a promising approach for therapeutic intervention in multiple myeloma.


2020 ◽  
Vol 11 (12) ◽  
Author(s):  
Lakshana Sreenivasan ◽  
Hui Wang ◽  
Shyong Quin Yap ◽  
Pascal Leclair ◽  
Anthony Tam ◽  
...  

AbstractMedulloblastoma (MB) is a high-grade pediatric brain malignancy that originates from neuronal precursors located in the posterior cranial fossa. In this study, we evaluated the role of STAT3 and IL-6 in a tumor microenvironment mediated drug resistance in human MBs. We established that the Group 3 MB cell line, Med8A, is chemosensitive (hence Med8A-S), and this is correlated with a basal low phosphorylated state of STAT3, while treatment with IL-6 induced robust increases in pY705-STAT3. Via incremental selection with vincristine, we derived the stably chemoresistant variant, Med8A-R, that exhibited multi-drug resistance, enhanced IL-6 induced pY705-STAT3 levels, and increased IL6R expression. Consequently, abrogation of STAT3 or IL6R expression in Med8A-R led to restored chemosensitivity to vincristine, highlighting a prominent role for canonical IL-6/STAT3 signaling in acquired drug resistance. Furthermore, Med8A-S subjected to conditioning exposure with IL-6, termed Med8A-IL6+ cells, exhibited enhanced vincristine resistance, increased expression of pY705-STAT3 and IL6R, and increased secretion of IL-6. When cocultured with Med8A-IL6+ cells, Med8A-S cells exhibited increased pY705-STAT3 and increased IL-6 secretion, suggesting a cytokine feedback loop responsible for amplifying STAT3 activity. Similar IL-6 induced phenomena were also observed in the Group 3 MB cell lines, D283 and D341, including increased pY705-STAT3, drug resistance, IL-6 secretion and IL6R expression. Our study unveiled autocrine IL-6 as a promoter of STAT3 signaling in development of drug resistance, and suggests therapeutic benefits for targeting the IL-6/STAT3 signaling axis in Group 3 MBs.


2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Fengjie Jiang ◽  
Xiaozhu Tang ◽  
Chao Tang ◽  
Zhen Hua ◽  
Mengying Ke ◽  
...  

AbstractN6-methyladenosine (m6A) modification is the most prevalent modification in eukaryotic RNAs while accumulating studies suggest that m6A aberrant expression plays an important role in cancer. HNRNPA2B1 is a m6A reader which binds to nascent RNA and thus affects a perplexing array of RNA metabolism exquisitely. Despite unveiled facets that HNRNPA2B1 is deregulated in several tumors and facilitates tumor growth, a clear role of HNRNPA2B1 in multiple myeloma (MM) remains elusive. Herein, we analyzed the function and the regulatory mechanism of HNRNPA2B1 in MM. We found that HNRNPA2B1 was elevated in MM patients and negatively correlated with favorable prognosis. The depletion of HNRNPA2B1 in MM cells inhibited cell proliferation and induced apoptosis. On the contrary, the overexpression of HNRNPA2B1 promoted cell proliferation in vitro and in vivo. Mechanistic studies revealed that HNRNPA2B1 recognized the m6A sites of ILF3 and enhanced the stability of ILF3 mRNA transcripts, while AKT3 downregulation by siRNA abrogated the cellular proliferation induced by HNRNPA2B1 overexpression. Additionally, the expression of HNRNPA2B1, ILF3 and AKT3 was positively associated with each other in MM tissues tested by immunohistochemistry. In summary, our study highlights that HNRNPA2B1 potentially acts as a therapeutic target of MM through regulating AKT3 expression mediated by ILF3-dependent pattern.


2020 ◽  
Author(s):  
Yue Shen ◽  
HaiXiang Ma ◽  
XiTing Lian ◽  
LeYuan Gu ◽  
Qian Yu ◽  
...  

AbstractSudden unexpected death in epilepsy (SUDEP) is the fatal cause leading to the death of epilepsy patients with anti-epileptic drug resistance. However, the underlying mechanism of SUDEP remains to be elusive. Our previous study demonstrated that enhancement of serotonin (5-HT) synthesis by intraperitoneal (IP) injection of 5-hydroxytryptophan in brain significantly reduced the incidence of seizure-induced respiratory arrest (S-IRA) in DBA/1 mice SUDEP models. Given that 5-HT2A receptor (5-HT2AR) acts an important role in mediating respiration system in brain, we hypothesized that 5-HT2AR is of great significance to modulate S-IRA and SUDEP. To test this hypothesis, we examined whether the decreased incidence S-IRA evoked by either acoustic stimulation or PTZ by blocking 5-HT2AR by administration with ketanserin (KET), a selective antagonist of 5HT2AR, in DBA/1 mice SUDEP models to test the role of 5-HT2AR modulating S-IRA. Our results suggested that the decreased incidence of S-IRA by 5-Hydroxytryptophan (5-HTP), a precursor for central nervous system (CNS) serotonin (5-HT) synthesis, was significantly reversed by IP and intracerebroventricularly (ICV) injection of ketanserin in our models. Thus, our data suggested that 5-HT2AR in the brain may be a potential and specific target to prevent SUDEP.


2017 ◽  
Vol 2017 ◽  
pp. 1-7
Author(s):  
Dong Li ◽  
Aixin Li ◽  
Hairui Zhou ◽  
Xi Wang ◽  
Peng Li ◽  
...  

Drug-induced myopathy (DIM) is a rare side effect; however, the consequence could be fatal. There are few reports to systematically assess the underlying mechanism of DIM. In this study, we curated the comprehensive DIM drug list based on structured labeling products (SPLs) and carried out the analysis based on chemical structure space, drug protein interaction, side effect space, and transcriptomic profiling space. Some key features are enriched from each of analysis. Specifically, the similarity of DIM drugs is more significant than random chance, which shows that the chemical structure could distinguish the DIM-positive drugs from negatives. The cytochrome P450 (CYP) was identified to be shared by DIM drugs, which indicated the important role of metabolism in DIM. Three pathways including pathways in cancer, MAPK signaling pathway, and GnRH signaling pathway enriched based on transcriptomic analysis may explain the underlying mechanism of DIM. Although the DIM is the current focus of the study, the proposed approaches could be applied to other toxicity assessments and facilitate the safety evaluation.


Sign in / Sign up

Export Citation Format

Share Document