1
|
Goyal A, Murkute SL, Bhowmik S, Prasad CP, Mohapatra P. Belling the "cat": Wnt/β-catenin signaling and its significance in future cancer therapies. Biochim Biophys Acta Rev Cancer 2024; 1879:189195. [PMID: 39413855 DOI: 10.1016/j.bbcan.2024.189195] [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: 05/30/2024] [Revised: 09/15/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024]
Abstract
The WNT/β-catenin is among one of the most extensively studied cellular signaling pathways involved in the initiation and progression of several deadly cancers. It is now understood that the WNT/β-catenin signaling, during tumor progression operates in a very complex fashion beyond the earlier assumed simple WNT 'On' or 'Off' mode as it recruits numerous WNT ligands, receptors, transcriptional factors and also cross-talks with other signaling molecules including the noncanonical WNT regulators. WNT/β-catenin signaling molecules are often mutated in different cancers which makes them very challenging to inhibit and sometimes ranks them among the undruggable targets. Furthermore, due to the evolutionary conservation of this pathway, inhibiting WNT/β-catenin has caused significant toxicity in normal cells. These challenges are reflected in clinical trial data, where the use of WNT/β-catenin inhibitors as standalone treatments remains limited. In this review, we have highlighted the crucial functional associations of diverse WNT/β-catenin signaling regulators with cancer progression and the phenotypic switching of tumor cells. Next, we have shed light on the roles of WNT/β-catenin signaling in drug resistance, clonal evolution, tumor heterogeneity, and immune evasion. The present review also focuses on various classes of routine and novel WNT/β-catenin therapeutic regimes while addressing the challenges associated with targeting the regulators of this complex pathway. In the light of multiple case studies on WNT/β-catenin inhibitors, we also highlighted the challenges and opportunities for future clinical trial strategies involving these treatments. Additionally, we have proposed strategies for future WNT/β-catenin-based drug discovery trials, emphasizing the potential of combination therapies and AI/ML-driven prediction approaches. Overall, here we showcased the opportunities, possibilities, and potentialities of WNT/β-catenin signaling modulatory therapeutic regimes as promising precision cancer medicines for the future.
Collapse
Affiliation(s)
- Akansha Goyal
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India
| | - Satyajit Laxman Murkute
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India
| | - Sujoy Bhowmik
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India
| | - Chandra Prakash Prasad
- Department of Medical Oncology Lab, DR BRA-IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Purusottam Mohapatra
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India.
| |
Collapse
|
2
|
Zhou YQ, Cheng XX, He S, Liu SQ, Li YQ, Wei PP, Luo CL, Bei JX. A positive feedback loop between PLD1 and NF-κB signaling promotes tumorigenesis of nasopharyngeal carcinoma. J Genet Genomics 2024; 51:997-1006. [PMID: 38885836 DOI: 10.1016/j.jgg.2024.06.004] [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: 04/17/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024]
Abstract
Phospholipase D (PLD) lipid-signaling enzyme superfamily has been widely implicated in various human malignancies, but its role and underlying mechanism remain unclear in nasopharyngeal carcinoma (NPC). Here, we analyze the expressions of 6 PLD family members between 87 NPC and 10 control samples through transcriptome analysis. Our findings reveal a notable upregulation of PLD1 in both NPC tumors and cell lines, correlating with worse disease-free and overall survival in NPC patients. Functional assays further elucidate the oncogenic role of PLD1, demonstrating its pivotal promotion of critical tumorigenic processes such as cell proliferation and migration in vitro, as well as tumor growth in vivo. Notably, our study uncovers a positive feedback loop between PLD1 and the NF-κB signaling pathway to render NPC progression. Specifically, PLD1 enhances NF-κB activity by facilitating the phosphorylation and nuclear translocation of RELA, which in turn binds to the promoter of PLD1, augmenting its expression. Moreover, RELA overexpression markedly rescues the inhibitory effects in PLD1-depleted NPC cells. Importantly, the application of the PLD1 inhibitor, VU0155069, substantially inhibits NPC tumorigenesis in a patient-derived xenograft model. Together, our findings identify PLD1/NF-κB signaling as a positive feedback loop with promising therapeutic and prognostic potential in NPC.
Collapse
Affiliation(s)
- Ya-Qing Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Xi-Xi Cheng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Shuai He
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Shu-Qiang Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Yi-Qi Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Pan-Pan Wei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Chun-Ling Luo
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China.
| | - Jin-Xin Bei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China; Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China; Department of Medical Oncology, National Cancer Centre of Singapore, Singapore.
| |
Collapse
|
3
|
Lim SH, Lee H, Lee HJ, Kim K, Choi J, Han JM, Min DS. PLD1 is a key player in cancer stemness and chemoresistance: Therapeutic targeting of cross-talk between the PI3K/Akt and Wnt/β-catenin pathways. Exp Mol Med 2024; 56:1479-1487. [PMID: 38945955 PMCID: PMC11297275 DOI: 10.1038/s12276-024-01260-9] [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: 12/29/2023] [Revised: 03/04/2024] [Accepted: 03/19/2024] [Indexed: 07/02/2024] Open
Abstract
The development of chemoresistance is a major challenge in the treatment of several types of cancers in clinical settings. Stemness and chemoresistance are the chief causes of poor clinical outcomes. In this context, we hypothesized that understanding the signaling pathways responsible for chemoresistance in cancers is crucial for the development of novel targeted therapies to overcome drug resistance. Among the aberrantly activated pathways, the PI3K-Akt/Wnt/β-catenin signaling pathway is clinically implicated in malignancies such as colorectal cancer (CRC) and glioblastoma multiforme (GBM). Aberrant dysregulation of phospholipase D (PLD) has been implicated in several malignancies, and oncogenic activation of this pathway facilitates tumor proliferation, stemness, and chemoresistance. Crosstalk involving the PLD and Wnt/β-catenin pathways promotes the progression of CRC and GBM and reduces the sensitivity of cancer cells to standard therapies. Notably, both pathways are tightly regulated and connected at multiple levels by upstream and downstream effectors. Thus, gaining deeper insights into the interactions between these pathways would help researchers discover unique therapeutic targets for the management of drug-resistant cancers. Here, we review the molecular mechanisms by which PLD signaling stimulates stemness and chemoresistance in CRC and GBM. Thus, the current review aims to address the importance of PLD as a central player coordinating cross-talk between the PI3K/Akt and Wnt/β-catenin pathways and proposes the possibility of targeting these pathways to improve cancer therapy and overcome drug resistance.
Collapse
Affiliation(s)
- Seong Hun Lim
- Department of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Hyesung Lee
- Department of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Hyun Ji Lee
- Department of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Kuglae Kim
- Department of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Junjeong Choi
- Department of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Jung Min Han
- Department of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
- POSTECH Biotech Center, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Do Sik Min
- Department of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea.
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea.
| |
Collapse
|
4
|
Wang H, Zhao Y, Pan Y, Yang A, Li C, Wang S, Dong Z, Li M, Wang S, Zhang Z, Zhu Y, Zhang D, Sun G. Inhibition of phospholipase D1 ameliorates hepatocyte steatosis and non-alcoholic fatty liver disease. JHEP Rep 2023; 5:100726. [PMID: 37138676 PMCID: PMC10149370 DOI: 10.1016/j.jhepr.2023.100726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 02/14/2023] [Accepted: 02/27/2023] [Indexed: 05/05/2023] Open
Abstract
Background & Aims Phospholipase D1 (PLD1), a phosphatidylcholine-hydrolysing enzyme, is involved in cellular lipid metabolism. However, its involvement in hepatocyte lipid metabolism and consequently non-alcoholic fatty liver disease (NAFLD) has not been explicitly explored. Methods NAFLD was induced in hepatocyte-specific Pld1 knockout (Pld1(H)-KO) and littermate Pld1 flox/flox (Pld1-Flox) control mice feeding a high-fat diet (HFD) for 20 wk. Changes of the lipid composition in the liver were compared. Alpha mouse liver 12 (AML12) cells and mouse primary hepatocytes were incubated with oleic acid or sodium palmitate in vitro to explore the role of PLD1 in the development of hepatic steatosis. Hepatic PLD1 expression was evaluated in liver biopsy samples in patients with NAFLD. Results PLD1 expression levels were increased in the hepatocytes of patients with NAFLD and HFD-fed mice. Compared with Pld1-Flox mice, Pld1(H)-KO mice exhibited decreased plasma glucose and lipid levels as well as lipid accumulation in liver tissues after HFD feeding. Transcriptomic analysis showed that hepatocyte-specific deficiency of PLD1 decreased Cd36 expression in steatosis liver tissues, which was confirmed at the protein and gene levels. In vitro, specific inhibition of PLD1 with VU0155069 or VU0359595 decreased CD36 expression and lipid accumulation in oleic acid- or sodium palmitate-treated AML12 cells or primary hepatocytes. Inhibition of hepatocyte PLD1 significantly altered lipid composition, especially phosphatidic acid and lysophosphatidic acid levels in liver tissues with hepatic steatosis. Furthermore, phosphatidic acid, the downstream product of PLD1, increased the expression levels of CD36 in AML12 cells, which was reversed by a PPARγ antagonist. Conclusions Hepatocyte-specific Pld1 deficiency ameliorates lipid accumulation and NAFLD development by inhibiting the PPARγ/CD36 pathway. PLD1 may be a new target for the treatment of NAFLD. Impact and implications The involvement of PLD1 in hepatocyte lipid metabolism and NAFLD has not been explicitly explored. In this study, we found that the inhibition of hepatocyte PLD1 exerted potent protective effects against HFD-induced NAFLD, which were attributable to a reduction in PPARγ/CD36 pathway-mediated lipid accumulation in hepatocytes. Targeting hepatocyte PLD1 may be a new target for the treatment of NAFLD.
Collapse
Affiliation(s)
- Huan Wang
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
- Beijing Clinical Research Institute, Beijing, China
| | - Yushang Zhao
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
- Beijing Clinical Research Institute, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Beijing Laboratory of Oral Health, Capital Medical University School of Basic Medicine, Beijing, China
| | - Yuhualei Pan
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
- Beijing Clinical Research Institute, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Beijing Laboratory of Oral Health, Capital Medical University School of Basic Medicine, Beijing, China
| | - Aiting Yang
- Beijing Clinical Research Institute, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Changying Li
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
- Beijing Clinical Research Institute, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Beijing Laboratory of Oral Health, Capital Medical University School of Basic Medicine, Beijing, China
| | - Song Wang
- Beijing Clinical Research Institute, Beijing, China
| | - Zhao Dong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Mengyi Li
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Songlin Wang
- Beijing Laboratory of Oral Health, Capital Medical University School of Basic Medicine, Beijing, China
| | - Zhongtao Zhang
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yanbing Zhu
- Beijing Clinical Research Institute, Beijing, China
- Corresponding author. Address: Capital Medical University Affiliated Beijing Friendship Hospital, 95 Yongan Road, Xicheng District, Beijing 100050, China. Tel.: (8610)63139309, fax: (8610)63139421.
| | - Dong Zhang
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
- Beijing Clinical Research Institute, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Beijing Laboratory of Oral Health, Capital Medical University School of Basic Medicine, Beijing, China
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Corresponding author. Address: Capital Medical University Affiliated Beijing Friendship Hospital, 95 Yongan Road, Xicheng District, Beijing 100050, China. Tel.: (8610)63139309, fax: (8610)63139421.
| | - Guangyong Sun
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
- Beijing Clinical Research Institute, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Beijing Laboratory of Oral Health, Capital Medical University School of Basic Medicine, Beijing, China
- Corresponding author. Address: Capital Medical University Affiliated Beijing Friendship Hospital, 95 Yongan Road, Xicheng District, Beijing 100050, China. Tel.: (8610)63139309, fax: (8610)63139421.
| |
Collapse
|
5
|
Xu X, Wang J, Du S, Shen X, Lian J, Zhou J, Wang M, Feng W, Lv Z, Zhu J, Jin L, Sun H, Wu L, Wang X, Qiu H, Wang W, Teng H, Wang Y, Huang Z. Yes-associated protein regulates glutamate homeostasis through promoting the expression of excitatory amino acid transporter-2 in astrocytes via β-catenin signaling. Glia 2023; 71:1197-1216. [PMID: 36617748 DOI: 10.1002/glia.24332] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/07/2022] [Accepted: 12/21/2022] [Indexed: 01/10/2023]
Abstract
The homeostasis of glutamate is mainly regulated by the excitatory amino acid transporters (EAATs), especially by EAAT2 in astrocytes. Excessive glutamate in the synaptic cleft caused by dysfunction or dysregulation of EAAT2 can lead to excitotoxicity, neuronal death and cognitive dysfunction. However, it remains unclear about the detailed regulation mechanism of expression and function of astrocytic EAAT2. In this study, first, we found increased neuronal death and impairment of cognitive function in YAPGFAP -CKO mice (conditionally knock out Yes-associated protein [YAP] in astrocytes), and identified EAAT2 as a downstream target of YAP through RNA sequencing. Second, the expression of EAAT2 was decreased in cultured YAP-/- astrocytes and the hippocampus of YAPGFAP -CKO mice, and glutamate uptake was reduced in YAP-/- astrocytes, but increased in YAP-upregulated astrocytes. Third, further investigation of the mechanism showed that the mRNA and protein levels of β-catenin were decreased in YAP-/- astrocytes and increased in YAP-upregulated astrocytes. Wnt3a activated YAP signaling and up-regulated EAAT2 through β-catenin. Furthermore, over-expression or activation of β-catenin partially restored the downregulation of EAAT2, the impairment of glutamate uptake, neuronal death and cognitive decline that caused by YAP deletion. Finally, activation of EAAT2 also rescued neuronal death and cognitive decline in YAPGFAP -CKO mice. Taken together, our study identifies an unrecognized role of YAP signaling in the regulation of glutamate homeostasis through the β-catenin/EAAT2 pathway in astrocytes, which may provide novel insights into the pathogenesis of brain diseases that closely related to the dysfunction or dysregulation of EAAT2, and promote the development of clinical strategy.
Collapse
Affiliation(s)
- Xingxing Xu
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jiaojiao Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Siyu Du
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Xiya Shen
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jiashu Lian
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian Zhou
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mianxian Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenjin Feng
- Zhejiang Sinogen Medical Equipment Co., Ltd., Wenzhou, China
| | - Zhaoting Lv
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Junzhe Zhu
- School of the First Clinical Medical Sciences (School of Information and Engineering), Wenzhou Medical University, Wenzhou, China
| | - Lingting Jin
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Huankun Sun
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Lihao Wu
- School of the First Clinical Medical Sciences (School of Information and Engineering), Wenzhou Medical University, Wenzhou, China
| | - Xiaoning Wang
- School of the First Clinical Medical Sciences (School of Information and Engineering), Wenzhou Medical University, Wenzhou, China
| | - Haoyu Qiu
- School of the First Clinical Medical Sciences (School of Information and Engineering), Wenzhou Medical University, Wenzhou, China
| | - Wei Wang
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Honglin Teng
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ying Wang
- Clinical Research Center, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihui Huang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
- Department of Orthopedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
6
|
Inhibition of phospholipase D1 induces immunogenic cell death and potentiates cancer immunotherapy in colorectal cancer. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1563-1576. [PMID: 36131027 PMCID: PMC9535023 DOI: 10.1038/s12276-022-00853-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/07/2022] [Accepted: 07/21/2022] [Indexed: 12/02/2022]
Abstract
Phospholipase D (PLD) is a potential therapeutic target against cancer. However, the contribution of PLD inhibition to the antitumor response remains unknown. We developed a potent and selective PLD1 inhibitor based on computer-aided drug design. The inhibitor enhanced apoptosis in colorectal cancer (CRC) cells but not in normal colonic cells, and in vitro cardiotoxicity was not observed. The inhibitor downregulated the Wnt/β-catenin signaling pathway and reduced the migration, invasion, and self-renewal capacity of CRC cells. In cancer, therapeutic engagement of immunogenic cell death (ICD) leads to more effective responses by eliciting the antitumor immunity of T cells. The CRC cells treated with the inhibitor showed hallmarks of ICD, including downregulation of “do not eat-me” signals (CD24, CD47, programmed cell death ligand 1 [PD-L1]), upregulation of “eat-me” signal (calreticulin), release of high-mobility group Box 1, and ATP. PLD1 inhibition subsequently enhanced the phagocytosis of cancer cells by macrophages through the surface expression of costimulatory molecules; as a result, the cancer cells were more susceptible to cytotoxic T-cell-mediated killing. Moreover, PLD1 inhibition attenuated colitis-associated CRC and orthotopically injected tumors, probably by controlling multiple pathways, including Wnt signaling, phagocytosis checkpoints, and immune signaling. Furthermore, combination therapy with a PLD1 inhibitor and an anti-PD-L1 antibody further enhanced tumor regression via immune activation in the tumor environment. Collectively, in this study, PLD1 was identified as a critical regulator of the tumor microenvironment in colorectal cancer, suggesting the potential of PLD1 inhibitors for cancer immunotherapy based on ICD and immune activation. PLD1 inhibitors may act as promising immune modulators in antitumor treatment via ICD. A novel drug that can inhibit an enzyme involved in colorectal cancer progression shows promise in trials on mouse models. The phospholipase D1 (PLD1) enzyme reinforces a critical signaling pathway that promotes cancer progression and drug resistance. Using computer-aided drug design, South Korean researchers led by Do Sik Min and Gyoonhee Han at Yonsei University in Incheon and Seoul, respectively, have developed a drug that specifically binds to and inhibits PLD1. In trials, the researchers observed downregulation of PLD1’s associated signaling pathway, and reductions in the ability of colorectal cancer cells to migrate, invade and replicate. The drug suppressed the cancer cells’ “don’t-eat-me” signals and upregulated “eat-me” signals, triggering improved responses from the immune system. The drug was even more effective when used in combination with an immunotherapy agent.
Collapse
|
7
|
Huang Y, Zhang X, PengWang, Li Y, Yao J. Identification of hub genes and pathways in colitis-associated colon cancer by integrated bioinformatic analysis. BMC Genom Data 2022; 23:48. [PMID: 35733095 PMCID: PMC9219145 DOI: 10.1186/s12863-022-01065-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/13/2022] [Indexed: 12/25/2022] Open
Abstract
Background Colitis-associated colon cancer (CAC) patients have a younger age of onset, more multiple lesions and invasive tumors than sporadic colon cancer patients. Early detection of CAC using endoscopy is challenging, and the incidence of septal colon cancer remains high. Therefore, identifying biomarkers that can predict the tumorigenesis of CAC is in urgent need. Results A total of 275 DEGs were identified in CAC. IGF1, BMP4, SPP1, APOB, CCND1, CD44, PTGS2, CFTR, BMP2, KLF4, and TLR2 were identified as hub DEGs, which were significantly enriched in the PI3K-Akt pathway, stem cell pluripotency regulation, focal adhesion, Hippo signaling, and AMPK signaling pathways. Sankey diagram showed that the genes of both the PI3K-AKT signaling and focal adhesion pathways were upregulated (e.g., SPP1, CD44, TLR2, CCND1, and IGF1), and upregulated genes were predicted to be regulated by the crucial miRNAs: hsa-mir-16-5p, hsa-mir-1-3p, et al. Hub gene-TFs network revealed FOXC1 as a core transcription factor. In ulcerative colitis (UC) patients, KLF4, CFTR, BMP2, TLR2 showed significantly lower expression in UC-associated cancer. BMP4 and IGF1 showed higher expression in UC-Ca compared to nonneoplastic mucosa. Survival analysis showed that the differential expression of SPP1, CFRT, and KLF4 were associated with poor prognosis in colon cancer. Conclusion Our study provides novel insights into the mechanism underlying the development of CAC. The hub genes and signaling pathways may contribute to the prevention, diagnosis and treatment of CAC. Supplementary Information The online version contains supplementary material available at 10.1186/s12863-022-01065-7.
Collapse
Affiliation(s)
- Yongming Huang
- Department of General Surgery, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, 272000, Shandong Province, China
| | - Xiaoyuan Zhang
- Key Laboratory of Precision Oncology in Universities of Shandong, Department of Pathology and Institute of Precision Medicine, Taibai Lake New Area, Jining Medical University, 133 Hehua Road, Jining, 272067, Shandong Province, China
| | - PengWang
- Department of General Surgery, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, 272000, Shandong Province, China
| | - Yansen Li
- Department of General Surgery, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, 272000, Shandong Province, China
| | - Jie Yao
- Department of Oncology, Jining Hospital of Traditional Chinese Medicine, 3 Huancheng North Road, Jining, 272000, Shandong Province, China.
| |
Collapse
|
8
|
Wnt/β-catenin signalling: function, biological mechanisms, and therapeutic opportunities. Signal Transduct Target Ther 2022; 7:3. [PMID: 34980884 PMCID: PMC8724284 DOI: 10.1038/s41392-021-00762-6] [Citation(s) in RCA: 699] [Impact Index Per Article: 349.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/28/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023] Open
Abstract
The Wnt/β-catenin pathway comprises a family of proteins that play critical roles in embryonic development and adult tissue homeostasis. The deregulation of Wnt/β-catenin signalling often leads to various serious diseases, including cancer and non-cancer diseases. Although many articles have reviewed Wnt/β-catenin from various aspects, a systematic review encompassing the origin, composition, function, and clinical trials of the Wnt/β-catenin signalling pathway in tumour and diseases is lacking. In this article, we comprehensively review the Wnt/β-catenin pathway from the above five aspects in combination with the latest research. Finally, we propose challenges and opportunities for the development of small-molecular compounds targeting the Wnt signalling pathway in disease treatment.
Collapse
|
9
|
Xu Z, Qu H, Ren Y, Gong Z, Ri HJ, Chen X. An Update on the Potential Roles of E2F Family Members in Colorectal Cancer. Cancer Manag Res 2021; 13:5509-5521. [PMID: 34276228 PMCID: PMC8277564 DOI: 10.2147/cmar.s320193] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/01/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is a major health burden worldwide, and thus, optimised diagnosis and treatments are imperative. E2F transcription factors (E2Fs) are a family of transcription factors consisting of eight genes, contributing to the oncogenesis and development of CRC. Importantly, E2Fs control not only the cell cycle but also apoptosis, senescence, DNA damage response, and drug resistance by interacting with multiple signaling pathways. However, the specific functions and intricate machinery of these eight E2Fs in human CRC remain unclear in many respects. Evidence on E2Fs and CRC has been scattered on the related regulatory genes, microRNAs (miRNAs), and competing endogenous RNAs (ceRNAs). Accordingly, some drugs targeting E2Fs have been transferred from preclinical to clinical application. Herein, we have systemically reviewed the current literature on the roles of various E2Fs in CRC with the purpose of providing possible clinical implications for patient diagnosis and prognosis and future treatment strategy design, thereby furthering the understanding of the E2Fs.
Collapse
Affiliation(s)
- ZhaoHui Xu
- Department of Hernia and Colorectal Surgery, The Second Hospital of Dalian Medical University, Dalian, 116023, People's Republic of China
| | - Hui Qu
- Department of Hernia and Colorectal Surgery, The Second Hospital of Dalian Medical University, Dalian, 116023, People's Republic of China
| | - YanYing Ren
- Department of Hernia and Colorectal Surgery, The Second Hospital of Dalian Medical University, Dalian, 116023, People's Republic of China
| | - ZeZhong Gong
- Department of Hernia and Colorectal Surgery, The Second Hospital of Dalian Medical University, Dalian, 116023, People's Republic of China
| | - Hyok Ju Ri
- Department of Hernia and Colorectal Surgery, The Second Hospital of Dalian Medical University, Dalian, 116023, People's Republic of China
| | - Xin Chen
- Department of Hernia and Colorectal Surgery, The Second Hospital of Dalian Medical University, Dalian, 116023, People's Republic of China
| |
Collapse
|
10
|
Ramaiah MJ, Kumar KR. mTOR-Rictor-EGFR axis in oncogenesis and diagnosis of glioblastoma multiforme. Mol Biol Rep 2021; 48:4813-4835. [PMID: 34132942 DOI: 10.1007/s11033-021-06462-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 06/01/2021] [Indexed: 12/12/2022]
Abstract
Glioblastoma multiforme (GBM) is one of the aggressive brain cancers with patients having less survival period upto 12-15 months. Mammalian target of rapamycin (mTOR) is a serine/threonine kinase, belongs to the phosphatidylinositol 3-kinases (PI3K) pathway and is involved in various cellular processes of cancer cells. Cancer metabolism is regulated by mTOR and its components. mTOR forms two complexes as mTORC1 and mTORC2. Studies have identified the key component of the mTORC2 complex, Rapamycin-insensitive companion of mammalian target of rapamycin (Rictor) plays a prominent role in the regulation of cancer cell proliferation and metabolism. Apart, growth factor receptor signaling such as epidermal growth factor signaling mediated by epidermal growth factor receptor (EGFR) regulates cancer-related processes. In EGFR signaling various other signaling cascades such as phosphatidyl-inositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR pathway) and Ras/Raf/mitogen-activated protein kinase/ERK kinase (MEK)/extracellular-signal-regulated kinase (ERK) -dependent signaling cross-talk each other. From various studies about GBM, it is very well established that Rictor and EGFR mediated signaling pathways majorly playing a pivotal role in chemoresistance and tumor aggressiveness. Recent studies have shown that non-coding RNAs such as microRNAs (miRs) and long non-coding RNAs (lncRNAs) regulate the EGFR and Rictor and sensitize the cells towards chemotherapeutic agents. Thus, understanding of microRNA mediated regulation of EGFR and Rictor will help in cancer prevention and management as well as a future therapy.
Collapse
Affiliation(s)
- M Janaki Ramaiah
- Functional Genomics and Disease Biology Laboratory, School of Chemical and Biotechnology (SCBT), SASTRA Deemed University, Tirumalaisamudram, Thanjavur, 613401, Tamil Nadu, India.
- School of Chemical and Biotechnology (SCBT), SASTRA Deemed University, Tirumalaisamudram, Thanjavur, 613401, Tamil Nadu, India.
| | - K Rohil Kumar
- Functional Genomics and Disease Biology Laboratory, School of Chemical and Biotechnology (SCBT), SASTRA Deemed University, Tirumalaisamudram, Thanjavur, 613401, Tamil Nadu, India
| |
Collapse
|
11
|
Phospholipase Signaling in Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 33983572 DOI: 10.1007/978-981-32-9620-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Breast cancer progression results from subversion of multiple intra- or intercellular signaling pathways in normal mammary tissues and their microenvironment, which have an impact on cell differentiation, proliferation, migration, and angiogenesis. Phospholipases (PLC, PLD and PLA) are essential mediators of intra- and intercellular signaling. They hydrolyze phospholipids, which are major components of cell membrane that can generate many bioactive lipid mediators, such as diacylglycerol, phosphatidic acid, lysophosphatidic acid, and arachidonic acid. Enzymatic processing of phospholipids by phospholipases converts these molecules into lipid mediators that regulate multiple cellular processes, which in turn can promote breast cancer progression. Thus, dysregulation of phospholipases contributes to a number of human diseases, including cancer. This review describes how phospholipases regulate multiple cancer-associated cellular processes, and the interplay among different phospholipases in breast cancer. A thorough understanding of the breast cancer-associated signaling networks of phospholipases is necessary to determine whether these enzymes are potential targets for innovative therapeutic strategies.
Collapse
|
12
|
Bjørnetrø T, Steffensen LA, Vestad B, Brusletto BS, Olstad OK, Trøseid AM, Aass HCD, Haug KBF, Llorente A, Bøe SO, Lång A, Samiappan R, Redalen KR, Øvstebø R, Ree AH. Uptake of circulating extracellular vesicles from rectal cancer patients and differential responses by human monocyte cultures. FEBS Open Bio 2021; 11:724-740. [PMID: 33512765 PMCID: PMC7931235 DOI: 10.1002/2211-5463.13098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/13/2021] [Accepted: 01/26/2021] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EVs) released by tumor cells can directly or indirectly modulate the phenotype and function of the immune cells of the microenvironment locally or at distant sites. The uptake of circulating EVs and the responses by human monocytes in vitro may provide new insights into the underlying biology of the invasive and metastatic processes in cancer. Although a mixed population of vesicles is obtained with most isolation techniques, we predominantly isolated exosomes (small EVs) and microvesicles (medium EVs) from the SW480 colorectal cancer cell line (established from a primary adenocarcinoma of the colon) by sequential centrifugation and ultrafiltration, and plasma EVs were prepared from 22 patients with rectal adenoma polyps or invasive adenocarcinoma by size‐exclusion chromatography. The EVs were thoroughly characterized. The uptake of SW480 EVs was analyzed, and small SW480 EVs were observed to be more potent than medium SW480 EVs in inducing monocyte secretion of cytokines. The plasma EVs were also internalized by monocytes; however, their cytokine‐releasing potency was lower than that of the cell line‐derived vesicles. The transcriptional changes in the monocytes highlighted differences between adenoma and adenocarcinoma patient EVs in their ability to regulate biological functions, whereas the most intriguing changes were found in monocytes receiving EVs from patients with metastatic compared with localized cancer.
Collapse
Affiliation(s)
- Tonje Bjørnetrø
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Norway
| | - Lilly Alice Steffensen
- The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Norway
| | - Beate Vestad
- The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Norway
| | - Berit Sletbakk Brusletto
- The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Norway
| | - Ole Kristoffer Olstad
- The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Norway
| | - Anne-Marie Trøseid
- The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Norway
| | | | - Kari Bente Foss Haug
- The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Norway
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute of Cancer Research, Oslo University Hospital, Norway
| | - Stig Ove Bøe
- Department of Medical Biochemistry, Oslo University Hospital, Norway
| | - Anna Lång
- Department of Medical Biochemistry, Oslo University Hospital, Norway
| | | | - Kathrine Røe Redalen
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway.,Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Reidun Øvstebø
- The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Norway
| | - Anne Hansen Ree
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Norway
| |
Collapse
|
13
|
Kang DW, Hwang WC, Noh YN, Che X, Lee SH, Jang Y, Choi KY, Choi JY, Min DS. Deletion of phospholipase D1 decreases bone mass and increases fat mass via modulation of Runx2, β-catenin-osteoprotegerin, PPAR-γ and C/EBPα signaling axis. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166084. [PMID: 33497821 DOI: 10.1016/j.bbadis.2021.166084] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/12/2021] [Accepted: 01/20/2021] [Indexed: 11/16/2022]
Abstract
In osteoporosis, mesenchymal stem cells (MSCs) prefer to differentiate into adipocytes at the expense of osteoblasts. Although the balance between adipogenesis and osteogenesis has been closely examined, the mechanism of commitment determination switch is unknown. Here we demonstrate that phospholipase D1 (PLD1) plays a key switch in determining the balance between bone and fat mass. Ablation of Pld1 reduced bone mass but increased fat in mice. Mechanistically, Pld1/- MSCs inhibited osteoblast differentiaion with diminished Runx2 expression, while osteoclast differentiation was accelerated in Pld1-/- bone marrow-derived macrophages. Pld1-/- osteoblasts showed decreased expression of osteogenic makers. Increased number and resorption activity of osteoclasts in Pld1-/- mice were corroborated with upregulation of osteoclastogenic markers. Moreover, Pld1-/- osteoblasts reduced β-catenin mediated-osteoprotegerin (OPG) with increased RANKL/OPG ratio which resulted in accelerated osteoclast differentiation. Thus, low bone mass with upregulated osteoclasts could be due to the contribution of both osteoblasts and osteoclasts during bone remodeling. Moreover, ablation of Pld1 further increased bone loss in ovariectomized mice, suggesting that PLD1 is a negative regulator of osteoclastogenesis. Furthermore, loss of PLD1 increased adipogenesis, body fat mass, and hepatic steatosis along with upregulation of PPAR-γ and C/EBPα. Interestingly, adipocyte-specific Pld1 transgenic mice rescued the compromised phenotypes of fat mass and adipogenesis in Pld1 knockout mice. Collectively, PLD1 regulated the bifurcating pathways of mesenchymal cell lineage into increased osteogenesis and decreased adipogenesis, which uncovered a previously unrecognized role of PLD1 in homeostasis between bone and fat mass.
Collapse
Affiliation(s)
- Dong Woo Kang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 609-735, Republic of Korea; Institute for Innovative Cancer Research, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Won Chan Hwang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 609-735, Republic of Korea; College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
| | - Yu Na Noh
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 609-735, Republic of Korea
| | - Xiangguo Che
- Department of Biochemistry and Cell Biology, Korea Mouse Phenotyping Center, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Soung-Hoon Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Younghoon Jang
- Department of Biology and Chemistry, Changwon National University, Changwon, Republic of Korea
| | - Kang-Yell Choi
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Je-Yong Choi
- Department of Biochemistry and Cell Biology, Korea Mouse Phenotyping Center, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Do Sik Min
- College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea.
| |
Collapse
|
14
|
Auclair N, Sané AT, Delvin E, Spahis S, Levy E. Phospholipase D as a Potential Modulator of Metabolic Syndrome: Impact of Functional Foods. Antioxid Redox Signal 2021; 34:252-278. [PMID: 32586106 DOI: 10.1089/ars.2020.8081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Significance: Cardiometabolic disorders (CMD) are composed of a plethora of metabolic dysfunctions such as dyslipidemia, nonalcoholic fatty liver disease, insulin resistance, and hypertension. The development of these disorders is highly linked to inflammation and oxidative stress (OxS), two metabolic states closely related to physiological and pathological conditions. Given the drastically rising CMD prevalence, the discovery of new therapeutic targets/novel nutritional approaches is of utmost importance. Recent Advances: The tremendous progress in methods/technologies and animal modeling has allowed the clarification of phospholipase D (PLD) critical roles in multiple cellular processes, whether directly or indirectly via phosphatidic acid, the lipid product mediating signaling functions. In view of its multiple features and implications in various diseases, PLD has emerged as a drug target. Critical Issues: Although insulin stimulates PLD activity and, in turn, PLD regulates insulin signaling, the impact of the two important PLD isoforms on the metabolic syndrome components remains vague. Therefore, after outlining PLD1/PLD2 characteristics and functions, their role in inflammation, OxS, and CMD has been analyzed and critically reported in the present exhaustive review. The influence of functional foods and nutrients in the regulation of PLD has also been examined. Future Directions: Available evidence supports the implication of PLD in CMD, but only few studies emphasize its mechanisms of action and specific regulation by nutraceutical compounds. Therefore, additional investigations are first needed to clarify the functional role of nutraceutics and, second, to elucidate whether targeting PLDs with food compounds represents an appropriate therapeutic strategy to treat CMD. Antioxid. Redox Signal. 34, 252-278.
Collapse
Affiliation(s)
- Nickolas Auclair
- Research Center, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada.,Department of Pharmacology & Physiology and Université de Montréal, Montreal, Quebec, Canada
| | - Alain T Sané
- Research Center, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Edgard Delvin
- Research Center, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Schohraya Spahis
- Research Center, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada.,Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Emile Levy
- Research Center, CHU Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada.,Department of Pharmacology & Physiology and Université de Montréal, Montreal, Quebec, Canada.,Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
15
|
Li YF, Tsai WC, Chou CH, Huang LC, Huang SM, Hueng DY, Tsai CK. CKAP2L Knockdown Exerts Antitumor Effects by Increasing miR-4496 in Glioblastoma Cell Lines. Int J Mol Sci 2020; 22:ijms22010197. [PMID: 33375517 PMCID: PMC7796349 DOI: 10.3390/ijms22010197] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 02/06/2023] Open
Abstract
Despite advances in the diagnosis and treatment of the central nervous system malignancy glioma, overall survival remains poor. Cytoskeleton-associated protein 2-like (CKAP2L), which plays key roles in neural progenitor cell division, has also been linked to poor prognosis in lung cancer. In the present study, we investigated the role of CKAP2L in glioma. From bioinformatics analyses of datasets from The Cancer Gene Atlas and the Chinese Glioma Genome Atlas, we found that CKAP2L expression correlates with tumor grade and overall survival. Gene set enrichment analysis (GSEA) showed that MITOTIC_SPINDLE, G2M_CHECKPOINT, and E2F_TARGETS are crucially enriched phenotypes associated with high CKAP2L expression. Using U87MG, U118MG, and LNZ308 human glioma cells, we confirmed that CKAP2L knockdown with siCKAP2L inhibits glioma cell proliferation, migration, invasion, and epithelial-mesenchymal transition. Interestingly, CKAP2L knockdown also induced cell cycle arrest at G2/M phase, which is consistent with the GSEA finding. Finally, we observed that CKAP2L knockdown led to significant increases in miR-4496. Treating cells with exogenous miR-4496 mimicked the effect of CKAP2L knockdown, and the effects of CKAP2L knockdown could be suppressed by miR-4496 inhibition. These findings suggest that CKAP2L is a vital regulator of miR-4496 activity and that CKAP2L is a potentially useful prognostic marker in glioma.
Collapse
Affiliation(s)
- Yao-Feng Li
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-F.L.); (W.-C.T.)
| | - Wen-Chiuan Tsai
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-F.L.); (W.-C.T.)
| | - Chung-Hsing Chou
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan;
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan; (S.-M.H.); (D.-Y.H.)
| | - Li-Chun Huang
- Department of Biochemistry, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Shih-Ming Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan; (S.-M.H.); (D.-Y.H.)
- Department of Biochemistry, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Dueng-Yuan Hueng
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan; (S.-M.H.); (D.-Y.H.)
- Department of Biochemistry, National Defense Medical Center, Taipei 11490, Taiwan;
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan
| | - Chia-Kuang Tsai
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan;
- Correspondence:
| |
Collapse
|
16
|
Phospholipase D1 and D2 Synergistically Regulate Thrombus Formation. Int J Mol Sci 2020; 21:ijms21186954. [PMID: 32971863 PMCID: PMC7555624 DOI: 10.3390/ijms21186954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/17/2020] [Accepted: 09/20/2020] [Indexed: 11/16/2022] Open
Abstract
Previously, we reported that phospholipase D1 (PLD1) and PLD2 inhibition by selective PLD1 and PLD2 inhibitors could prevent platelet aggregation in humans, but not in mice. Moreover, only the PLD1 inhibitor, but not PLD2 inhibitor, could effectively prevent thrombus formation in mice, indicating that PLD might play different roles in platelet function in humans and mice. Although PLD1 and PLD2 were reported to be implicated in thrombotic events, the role of PLD in mice remains not completely clear. Here, we investigated the role of PLD1 and PLD2 in acute pulmonary thrombosis and transient middle cerebral artery occlusion-induced brain injury in mice. The data revealed that inhibition of PLD1, but not of PLD2, could partially prevent pulmonary thrombosis-induced death. Moreover, concurrent PLD1 and PLD2 inhibition could considerably increase survival rate. Likewise, inhibition of PLD1, but not PLD2, partially improved ischemic stroke and concurrent inhibition of PLD1, and PLD2 exhibited a relatively better protection against ischemic stroke, as evidenced by the infarct size, brain edema, modified neurological severity score, rotarod test, and the open field test. In conclusion, PLD1 might play a more important role than PLD2, and both PLD1 and PLD2 could act synergistically or have partially redundant functions in regulating thrombosis-relevant events.
Collapse
|
17
|
Kang DW, Hwang WC, Noh YN, Park KS, Min DS. Phospholipase D1 inhibition sensitizes glioblastoma to temozolomide and suppresses its tumorigenicity. J Pathol 2020; 252:304-316. [PMID: 32725633 PMCID: PMC7693208 DOI: 10.1002/path.5519] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/12/2020] [Accepted: 07/24/2020] [Indexed: 12/15/2022]
Abstract
Resistance of glioblastoma to the chemotherapeutic compound temozolomide is associated with the presence of glioblastoma stem cells in glioblastoma and is a key obstacle for the poor prognosis of glioblastoma. Here, we show that phospholipase D1 is elevated in CD44High glioblastoma stem cells and in glioblastoma, especially recurring glioblastoma. Phospholipase D1 elevation positively correlated with the level of CD44 and poor prognosis in glioblastoma patients. Temozolomide significantly upregulated the expression of phospholipase D1 in the low and moderate CD44 populations of glioblastoma stem cells, but not in the CD44High population in which phospholipase D1 is highly expressed. Phospholipase D1 conferred resistance to temozolomide in CD44High glioblastoma stem cells and increased their self‐renewal capacity and maintenance. Phospholipase D1 expression significantly correlated with levels of temozolomide resistance factors, which were suppressed by microRNA‐320a and ‐4496 induced by phospholipase D1 inhibition. Genetic and pharmacological targeting of phospholipase D1 attenuated glioblastoma stem cell‐derived intracranial tumors of glioblastoma using the microRNAs, and improved survival. Treatment solely with temozolomide produced no benefits on the glioblastoma, whereas in combination, phospholipase D1 inhibition sensitized glioblastoma stem cells to temozolomide and reduced glioblastoma tumorigenesis. Together, these findings indicate that phospholipase D1 inhibition might overcome resistance to temozolomide and represents a potential treatment strategy for glioblastoma. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Dong Woo Kang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Won Chan Hwang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea.,College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Yu Na Noh
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Kang Seo Park
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea.,Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Do Sik Min
- College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| |
Collapse
|
18
|
Kang DW, Hwang WC, Noh YN, Kang Y, Jang Y, Kim JA, Min DS. Phospholipase D1 is upregulated by vorinostat and confers resistance to vorinostat in glioblastoma. J Cell Physiol 2020; 236:549-560. [PMID: 32869317 PMCID: PMC7692931 DOI: 10.1002/jcp.29882] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/25/2022]
Abstract
Glioblastoma (GBM) is an aggressive brain tumor and drug resistance remains a major barrier for therapeutics. Epigenetic alterations are implicated in GBM pathogenesis, and epigenetic modulators including histone deacetylase (HDAC) inhibitors are exploited as promising anticancer therapies. Here, we demonstrate that phospholipase D1 (PLD1) is a transcriptional target of HDAC inhibitors and confers resistance to HDAC inhibitor in GBM. Treatment of vorinostat upregulates PLD1 through PKCζ‐Sp1 axis. Vorinostat induces dynamic changes in the chromatin structure and transcriptional machinery associated with PLD1 promoter region. Cotreatment of vorinostat with PLD1 inhibitor further attenuates invasion, angiogenesis, colony‐forming capacity, and self‐renewal capacity, compared with those of either treatment. PLD1 inhibitor overcomes resistance to vorinostat in GBM cells intracranial GBM tumors. Our finding provides new insight into the role of PLD1 as a target of resistance to vorinostat, and PLD1 inhibitor might provide the basis for therapeutic combinations with improved efficacy of HDAC inhibitor.
Collapse
Affiliation(s)
- Dong Woo Kang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Won Chan Hwang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea.,College of Pharmacy, Yonsei University, Incheon, South Korea
| | - Yu Na Noh
- Institute for Innovative Cancer Research, Biomedical Research Center, Asan Medical Center, Seoul, Republic of Korea
| | - Youra Kang
- College of Pharmacy, Yeungnam University, Gyeongsan, South Korea
| | - Younghoon Jang
- Department of Biology and Chemistry, Changwon National University, Changwon, Korea
| | - Jung-Ae Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, South Korea
| | - Do Sik Min
- College of Pharmacy, Yonsei University, Incheon, South Korea
| |
Collapse
|
19
|
Chen X, Wang C, Jiang Y, Wang Q, Tao Y, Zhang H, Zhao Y, Hu Y, Li C, Ye D, Liu D, Jiang W, Chin EY, Chen S, Liu Y, Wang M, Liu S, Zhang X. Bcl-3 promotes Wnt signaling by maintaining the acetylation of β-catenin at lysine 49 in colorectal cancer. Signal Transduct Target Ther 2020; 5:52. [PMID: 32355204 PMCID: PMC7193563 DOI: 10.1038/s41392-020-0138-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/24/2019] [Accepted: 02/18/2020] [Indexed: 11/09/2022] Open
Abstract
Wnt/β-catenin signaling plays a critical role in colorectal cancer (CRC) tumorigenesis and the homeostasis of colorectal cancer stem cells (CSCs), but its molecular mechanism remains unclear. B-cell lymphoma 3 (Bcl-3), a member of the IκB family, is overexpressed in CRC and promotes tumorigenicity. Here, we report a novel function of Bcl-3 in maintaining colorectal CSC homeostasis by activating Wnt/β-catenin signaling. Silencing Bcl-3 suppresses the self-renewal capacity of colorectal CSCs and sensitizes CRC cells to chemotherapeutic drugs through a decrease in Wnt/β-catenin signaling. Moreover, our data show that Bcl-3 is a crucial component of Wnt/β-catenin signaling and is essential for β-catenin transcriptional activity in CRC cells. Interestingly, Wnt3a increases the level and nuclear translocation of Bcl-3, which binds directly to β-catenin and enhances the acetylation of β-catenin at lysine 49 (Ac-K49-β-catenin) and transcriptional activity. Bcl-3 depletion decreases the Ac-K49-β-catenin level by increasing the level of histone deacetylase 1 to remove acetyl groups from β-catenin, thus interrupting Wnt/β-catenin activity. In CRC clinical specimens, Bcl-3 expression negatively correlates with the overall survival of CRC patients. A significantly positive correlation was found between the expression of Bcl-3 and Ac-K49-β-catenin. Collectively, our data reveal that Bcl-3 plays a crucial role in CRC chemoresistance and colorectal CSC maintenance via its modulation of the Ac-K49-β-catenin, which serves as a promising therapeutic target for CRC.
Collapse
Affiliation(s)
- Xi Chen
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
| | - Chen Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China.,Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Yuhang Jiang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China.,Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China
| | - Qi Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
| | - Yu Tao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China.,Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China
| | - Haohao Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China.,Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China
| | - Yongxu Zhao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
| | - Yiming Hu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China.,Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China
| | - Cuifeng Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China.,Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China
| | - Deji Ye
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
| | - Dandan Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
| | - Wenxia Jiang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
| | - Eugene Y Chin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
| | - Sheng Chen
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yongzhong Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Mingliang Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Sanhong Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China. .,Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China.
| | - Xiaoren Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China. .,Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, Guangzhou, 510000, China.
| |
Collapse
|
20
|
Yao Y, Wang X, Li H, Fan J, Qian X, Li H, Xu Y. Phospholipase D as a key modulator of cancer progression. Biol Rev Camb Philos Soc 2020; 95:911-935. [PMID: 32073216 DOI: 10.1111/brv.12592] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 02/01/2020] [Accepted: 02/06/2020] [Indexed: 12/15/2022]
Abstract
The phospholipase D (PLD) family has a ubiquitous expression in cells. PLD isoforms (PLDs) and their hydrolysate phosphatidic acid (PA) have been demonstrated to engage in multiple stages of cancer progression. Aberrant expression of PLDs, especially PLD1 and PLD2, has been detected in various cancers. Inhibition or elimination of PLDs activity has been shown to reduce tumour growth and metastasis. PLDs and PA also serve as downstream effectors of various cell-surface receptors, to trigger and regulate propagation of intracellular signals in the process of tumourigenesis and metastasis. Here, we discuss recent advances in understanding the functions of PLDs and PA in discrete stages of cancer progression, including cancer cell growth, invasion and migration, and angiogenesis, with special emphasis on the tumour-associated signalling pathways mediated by PLDs and PA and the functional importance of PLDs and PA in cancer therapy.
Collapse
Affiliation(s)
- Yuanfa Yao
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China.,Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyi Wang
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China.,Department of Clinical Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hanbing Li
- Institute of Pharmacology, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Jiannan Fan
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China
| | - Xiaohan Qian
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China.,Department of Respiratory Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Li
- Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yingke Xu
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China.,Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
21
|
Vahkal B, Yegorov S, Onyilagha C, Donner J, Reddick D, Shrivastav A, Uzonna J, Good SV. Immune System Effects of Insulin-Like Peptide 5 in a Mouse Model. Front Endocrinol (Lausanne) 2020; 11:610672. [PMID: 33519716 PMCID: PMC7841425 DOI: 10.3389/fendo.2020.610672] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/16/2020] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION Insulin-like peptide 5 (INSL5) is a peptide hormone with proposed actions in glucose homeostasis and appetite regulation via its cognate receptor, relaxin family peptide receptor 4 (RXFP4). Here, we look for evidence for their involvement in the immune system using a mouse model. METHODS In silico analyses: we queried public databases for evidence of expression of INSL5-RXFP4 in immune system tissues/cells (NCBI's SRA and GeoProfiles) and disorders (EMBO-EBI) and performed phylogenetic footprinting to look for evidence that they are regulated by immune-associated transcription factors (TFs). Experimental analyses: We characterized the expression and correlation of INSL5/RXFP4 and other immune system markers in central and peripheral immune organs from C57/bl6 mice in seven cohorts. We tested whether fluctuations in circulating INSL5 induce an immune response, by injecting mice with 30 μg/kg of INSL5 peptide in the peritoneum, and examining levels of immune markers and metabolic peptides in plasma. Lastly, we quantified the expression of Rxfp4 in T-cells, dendritic cells and cell lines derived from human and mouse and tested the hypothesis that co-incubation of ANA-1 cells in INSL5 and LPS alters cytokine expression. RESULTS We find Insl5 expression only in thymus (in addition to colon) where its expression was highly correlated with Il-7, a marker of thymocyte development. This result is consistent with our in silico findings that Insl5 is highly expressed in thymic DP, DN thymocytes and cortical TEC's, and with evidence that it is regulated by thymocyte-associated TF's. We find Rxfp4 expression in all immune organs, and moderately high levels in DCs, particularly splenic DCs, and evidence that it is regulated by immune-associated TF's, such as STAT's and GATA. Systemic effects: We observed significantly elevated concentrations of blood GLP-1, GIP, GCG and PYY following intraperitoneal injection of INSL5, and significantly altered expression of cytokines IL-5, IL-7, M-CSF, IL-15, IL-27 and MIP-2. Immune cell effects: Incubation of ANA-1 cells with INSL5 impeded cell growth and led to a transient elevation of IL-15 and sustained reduction in IL-1β, IL-6 and TNFα. CONCLUSION We propose that INSL5-RXFP4 play a novel role in both central and peripheral immune cell signaling.
Collapse
Affiliation(s)
- Brett Vahkal
- Department of Biology, University of Winnipeg, Winnipeg, MB, Canada
- *Correspondence: Brett Vahkal, ; Sara V. Good,
| | - Sergey Yegorov
- Department of Biology, University of Winnipeg, Winnipeg, MB, Canada
| | | | | | - Dean Reddick
- Department of Biology, University of Winnipeg, Winnipeg, MB, Canada
| | | | - Jude Uzonna
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Sara V. Good
- Department of Biology, University of Winnipeg, Winnipeg, MB, Canada
- *Correspondence: Brett Vahkal, ; Sara V. Good,
| |
Collapse
|
22
|
Lee YY, Choi HJ, Lee SY, Park SY, Kang MJ, Han J, Han JS. Bcl-2 Overexpression Induces Neurite Outgrowth via the Bmp4/Tbx3/NeuroD1 Cascade in H19-7 Cells. Cell Mol Neurobiol 2020; 40:153-166. [PMID: 31493044 DOI: 10.1007/s10571-019-00732-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/29/2019] [Indexed: 12/25/2022]
Abstract
Bcl-2 is overexpressed in the nervous system during neural development and plays an important role in modulating cell survival. In addition to its anti-apoptotic function, it has been suggested previously that Bcl-2 might act as a mediator of neuronal differentiation. However, the mechanism by which Bcl-2 might influence neurogenesis is not sufficiently understood. In this study, we aimed to determine the non-apoptotic functions of Bcl-2 during neuronal differentiation. First, we used microarrays to analyze the whole-genome expression patterns of rat neural stem cells overexpressing Bcl-2 and found that Bcl-2 overexpression induced the expression of various neurogenic genes. Moreover, Bcl-2 overexpression increased the neurite length as well as expression of Bmp4, Tbx3, and proneural basic helix-loop-helix genes, such as NeuroD1, NeuroD2, and Mash1, in H19-7 rat hippocampal precursor cells. To determine the hierarchy of these molecules, we selectively depleted Bmp4, Tbx3, and NeuroD1 in Bcl-2-overexpressing cells. Bmp4 depletion suppressed the upregulation of Tbx3 and NeuroD1 as well as neurite outgrowth, which was induced by Bcl-2 overexpression. Although Tbx3 knockdown repressed Bcl-2-mediated neurite elaboration and downregulated NeuroD1 expression, it did not affect Bcl-2-induced Bmp4 expression. While the depletion of NeuroD1 had no effect on the expression of Bcl-2, Bmp4, or Tbx3, Bcl-2-mediated neurite outgrowth was suppressed. Taken together, these results demonstrate that Bcl-2 regulates neurite outgrowth through the Bmp4/Tbx3/NeuroD1 cascade in H19-7 cells, indicating that Bcl-2 may have a direct role in neuronal development in addition to its well-known anti-apoptotic function in response to environmental insults.
Collapse
Affiliation(s)
- Yun Young Lee
- Department of Biomedical Sciences, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea
- Biomedical Research Institute and Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, 04763, Republic of Korea
| | - Hye-Jin Choi
- Biomedical Research Institute and Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, 04763, Republic of Korea
| | - So Young Lee
- Department of Biomedical Sciences, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Shin-Young Park
- Biomedical Research Institute and Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, 04763, Republic of Korea
| | - Min-Jeong Kang
- Department of Biomedical Sciences, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Jinil Han
- Department of Biomedical Sciences, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea
- Gencurix, Inc, Hanwha Bizmetro 1, Guro 3-dong, Guro-gu, Seoul, 08394, Republic of Korea
| | - Joong-Soo Han
- Department of Biomedical Sciences, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea.
- Biomedical Research Institute and Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
23
|
Thakur R, Naik A, Panda A, Raghu P. Regulation of Membrane Turnover by Phosphatidic Acid: Cellular Functions and Disease Implications. Front Cell Dev Biol 2019; 7:83. [PMID: 31231646 PMCID: PMC6559011 DOI: 10.3389/fcell.2019.00083] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 05/03/2019] [Indexed: 01/23/2023] Open
Abstract
Phosphatidic acid (PA) is a simple glycerophospholipid with a well-established role as an intermediate in phospholipid biosynthesis. In addition to its role in lipid biosynthesis, PA has been proposed to act as a signaling molecule that modulates several aspects of cell biology including membrane transport. PA can be generated in eukaryotic cells by several enzymes whose activity is regulated in the context of signal transduction and enzymes that can metabolize PA thus terminating its signaling activity have also been described. Further, several studies have identified PA binding proteins and changes in their activity are proposed to be mediators of the signaling activity of this lipid. Together these enzymes and proteins constitute a PA signaling toolkit that mediates the signaling functions of PA in cells. Recently, a number of novel genetic models for the analysis of PA function in vivo and analytical methods to quantify PA levels in cells have been developed and promise to enhance our understanding of PA functions. Studies of several elements of the PA signaling toolkit in a single cell type have been performed and are presented to provide a perspective on our understanding of the biochemical and functional organization of pools of PA in a eukaryotic cell. Finally, we also provide a perspective on the potential role of PA in human disease, synthesizing studies from model organisms, human disease genetics and analysis using recently developed PLD inhibitors.
Collapse
Affiliation(s)
- Rajan Thakur
- National Centre for Biological Sciences-TIFR, Bengaluru, India
| | - Amruta Naik
- National Centre for Biological Sciences-TIFR, Bengaluru, India
| | - Aniruddha Panda
- National Centre for Biological Sciences-TIFR, Bengaluru, India
| | - Padinjat Raghu
- National Centre for Biological Sciences-TIFR, Bengaluru, India
| |
Collapse
|
24
|
Phospholipase D and the Mitogen Phosphatidic Acid in Human Disease: Inhibitors of PLD at the Crossroads of Phospholipid Biology and Cancer. Handb Exp Pharmacol 2019; 259:89-113. [PMID: 31541319 DOI: 10.1007/164_2019_216] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Lipids are key building blocks of biological membranes and are involved in complex signaling processes such as metabolism, proliferation, migration, and apoptosis. Extracellular signaling by growth factors, stress, and nutrients is transmitted through receptors that activate lipid-modifying enzymes such as the phospholipases, sphingosine kinase, or phosphoinositide 3-kinase, which then modify phospholipids, sphingolipids, and phosphoinositides. One such important enzyme is phospholipase D (PLD), which cleaves phosphatidylcholine to yield phosphatidic acid and choline. PLD isoforms have dual role in cells. The first involves maintaining cell membrane integrity and cell signaling, including cell proliferation, migration, cytoskeletal alterations, and invasion through the PLD product PA, and the second involves protein-protein interactions with a variety of binding partners. Increased evidence of elevated PLD expression and activity linked to many pathological conditions, including cancer, neurological and inflammatory diseases, and infection, has motivated the development of dual- and isoform-specific PLD inhibitors. Many of these inhibitors are reported to be efficacious and safe in cells and mouse disease models, suggesting the potential for PLD inhibitors as therapeutics for cancer and other diseases. Current knowledge and ongoing research of PLD signaling networks will help to evolve inhibitors with increased efficacy and safety for clinical studies.
Collapse
|
25
|
Lu WJ, Chung CL, Chen RJ, Huang LT, Lien LM, Chang CC, Lin KH, Sheu JR. An Antithrombotic Strategy by Targeting Phospholipase D in Human Platelets. J Clin Med 2018; 7:jcm7110440. [PMID: 30441821 PMCID: PMC6262437 DOI: 10.3390/jcm7110440] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/06/2018] [Accepted: 11/13/2018] [Indexed: 01/16/2023] Open
Abstract
Phospholipase D (PLD) is involved in many biological processes. PLD1 plays a crucial role in regulating the platelet activity of mice; however, the role of PLD in the platelet activation of humans remains unclear. Therefore, we investigated whether PLD is involved in the platelet activation of humans. Our data revealed that inhibition of PLD1 or PLD2 using pharmacological inhibitors effectively inhibits platelet aggregation in humans. However, previous studies have showed that PLD1 or PLD2 deletion did not affect mouse platelet aggregation in vitro, whereas only PLD1 deletion inhibited thrombus formation in vivo. Intriguingly, our data also showed that the pharmacological inhibition of PLD1 or PLD2 does not affect mouse platelet aggregation in vitro, whereas the inhibition of only PLD1 delayed thrombus formation in vivo. These findings indicate that PLD may play differential roles in humans and mice. In humans, PLD inhibition attenuates platelet activation, adhesion, spreading, and clot retraction. For the first time, we demonstrated that PLD1 and PLD2 are essential for platelet activation in humans, and PLD plays different roles in platelet function in humans and mice. Our findings also indicate that targeting PLD may provide a safe and alternative therapeutic approach for preventing thromboembolic disorders.
Collapse
Affiliation(s)
- Wan Jung Lu
- Department of Medical Research, Taipei Medical University Hospital, Taipei 110, Taiwan.
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Graduate Institute of Metabolism and Obesity Sciences, College of Public Health and Nutrition, Taipei Medical University, Taipei 110, Taiwan.
| | - Chi Li Chung
- Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan.
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Ray Jade Chen
- Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan.
- School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Li Ting Huang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Li Ming Lien
- School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Neurology, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan.
| | - Chao Chien Chang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Kuan Hung Lin
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Central Laboratory, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan.
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City 252, Taiwan.
| | - Joen Rong Sheu
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
26
|
Zhou W, Shi K, Ji L, Wu R, Chen Y, Tu H, Zhou B, Wang Z, Zhang M. Inhibition of Phospholipase D1 mRNA Expression Slows Down the Proliferation Rate of Prostate Cancer Cells That Have Transited to Androgen Independence. J Cancer 2018; 9:3620-3625. [PMID: 30310520 PMCID: PMC6171019 DOI: 10.7150/jca.26689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 08/06/2018] [Indexed: 12/12/2022] Open
Abstract
To explore the role of phospholipase D1 (PLD1) mRNA in transition of prostate cancer (PCa) cells to androgen independence, we used Arraystar Human LncRNA Microarray V3.0 to detect and compare the differential expression of PLD1 and its signaling pathway-related gene in standard androgen dependence prostate cancer (ADPC) cell line LNCaP before and after the occurrence of androgen independence prostate cancer (AIPC) transition. In addition, we used the shRNA lentiviral vector to inhibit the PLD1 mRNA expression and observed its effect on LNCaP cell proliferation after AIPC transition by using MTS method. The results showed that the expression level of PLD1 mRNA was increased by 373-fold after AIPC transition (P<0.05); the PI3K/AKT signaling pathway-related gene expression was also elevated (P<0.05); the growth rate of LNCaP cells that had transited to androgen independence was reduced by about 30% when the PLD1 mRNA expression was inhibited by the shRNA lentivirus as compared with the negative control group (P<0.05). All these results suggest that PLD1 mRNA and the related PI3K/AKT signaling pathway may play an important role in AIPC. Down-regulating the expression of PLD1 mRNA could to some extent inhibit the proliferation rate of PCa cells after AIPC transition.
Collapse
Affiliation(s)
- Wu Zhou
- Department of Medical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang Wenzhou, 325000, China
| | - Keqing Shi
- Liver Disease Center, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang Wenzhou, 325000,China
| | - Lili Ji
- Department of Medical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang Wenzhou, 325000, China
| | - Ruihao Wu
- Department of Medical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang Wenzhou, 325000, China
| | - Yuehui Chen
- Department of Medical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang Wenzhou, 325000, China
| | - Hongxiang Tu
- Department of Medical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang Wenzhou, 325000, China
| | - Beibei Zhou
- Department of Medical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang Wenzhou, 325000, China
| | - Zhongyong Wang
- Department of Medical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang Wenzhou, 325000, China
| | - Meijuan Zhang
- Department of Medical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Zhejiang Wenzhou, 325000, China
| |
Collapse
|
27
|
Androgen receptor suppresses prostate cancer cell invasion via altering the miR-4496/β-catenin signals. Biochem Biophys Res Commun 2018; 504:82-88. [PMID: 30177389 DOI: 10.1016/j.bbrc.2018.08.134] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 08/22/2018] [Indexed: 12/12/2022]
Abstract
Previous study found that AR in prostate may act as both a proliferator and a suppressor to promote or suppress the metastasis of prostate cancer (PCa). In current work, we demonstrated that AR could suppress PCa cell invasion through altering the miR-4496/β-catenin signals. And mechanisms dissection found that AR could negatively regulate the expression of β-catenin through enhancing the miR-4496 expression via directly binding to the AR-response-elements (AREs) of miR-4496 promoter, subsequently, the miRNA could directly target the 3' UTR of the β-catenin-mRNA to reduce its expression. To conclude, our work suggests that AR might play an important role to suppress PCa cell invasion, targeting the newly identified AR/miR-4496/β-catenin signaling with small molecules may help us to build up new therapeutic approaches to better suppress the metastasis of PCa.
Collapse
|
28
|
Lee YY, Lee SY, Park SY, Choi HJ, Kim EG, Han JS. Therapeutic potential of a phospholipase D1 inhibitory peptide fused with a cell-penetrating peptide as a novel anti-asthmatic drug in a Der f 2-induced airway inflammation model. Exp Mol Med 2018; 50:1-11. [PMID: 29717122 PMCID: PMC5938051 DOI: 10.1038/s12276-018-0083-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/12/2018] [Accepted: 03/05/2018] [Indexed: 01/13/2023] Open
Abstract
Asthma is a chronic lung disease that causes airflow obstruction due to airway inflammation. However, its therapeutics remain inadequate. We previously reported that phospholipase D1 (PLD1) is a key enzyme involved in the production of pro-inflammatory cytokines in airway inflammation induced by the house dust mite allergen Dermatophagoides farinae 2 (Der f 2). We also revealed that PLD1 is specifically inactivated by AP180 (assembly protein, 180 kDa) and identified the PLD1-specific binding motif (TVTSP) of AP180. Therefore, the aims of this study were to develop a novel anti-asthmatic agent that could suppress airway inflammation by inhibiting PLD1 and examine its acute and chronic toxicity. We designed TAT-TVTSP, a PLD1-inhibitory peptide fused with a cell-penetrating peptide (CPP) delivery system. TAT-TVTSP was efficiently delivered to bronchial epithelial cells and significantly reduced Der f 2-induced PLD activation and Interleukin 13 (IL-13) production. Intranasally administered TAT-TVTSP was also efficiently transferred to airway tissues and ameliorated airway inflammation in a Der f 2-induced allergic asthma mouse model. Moreover, we investigated the safety of TAT-TVTSP as a therapeutic agent through single- and repeated-dose toxicity studies in a mouse model. Taken together, these results indicated that a PLD1-inhibitory peptide fused with a cell-penetrating peptide may be useful for treating allergic inflammatory asthma induced by house dust mites (HDMs). A drug that targets a key enzyme involved in airway tissue inflammation shows promise in the treatment of allergic asthma. The enzyme phospholipase D1 (PLD1) triggers airway inflammation in allergic asthma brought on by house dust mites. Joong-Soo Han at Hanyang University in Seoul, Eung-Gook Kim at Chungbuk National University, Cheongju, South Korea, and co-workers have developed a treatment aimed at suppressing PLD1 and trialed the drug on mouse models of dust-mite allergy. The team designed a carrier system capable of accurately delivering a PLD1-inhibitory peptide to airway tissues and cells. They found that airway inflammation was significantly reduced in the treated mice. The drug appeared to be relatively safe when used in repeated doses, although further investigations are needed to verify this. The team hope their treatment will improve therapies for allergic asthma.
Collapse
Affiliation(s)
- Yun Young Lee
- Department of Biomedical Sciences, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - So Young Lee
- Department of Biomedical Sciences, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Shin-Young Park
- Biomedical Research Institute and Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, 04763, Republic of Korea
| | - Hye-Jin Choi
- Biomedical Research Institute and Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, 04763, Republic of Korea
| | - Eung-Gook Kim
- Department of Biochemistry and Signaling Disorder Research Center, College of Medicine, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| | - Joong-Soo Han
- Department of Biomedical Sciences, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea. .,Biomedical Research Institute and Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
29
|
Phospholipases play multiple cellular roles including growth, stress tolerance, sexual development, and virulence in fungi. Microbiol Res 2018; 209:55-69. [DOI: 10.1016/j.micres.2017.12.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 12/21/2017] [Accepted: 12/31/2017] [Indexed: 12/16/2022]
|
30
|
Wils LJ, Bijlsma MF. Epigenetic regulation of the Hedgehog and Wnt pathways in cancer. Crit Rev Oncol Hematol 2018; 121:23-44. [DOI: 10.1016/j.critrevonc.2017.11.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 11/17/2017] [Accepted: 11/17/2017] [Indexed: 12/14/2022] Open
|
31
|
Wang Z, Zhang F, He J, Wu P, Tay LWR, Cai M, Nian W, Weng Y, Qin L, Chang JT, McIntire LB, Di Paolo G, Xu J, Peng J, Du G. Binding of PLD2-Generated Phosphatidic Acid to KIF5B Promotes MT1-MMP Surface Trafficking and Lung Metastasis of Mouse Breast Cancer Cells. Dev Cell 2017; 43:186-197.e7. [PMID: 29033361 DOI: 10.1016/j.devcel.2017.09.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 07/19/2017] [Accepted: 09/14/2017] [Indexed: 12/22/2022]
Abstract
Little is known about the cellular events promoting metastasis. We show that knockout of phospholipase D2 (PLD2), which generates the signaling lipid phosphatidic acid (PA), inhibits lung metastases in the mammary tumor virus (MMTV)-Neu transgenic mouse breast cancer model. PLD2 promotes local invasion through the regulation of the plasma membrane targeting of MT1-MMP and its associated invadopodia. A liposome pull-down screen identifies KIF5B, the heavy chain of the motor protein kinesin-1, as a new PA-binding protein. In vitro assays reveal that PA specifically and directly binds to the C terminus of KIF5B. The binding between PLD2-generated PA and KIF5B is required for the vesicular association of KIF5B, surface localization of MT1-MMP, invadopodia, and invasion in cancer cells. Taken together, these results identify a role of PLD2-generated PA in the regulation of kinesin-1 motor functions and breast cancer metastasis and suggest PLD2 as a potential therapeutic target for metastatic breast cancer.
Collapse
Affiliation(s)
- Ziqing Wang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | - Feng Zhang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA; Core Facility, Department of Clinical Laboratory, Quzhou People's Hospital, Quzhou, Zhejiang, China
| | - Jingquan He
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | - Ping Wu
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | - Li Wei Rachel Tay
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | - Ming Cai
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA; Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430022, China
| | - Weiqi Nian
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA; Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing Cancer Hospital & Institute & Cancer Center, Chongqing 400030, China
| | - Yuanyuan Weng
- Core Facility, Department of Clinical Laboratory, Quzhou People's Hospital, Quzhou, Zhejiang, China
| | - Li Qin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jeffrey T Chang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA
| | - Laura B McIntire
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Gilbert Di Paolo
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Proteomics Facility, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Guangwei Du
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX 77030, USA.
| |
Collapse
|
32
|
Kang DW, Lee BH, Suh YA, Choi YS, Jang SJ, Kim YM, Choi KY, Min DS. Phospholipase D1 Inhibition Linked to Upregulation of ICAT Blocks Colorectal Cancer Growth Hyperactivated by Wnt/β-Catenin and PI3K/Akt Signaling. Clin Cancer Res 2017; 23:7340-7350. [DOI: 10.1158/1078-0432.ccr-17-0749] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 07/19/2017] [Accepted: 09/18/2017] [Indexed: 11/16/2022]
|
33
|
PLD1 overexpression promotes invasion and migration and function as a risk factor for Chinese glioma patients. Oncotarget 2017; 8:57039-57046. [PMID: 28915652 PMCID: PMC5593623 DOI: 10.18632/oncotarget.18961] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 06/18/2017] [Indexed: 12/13/2022] Open
Abstract
Glioma is a lethal disease with few effective therapeutic options. Recently, insights into cancer biology had suggested that abnormal lipid metabolism was a risk factor for various human malignancies, including glioma. As a key enzyme implicated in lipid metabolism, PLD1 was overexpression in multiple human cancers, and it was stated to be responsible for aggressive phenotypes, such as angiogenesis and chemoresistance. However, there was still much to know about its expression and function in glioma. In the present study, we showed that PLD1 was overexpression in clinical samples of glioma. In addition, the correlation assay revealed that PLD1 overexpression was correlated with poor differentiation (p = 0.04), and it was responsible for a poor prognosis for the patients (p = 0.009). Furthermore, we showed in COX regression assay that PLD1 was a risk factor for glioma (p = 0.018, HR = 0.461, 95% CI = 0.243–0.887). Consistently, we found that PLD1 was overexpression in glioma cell lines, and it could facilitate the proliferation and migration. Taken together, our study suggested that PLD1 was pro-tumoral in glioma, and that further studies were urgently needed so as to define whether it was a novel therapeutic target for the disease.
Collapse
|
34
|
Liu Y, Li H, Ban Z, Nai M, Yang L, Chen Y, Xu Y. Annexin A2 inhibition suppresses ovarian cancer progression via regulating β-catenin/EMT. Oncol Rep 2017; 37:3643-3650. [PMID: 28440436 DOI: 10.3892/or.2017.5578] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 03/16/2017] [Indexed: 01/23/2023] Open
Abstract
Annexin A2 is a member of the Annexin family that acts as a Ca2+-dependent phospholipid and membrane binding protein, which is associated with the survival and spread of multiple neoplasms. However, the function of Annexin A2 in ovarian cancer progression remains unclear. In this study, we aimed to investigate the role and underlying molecular mechanism of Annexin A2 in cell proliferation and invasion in ovarian cancer. We found that the mRNA expression of Annexin A2 was upregulated in ovarian cancer tissues and cell lines. In the loss-of-function of Annexin A2, β-catenin was indicated to be significantly suppressed and EMT constrained. Moreover, cell proliferation and invasion were both markedly inhibited by the downregulation of Annexin A2. Additionally, the overexpression of β-catenin obviously reversed the effect of Annexin A2 on EMT, and cell proliferation and invasion, indicating that Annexin A2 suppression regulated EMT through controlling β-catenin. Taken together, this study showed that Annexin A2 inhibition suppresses proliferation and invasion in ovarian cancer via β-catenin/EMT, proposing the potential role of Annexin A2 in the prevention and treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yan Liu
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Hongyu Li
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Zhenying Ban
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Manman Nai
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Li Yang
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Yannan Chen
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Yiming Xu
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
35
|
Kang DW, Yang ES, Noh YN, Hwang WC, Jo SY, Suh YA, Park WS, Choi KY, Min DS. MicroRNA-320a and microRNA-4496 attenuate Helicobacter pylori cytotoxin-associated gene A (CagA)-induced cancer-initiating potential and chemoresistance by targeting β-catenin and ATP-binding cassette, subfamily G, member 2. J Pathol 2017; 241:614-625. [PMID: 28008607 DOI: 10.1002/path.4866] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 12/01/2016] [Accepted: 12/15/2016] [Indexed: 01/04/2023]
Abstract
Infection with Helicobacter pylori is closely linked to an increased risk of gastric cancer. Although cytotoxin-associated gene A (CagA), a major virulence factor of H. pylori, is known to be a causal factor for gastric carcinogenesis, the molecular link between CagA and gastric cancer-initiating cell (CIC)-like properties remains elusive. Here, we demonstrate that CagA is required for increased expression of β-catenin and its target CIC markers via downregulation of microRNA (miR)-320a and miR-4496. CagA promoted gastric CIC properties and was responsible for chemoresistance. miR-320a and miR-4496 attenuated the in vitro self-renewal and tumour-initiating capacity of CagA-expressing CICs by targeting β-catenin. Moreover, miR-320a and miR-4496 decreased CagA-induced chemoresistance by targeting ATP-binding cassette, subfamily G, member 2 (ABCG2) at the transcriptional and post-transcriptional levels, respectively. Combination therapy with 5-fluorouracil and miR-320a/miR-4496 suppressed gastric tumourigenesis and metastatic potential in an orthotopic mouse model, probably via suppression of CagA-induced CIC properties and chemoresistance. Our results provide novel evidence that CIC properties, chemoresistance and tumourigenesis associated with H. pylori are linked to CagA-induced upregulation of β-catenin and ABCG2. These data provide novel insights into the molecular mechanisms of CagA-induced carcinogenisis and the therapeutic potential of of miR-320a and miR-4496. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Dong Woo Kang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea.,Institute of Innovative Cancer Research, Asan Institute for Life Science, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eun Sun Yang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Yu Na Noh
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Won Chan Hwang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Se-Young Jo
- Institute of Innovative Cancer Research, Asan Institute for Life Science, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young-Ah Suh
- Institute of Innovative Cancer Research, Asan Institute for Life Science, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Won Sang Park
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kang-Yell Choi
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.,Translational Research Centre for Protein Function Control, Yonsei University, Seoul, Republic of Korea
| | - Do Sik Min
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea.,Translational Research Centre for Protein Function Control, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
36
|
Cheng M, Bhujwalla ZM, Glunde K. Targeting Phospholipid Metabolism in Cancer. Front Oncol 2016; 6:266. [PMID: 28083512 PMCID: PMC5187387 DOI: 10.3389/fonc.2016.00266] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 12/14/2016] [Indexed: 12/14/2022] Open
Abstract
All cancers tested so far display abnormal choline and ethanolamine phospholipid metabolism, which has been detected with numerous magnetic resonance spectroscopy (MRS) approaches in cells, animal models of cancer, as well as the tumors of cancer patients. Since the discovery of this metabolic hallmark of cancer, many studies have been performed to elucidate the molecular origins of deregulated choline metabolism, to identify targets for cancer treatment, and to develop MRS approaches that detect choline and ethanolamine compounds for clinical use in diagnosis and treatment monitoring. Several enzymes in choline, and recently also ethanolamine, phospholipid metabolism have been identified, and their evaluation has shown that they are involved in carcinogenesis and tumor progression. Several already established enzymes as well as a number of emerging enzymes in phospholipid metabolism can be used as treatment targets for anticancer therapy, either alone or in combination with other chemotherapeutic approaches. This review summarizes the current knowledge of established and relatively novel targets in phospholipid metabolism of cancer, covering choline kinase α, phosphatidylcholine-specific phospholipase D1, phosphatidylcholine-specific phospholipase C, sphingomyelinases, choline transporters, glycerophosphodiesterases, phosphatidylethanolamine N-methyltransferase, and ethanolamine kinase. These enzymes are discussed in terms of their roles in oncogenic transformation, tumor progression, and crucial cancer cell properties such as fast proliferation, migration, and invasion. Their potential as treatment targets are evaluated based on the current literature.
Collapse
Affiliation(s)
- Menglin Cheng
- Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kristine Glunde
- Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
37
|
Hu J, Hu H, Hang JJ, Yang HY, Wang ZY, Wang L, Chen DH, Wang LW. Simultaneous high expression of PLD1 and Sp1 predicts a poor prognosis for pancreatic ductal adenocarcinoma patients. Oncotarget 2016; 7:78557-78565. [PMID: 27713167 PMCID: PMC5346659 DOI: 10.18632/oncotarget.12447] [Citation(s) in RCA: 19] [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: 03/09/2016] [Accepted: 09/20/2016] [Indexed: 01/05/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease with few therapeutic options. Recently, insight into cancer biology suggested abnormal lipid metabolism to be a risk factor for human malignancies. As a key enzyme implicated in lipid metabolism, PLD1 was elevated in various human cancer associating with malignant phenotypes. However, little was known about its expression and function in PDAC. We showed that PLD1 was elevated in both the cell lines and clinical samples of PDAC, and it positively correlated with vascular invasion (p = 0.041) and responsible for a poor prognosis (p = 0.009). Meanwhile, we also found Sp1 to be elevated in the disease, correlating with vascular invasion (p = 0.007). Moreover, the correlation assay suggested that PLD1 positively correlated with Sp1 in the clinical sample (r = 0.390; p < 0.001) and the cell lines. Finally, we showed that co-high expression of both the factors confers the poorest prognosis for the patients, and that their simultaneous high expression might be an independent prognostic factor (p = 0.001; HR = 3.427; 95% CI 1.629-7.211).
Collapse
Affiliation(s)
- Jiong Hu
- Department of Medical Oncology, Shanghai General Hospital of Nanjing Medical University, Shanghai 201620, China
- Department of Medical Oncology and Pancreatic Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai 201620, China
| | - Hai Hu
- Department of Medical Oncology and Pancreatic Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai 201620, China
| | - Jun-jie Hang
- Department of Medical Oncology and Pancreatic Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai 201620, China
| | - Hai-yan Yang
- Department of Medical Oncology and Pancreatic Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai 201620, China
| | - Zhi-yong Wang
- Department of Medical Oncology and Pancreatic Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai 201620, China
| | - Lei Wang
- Department of Medical Oncology and Pancreatic Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai 201620, China
| | - Dong-hui Chen
- Department of Medical Oncology and Pancreatic Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai 201620, China
| | - Li-wei Wang
- Department of Medical Oncology, Shanghai General Hospital of Nanjing Medical University, Shanghai 201620, China
- Department of Medical Oncology and Pancreatic Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai 201620, China
| |
Collapse
|
38
|
Song HI, Yoon MS. PLD1 regulates adipogenic differentiation through mTOR - IRS-1 phosphorylation at serine 636/639. Sci Rep 2016; 6:36968. [PMID: 27872488 PMCID: PMC5181839 DOI: 10.1038/srep36968] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 10/24/2016] [Indexed: 01/08/2023] Open
Abstract
Phospholipase D1 (PLD1) plays a known role in several differentiation processes, but its role in adipogenic differentiation remains unknown. In the present study, we identified PLD1 as a negative regulator of adipogenic differentiation. We showed that PLD activity was downregulated by both 3-Isobutyl-1-methylxanthine (IBMX) and insulin upon induction of differentiation in 3T3-L1 adipogenic cells. In line with this observation, PLD activity decreased in both high fat diet (HFD)-fed mice and ob/ob mice. We also found that differentiation of 3T3-L1 preadipocytes was enhanced by the depletion of PLD1 levels or inhibition of PLD1 activity by VU0155069, a PLD1-specific inhibitor. Conversely, treatment with phosphatidic acid (PA), a PLD product, and overexpression of PLD1 both caused a decrease in adipogenic differentiation. Moreover, the elevated differentiation in PLD1-knockdown 3T3-L1 cells was reduced by either PA treatment or PLD1 expression, confirming negative roles of PLD1 and PA in adipogenic differentiation. Further investigation revealed that PA displaces DEP domain-containing mTOR-interacting protein (DEPTOR) from mTORC1, which subsequently phosphorylates insulin receptor substrate-1 (IRS-1) at serine 636/639 in 3T3-L1 cells. Taken together, our findings provide convincing evidence for a direct role of PLD1 in adipogenic differentiation by regulating IRS-1 phosphorylation at serine 636/639 through DEPTOR displacement and mTOR activation.
Collapse
Affiliation(s)
- Hae-In Song
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| | - Mee-Sup Yoon
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| |
Collapse
|
39
|
Kang DW, Lee SW, Hwang WC, Lee BH, Choi YS, Suh YA, Choi KY, Min DS. Phospholipase D1 Acts through Akt/TopBP1 and RB1 to Regulate the E2F1-Dependent Apoptotic Program in Cancer Cells. Cancer Res 2016; 77:142-152. [PMID: 27793841 DOI: 10.1158/0008-5472.can-15-3032] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 10/10/2016] [Accepted: 10/23/2016] [Indexed: 01/11/2023]
Abstract
The RB1/E2F1 signaling pathway is frequently deregulated in colorectal cancer and has been suggested to intersect with Wnt/β-catenin and PI3K/Akt pathways, but molecular evidence for this link is lacking. In this study, we demonstrate that phospholipase D1 (PLD1), a transcriptional target of β-catenin/TCF4, orchestrates functional interactions between these pathways during intestinal tumor development. Overexpression of PLD1 in intestinal epithelial cells protected cells from apoptosis induced by PLD1 ablation in the Apcmin/+ mouse model of intestinal tumorigenesis. Mechanistic investigations revealed that genetic and pharmacologic targeting of PLD1 promote the E2F1-dependent apoptotic program via both miR-192/4465-mediated downregulation of RB1 and inhibition of Akt-TopBP1 pathways. Moreover, the miRNA-RB1 axis and Akt pathway also contributed to the PLD1-mediated self-renewal capacity of colon cancer-initiating cells. Finally, PLD1-driven E2F1 target gene expression positively correlated with tumor stage in patients with colorectal cancer. Overall, our findings suggest that PLD1 mediates cross-talk between multiple major signaling pathways to promote the survival and malignancy of colon cancer cells and may therefore represent an ideal signaling node for therapeutic targeting. Cancer Res; 77(1); 142-52. ©2016 AACR.
Collapse
Affiliation(s)
- Dong Woo Kang
- Institute of Innovative Cancer Research, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Molecular Biology, Pusan National University, Busan, Republic of Korea
| | - Shin Wha Lee
- Institute of Innovative Cancer Research, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Obstetrics and Gynecology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Won Chan Hwang
- Department of Molecular Biology, Pusan National University, Busan, Republic of Korea
| | - Bo Hui Lee
- Department of Statistics, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Yong-Seok Choi
- Department of Statistics, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Young-Ah Suh
- Institute of Innovative Cancer Research, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kang-Yell Choi
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.,Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea
| | - Do Sik Min
- Department of Molecular Biology, Pusan National University, Busan, Republic of Korea. .,Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea.,Genetic Engineering Institute, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
40
|
Kang DW, Noh YN, Hwang WC, Choi KY, Min DS. Rebamipide attenuates Helicobacter pylori CagA-induced self-renewal capacity via modulation of β-catenin signaling axis in gastric cancer-initiating cells. Biochem Pharmacol 2016; 113:36-44. [PMID: 27265143 DOI: 10.1016/j.bcp.2016.06.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 06/01/2016] [Indexed: 02/07/2023]
Abstract
Rebamipide, a mucosal-protective agent, is used clinically for treatment of gastritis and peptic ulcers induced by Helicobacter pylori (H. pylori) which is associated with increased risk of gastric cancer. Although rebamipide is known to inhibit the growth of gastric cancer cells, the action mechanisms of rebamipide in gastric carcinogenesis remains elusive. Here, we show that rebamipide suppresses H. pylori CagA-induced β-catenin and its target cancer-initiating cells (C-IC) marker gene expression via upregulation of miRNA-320a and -4496. Rebamipide attenuated in vitro self-renewal capacity of H. pylori CagA-infected gastric C-IC via modulation of miRNA-320a/-4496-β-catenin signaling axis. Moreover, rebamipide enhanced sensitivity to chemotherapeutic drugs in CagA-expressed gastric C-IC. Furthermore, rebamipide suppressed tumor-initiating capacity of gastric C-IC, probably via suppression of CagA-induced C-IC properties. These data provide novel insights for the efficacy of rebamipide as a chemoprotective drug against H. pylori CagA-induced carcinogenic potential.
Collapse
Affiliation(s)
- Dong Woo Kang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 609-735, Republic of Korea; Institute of Innovative Cancer Research, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yu Na Noh
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 609-735, Republic of Korea
| | - Won Chan Hwang
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 609-735, Republic of Korea
| | - Kang-Yell Choi
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea; Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea
| | - Do Sik Min
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan 609-735, Republic of Korea; Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
41
|
Tropisetron suppresses colitis-associated cancer in a mouse model in the remission stage. Int Immunopharmacol 2016; 36:9-16. [PMID: 27104313 DOI: 10.1016/j.intimp.2016.04.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/10/2016] [Accepted: 04/12/2016] [Indexed: 02/07/2023]
Abstract
Patients with inflammatory bowel disease (IBD) have a high risk for development of colitis-associated cancer (CAC). Serotonin is a neurotransmitter produced by enterochromaffin cells of the intestine. Serotonin and its receptors, mainly 5-HT3 receptor, are overexpressed in IBD and promote development of CAC through production of inflammatory cytokines. In the present study, we demonstrated the in vivo activity of tropisetron, a 5-HT3 receptor antagonist, against experimental CAC. CAC was induced by azoxymethane (AOM)/dextran sodium sulfate (DDS) in BALB/c mice. The histopathology of colon tissue was performed. Beta-catenin and Cox-2 expression was evaluated by immunohistochemistry as well as quantitative reverse transcription-PCR (qRT-PCR). Alterations in the expression of 5-HT3 receptor and inflammatory-associated genes such as Il-1β, Tnf-α, Tlr4 and Myd88 were determined by qRT-PCR. Our results showed that tumor development in tropisetron-treated CAC group was significantly lower than the controls. The qRT-PCR analysis demonstrated that the expression of 5-HT3 receptor was significantly increased following CAC induction. In addition, tropisetron reduced expression of β-catenin and Cox-2 in the CAC experimental group. The levels of Il-1β, Tnf-α, Tlr4 and Myd88 were significantly decreased upon tropisetron treatment in the AOM/DSS group. Taken together, our data show that tropisetron inhibits development of CAC probably by attenuation of inflammatory reactions in the colitis.
Collapse
|
42
|
Park MH, Choi KY, Min DS. The pleckstrin homology domain of phospholipase D1 accelerates EGFR endocytosis by increasing the expression of the Rab5 effector, rabaptin-5. Exp Mol Med 2015; 47:e200. [PMID: 26680696 PMCID: PMC4686693 DOI: 10.1038/emm.2015.101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 10/21/2015] [Accepted: 10/23/2015] [Indexed: 01/28/2023] Open
Abstract
Endocytosis is differentially regulated by hypoxia-inducible factor-1α (HIF-1α) and phospholipase D (PLD). However, the relationship between HIF-1α and PLD in endocytosis is unknown. HIF-1α is degraded through the prolyl hydroxylase (PHD)/von Hippel–Lindau (VHL) ubiquitination pathway in an oxygen-dependent manner. Here, we show that PLD1 recovers the decrease in epidermal growth factor receptor (EGFR) endocytosis induced by HIF-1α independent of lipase activity via the Rab5-mediated endosome fusion pathway. EGF-induced interaction of PLD1 with HIF-1α, PHD and VHL may contribute to EGFR endocytosis. The pleckstrin homology domain (PH) of PLD1 itself promotes degradation of HIF-1α, then accelerates EGFR endocytosis via upregulation of rabaptin-5 and suppresses tumor progression. These findings reveal a novel role of the PLD1-PH domain as a positive regulator of endocytosis and provide a link between PLD1 and HIF-1α in the EGFR endocytosis pathway.
Collapse
Affiliation(s)
- Mi Hee Park
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Kang-Yell Choi
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.,Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea
| | - Do Sik Min
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea.,Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea.,Genetic Engineering Institute, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
43
|
Luteolin Inhibits Ischemia/Reperfusion-Induced Myocardial Injury in Rats via Downregulation of microRNA-208b-3p. PLoS One 2015; 10:e0144877. [PMID: 26658785 PMCID: PMC4685996 DOI: 10.1371/journal.pone.0144877] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/24/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Luteolin (LUT), a kind of flavonoid which is extracted from a variety of diets, has been reported to convey protective effects of various diseases. Recent researches have suggested that LUT can carry out cardioprotective effects during ischemia/reperfusion (I/R). However, there have no reports on whether LUT can exert protective effects against myocardial I/R injury through the actions of specific microRNAs (miRs). The purpose of this study was to determine which miRs and target genes LUT exerted such function through. METHODS Expression of various miRs in perfused rat hearts was detected using a gene chip. Target genes were predicted with TargetScan, MiRDB and MiRanda. Anoxia/reoxygenation was used to simulate I/R. Cells were transfected by miR-208b-3p mimic, inhibitor and small interfering RNA of Ets1 (avian erythroblastosis virus E26 (v ets) oncogene homolog 1). MiR-208b-3p and Ets1 mRNA were quantified by real-time quantitative polymerase chain reaction. The percentage of apoptotic cells was detected by annexin V-fluorescein isothiocyanate/propidium iodide dyeing and flow cytometry. The protein expression levels of cleaved caspase-3, Bcl-2, Bax, and Ets1 were examined by western blot analysis. A luciferase reporter assay was used to verify the combination between miR-208b-3p and the 3'-untranslated region of Ets1. RESULTS LUT pretreatment reduced miR-208b-3p expression in myocardial tissue, as compared to the I/R group. And LUT decreased miR-208b-3p expression and apoptosis caused by I/R. However, overexpression of miR-208b-3p further aggravated the changes caused by I/R and blocked all the effects of LUT. Knockdown of miR-208b-3p expression also attenuated apoptosis, while knockdown of Ets1 promoted apoptosis. Further, the luciferase reporter assay showed that miR-208b-3p could inhibit Ets1 expression. CONCLUSION LUT pretreatment conveys anti-apoptotic effects after myocardial I/R injury by decreasing miR-208b-3p and increasing Ets1 expression levels.
Collapse
|
44
|
Park MH, Bae SS, Choi KY, Min DS. Phospholipase D2 promotes degradation of hypoxia-inducible factor-1α independent of lipase activity. Exp Mol Med 2015; 47:e196. [PMID: 26611735 PMCID: PMC4673472 DOI: 10.1038/emm.2015.87] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 08/19/2015] [Accepted: 09/08/2015] [Indexed: 01/04/2023] Open
Abstract
Hypoxia-inducible factor-1α (HIF-1α) is a key transcriptional mediator that coordinates the expression of various genes involved in tumorigenesis in response to changes in oxygen tension. The stability of HIF-1α protein is determined by oxygen-dependent prolyl hydroxylation, which is required for binding of the von Hippel-Lindau protein (VHL), the recognition component of an E3 ubiquitin ligase that targets HIF-1α for ubiquitination and degradation. Here, we demonstrate that PLD2 protein itself interacts with HIF-1α, prolyl hydroxylase (PHD) and VHL to promote degradation of HIF-1α via the proteasomal pathway independent of lipase activity. PLD2 increases PHD2-mediated hydroxylation of HIF-1α by increasing the interaction of HIF-1α with PHD2. Moreover, PLD2 promotes VHL-dependent HIF-1α degradation by accelerating the association between VHL and HIF-1α. The interaction of the pleckstrin homology domain of PLD2 with HIF-1α also promoted degradation of HIF-1α and decreased expression of its target genes. These results indicate that PLD2 negatively regulates the stability of HIF-1α through the dynamic assembly of HIF-1α, PHD2 and VHL.
Collapse
Affiliation(s)
- Mi Hee Park
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Sun Sik Bae
- Department of Pharmacology, School of Medicine, Pusan National University, Busan, Republic of Korea
| | - Kang-Yell Choi
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea
| | - Do Sik Min
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea
- Genetic Engineering Institute, Pusan National University, Busan, Republic of Korea
| |
Collapse
|