1
|
Eckel O, Mirea MA, Gschwendtner A, Pistek M, Kinslechner K, Röhrl C, Stangl H, Hengstschläger M, Mikula M. Expression of the cholesterol transporter SR-B1 in melanoma cells facilitates inflammatory signaling leading to reduced cholesterol synthesis. Neoplasia 2025; 63:101154. [PMID: 40120430 PMCID: PMC11981749 DOI: 10.1016/j.neo.2025.101154] [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: 02/04/2025] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/25/2025]
Abstract
Scavenger receptor class B type 1 (SR-B1) is a cholesterol transporter, abundantly expressed in human melanoma, yet its precise role for melanoma progression is not fully understood. This study investigates the involvement of SR-B1 in cholesterol homeostasis of tumor cells and its implications for potential therapy. We found that SR-B1 depletion in melanoma cells does not alter total cholesterol levels, but induces cholesterol biosynthesis. This effect was characterized by an increased expression of HMG-CoA reductase (HMGCR), a rate limiting enzyme of cholesterol biosynthesis. Notably, further analyses indicated that this regulation occurs at the post-translational level, mediated via the hypoxia-inducible factor (HIF) signaling pathway. Importantly, we identified SR-B1 as a transporter of the lipid hormone sphingosine-1-phosphate (S1P) and we found that S1P exposure leads to HIF1A up-regulation. Finally, we used a pluripotent stem cell-derived skin organoid model to show that targeting SR-B1 in combination with targeted melanoma therapy can lead to increased apoptosis and suppressed proliferation of transplanted tumor cells. Our study shows that functional SR-B1 is linked to inflammatory signaling, which reduces cholesterol synthesis, while enabling melanoma cell survival during chemotherapy treatment.
Collapse
Affiliation(s)
- Oliver Eckel
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Madalina A Mirea
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Anna Gschwendtner
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Martina Pistek
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Katharina Kinslechner
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Clemens Röhrl
- University of Applied Sciences Upper Austria, Faculty of Engineering, Stelzhamerstraße 23, 4600, Wels, Austria
| | - Herbert Stangl
- Institute of Medical Chemistry, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Markus Hengstschläger
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Mario Mikula
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria.
| |
Collapse
|
2
|
Bolanle IO, de Liedekerke Beaufort GC, Weinberg PD. Transcytosis of LDL Across Arterial Endothelium: Mechanisms and Therapeutic Targets. Arterioscler Thromb Vasc Biol 2025; 45:468-480. [PMID: 40013359 PMCID: PMC11936472 DOI: 10.1161/atvbaha.124.321549] [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: 12/12/2024] [Accepted: 02/11/2025] [Indexed: 02/28/2025]
Abstract
Transport of LDL (low-density lipoprotein) from plasma to arterial intima is thought to be rate limiting in the development of atherosclerosis. Its variation likely determines where lesions develop within arteries and might account for some of the currently unexplained difference in disease susceptibility between individuals. It may also be critical in the development of lipid-rich, unstable plaques. Mechanisms have been controversial but recent evidence suggests that caveolar transcytosis across endothelial cells is the dominant pathway. Receptors involved are LDLR (LDL receptor), SR-B1 (scavenger receptor class B type 1), and ALK1 (activin receptor-like kinase 1). The role of LDLR is influenced by IL-1β (interleukin-1β); the role of SR-B1 by HDL (high-density lipoprotein), DOCK4 (dedicator of cytokinesis 4), GPER (G-protein-coupled estrogen receptor), and HMGB1 (high mobility group box 1); and the role of ALK1 by BMP (bone morphogenetic protein) 9. Additionally, BMP4 stimulates transcytosis and FSTL1 (follistatin-like 1 protein) inhibits it. Fundamental transcytotic mechanisms include caveola formation, undocking, trafficking, and docking; they are influenced by cholesterol-lowering agents, MYDGF (myeloid-derived growth factor), MFSD2a (major facilitator superfamily domain containing 2a) in the blood-brain barrier, and inhibitors of dynamin-2 and tubulin polymerization. The relative merits of different therapeutic approaches are discussed, with statins, colchicine, benzimidazoles, and metformin being existing drugs that might be repurposed and salidroside and glycyrrhizic acid being nutraceuticals worth investigating. Finally, we discuss evidence against the ferry-boat model of transcytosis, the contributions of receptor-mediated, fluid-phase, and active transcytosis, and where inhibition of transcytosis might be most beneficial.
Collapse
Affiliation(s)
- Israel O. Bolanle
- Department of Bioengineering, Imperial College London, United Kingdom
| | | | - Peter D. Weinberg
- Department of Bioengineering, Imperial College London, United Kingdom
| |
Collapse
|
3
|
Dai CL, Qiu ZY, Wang AQ, Yan S, Zhang LJ, Luan X. Targeting cholesterol metabolism: a promising therapy strategy for cancer. Acta Pharmacol Sin 2025:10.1038/s41401-025-01531-9. [PMID: 40133625 DOI: 10.1038/s41401-025-01531-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/03/2025] [Indexed: 03/27/2025]
Abstract
Cholesterol is a crucial structural component of cell membranes, playing a vital role in maintaining membrane fluidity and stability. Cholesterol metabolism involves four interconnected processes: de novo synthesis, uptake, efflux, and esterification. Disruptions in any of these pathways can lead to imbalances in cholesterol homeostasis, which are significantly associated with cancer progression. In recent years, traditional Chinese medicine (TCM) has emerged as a comprehensive therapeutic approach with multi-target and multi-pathway effects, demonstrating significant potential in regulating cholesterol metabolism. Research has shown that certain components of TCM can modulate enzymes, transport proteins, and signaling pathways involved in cholesterol metabolism, effectively interfering with survival and migration of cancer. These mechanisms highlight the unique advantages of TCM in inhibiting tumor progression. In this review we systematically describe the execution and regulation of the four key cholesterol metabolism processes, highlights the roles of critical proteins involved, and provides a comprehensive overview of natural products from TCM that modulate cholesterol metabolism. This review provides valuable insights for the development of novel drugs and cancer therapeutic strategies targeting cholesterol metabolism.
Collapse
Affiliation(s)
- Chun-Lan Dai
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zi-Yang Qiu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - An-Qi Wang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shen Yan
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li-Jun Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Xin Luan
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
4
|
Wu K, Lin F. Lipid Metabolism as a Potential Target of Liver Cancer. J Hepatocell Carcinoma 2024; 11:327-346. [PMID: 38375401 PMCID: PMC10875169 DOI: 10.2147/jhc.s450423] [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: 11/17/2023] [Accepted: 01/25/2024] [Indexed: 02/21/2024] Open
Abstract
Hepatocellular carcinoma (HCC) stands as a severe malignant tumor with a profound impact on overall health, often accompanied by an unfavorable prognosis. Despite some advancements in the diagnosis and treatment of this disease, improving the prognosis of HCC remains a formidable challenge. It is noteworthy that lipid metabolism plays a pivotal role in the onset, development, and progression of tumor cells. Existing research indicates the potential application of targeting lipid metabolism in the treatment of HCC. This review aims to thoroughly explore the alterations in lipid metabolism in HCC, offering a detailed account of the potential advantages associated with innovative therapeutic strategies targeting lipid metabolism. Targeting lipid metabolism holds promise for potentially enhancing the prognosis of HCC.
Collapse
Affiliation(s)
- Kangze Wu
- Department of Hepatobiliary Surgery, Shaoxing People’s Hospital, Shaoxing, People’s Republic of China
| | - Feizhuan Lin
- Department of Hepatobiliary Surgery, Shaoxing People’s Hospital, Shaoxing, People’s Republic of China
| |
Collapse
|
5
|
Zhang Q, Yuan Y, Cao S, Kang N, Qiu F. Withanolides: Promising candidates for cancer therapy. Phytother Res 2024; 38:1104-1158. [PMID: 38176694 DOI: 10.1002/ptr.8090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/14/2023] [Accepted: 11/28/2023] [Indexed: 01/06/2024]
Abstract
Natural products have played a significant role throughout history in the prevention and treatment of numerous diseases, particularly cancers. As a natural product primarily derived from various medicinal plants in the Withania genus, withanolides have been shown in several studies to exhibit potential activities in cancer treatment. Consequently, understanding the molecular mechanism of withanolides could herald the discovery of new anticancer agents. Withanolides have been studied widely, especially in the last 20 years, and attracted the attention of numerous researchers. Currently, over 1200 withanolides have been classified, with approximately a quarter of them having been reported in the literature to be able to modulate the survival and death of cancer cells through multiple avenues. To what extent, though, has the anticancer effects of these compounds been studied? How far are they from being developed into clinical drugs? What are their potential, characteristic features, and challenges? In this review, we elaborate on the current knowledge of natural compounds belonging to this class and provide an overview of their natural sources, anticancer activity, mechanism of action, molecular targets, and implications for anticancer drug research. In addition, direct targets and clinical research to guide the design and implementation of future preclinical and clinical studies to accelerate the application of withanolides have been highlighted.
Collapse
Affiliation(s)
- Qiang Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - YongKang Yuan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Shijie Cao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Ning Kang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Feng Qiu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| |
Collapse
|
6
|
Li Y, Xiong Z, Jiang WL, Tian D, Zhou H, Hou Q, Xiao L, Zhang M, Huang L, Zhong L, Zhou L, Zeng GG. An innovative viewpoint on the existing and prospectiveness of SR-B1. Curr Probl Cardiol 2024; 49:102226. [PMID: 38040207 DOI: 10.1016/j.cpcardiol.2023.102226] [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: 11/19/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023]
Abstract
Scavenger Receptor Class B Type 1 (SR-B1), a receptor protein expressed on the cell membrane, plays a crucial role in the metabolism and transport of cholesterol and other lipids, contributing significantly to the homeostasis of lipid levels within the body. Bibliometric analysis involves the application of mathematical and statistical methods to quantitatively analyze different types of documents. It involves the analysis of structural and temporal trends in scholarly articles, coupled with the identification of subject emphasis and variations. Through a bibliometric analysis, this study examines the historical background, current research trends, and future directions in the exploration of SR-B1. By offering insights into the research status and development of SR-B1, this paper aims to assist researchers in identifying novel pathways and areas of investigation in this field of study. Following the screening process, it can be concluded that research on SR-B1 has consistently remained a topic of significant interest over the past 17 years. Interestingly, SR-B1 has recently garnered attention in areas beyond its traditional research focus, including the field of cancer. The primary objective of this review is to provide a concise and accessible overview of the development process of SR-B1 that can help readers who are not well-versed in SR-B1 research quickly grasp its key aspects. Furthermore, this review aims to offer insights and suggestions to researchers regarding potential future research directions and areas of emphasis relating to SR-B1.
Collapse
Affiliation(s)
- Yonggui Li
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhijie Xiong
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wan-Li Jiang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Dandan Tian
- School of Nursing, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Haiyou Zhou
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Qin Hou
- Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Liang Xiao
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Mengjie Zhang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Liubin Huang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Lianping Zhong
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Department of Gastroenterology, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Li Zhou
- Department of Pathology, Chongqing Public Health Medical Center, Southwest University Public Health Hospital, Chongqing, China
| | - Guang-Gui Zeng
- The Second Affiliated Hospital, Department of Digestive Internal Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China; Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; 2020 Grade Excellent Doctor Class of Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
7
|
Chen Q, Wang L, Song H, Xing W, Shi J, Li Y, Wang Z, Chen J, Xie N, Zhao W. Deficiency of SR-B1 reduced the tumor load of colitis-induced or APC min /+ -induced colorectal cancer. Cancer Med 2023; 12:19744-19757. [PMID: 37766594 PMCID: PMC10587988 DOI: 10.1002/cam4.6534] [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: 05/25/2023] [Revised: 08/03/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common tumors in the world. Cholesterol plays an important role in the pathogenesis of tumors. One of the cholesterol transporters, scavenger receptor class B type 1 (SR-B1), a multi-ligand membrane receptor protein, is expressed in the intestines which also highly expressed in various tumors. But the potential mechanism of SR-B1 in CRC development has not been reported. AIMS This study aimed to clarify the importance of SR-B1 in the development and prognosis of CRC as much as possible to provide a possible strategy in CRC treatment. MATERIALS & METHODS In this study, we used SR-B1 gene knockdown mice to study the effect of SR-B1 on colitis-induced or APCmin/+ -induced CRC. The expression of related molecules were detected through the immunohistochemistry and hematoxylin-eosin staining, western blot analysis, and Flow cytometry. The gene expression and microbiota in microenvironment of CRC mice were analyzed through eukaryotic mRNA sequencing and 16S rRNA high-throughput sequencing. RESULTS The results showed that SR-B1 knockdown reduced the tumor load of colitis-induced or APCmin/+ -induced CRC. SR-B1 knockdown improved the immune microenvironment by affecting the level of tumor-associated macrophage (TAM), mononuclear myeloid-derived suppressor cells (M-MDSCs), granulocytic myeloid-derived suppressor cells (G-MDSCs), programmed cell death-ligand 1 (PD-L1), and human leukocyte antigen class I-B (HLA-B), and also reduced the level of low-density lipoprotein receptor (LDL-R), and increased the level of ATP binding cassette transporter A1 (ABCA1) to regulate the cholesterol metabolism, and regulated the expression of related genes and intestinal microbiota. SR-B1 knockdown can also trigger the anti-CRC effect of anti-PD 1 in colitis-induced CRC. DISCUSSION SR-B1 deficiency significantly improved the immunity in tumor microenvironment of colitis-induced or APCmin/+ -induced CRC. In addition, the microbiota changes caused by SR-B1 deficiency favor improving the immune response to chemotherapeutic drugs and anti-PD1 therapy. The mechanism of action of SR-B1 deficiency on the development of CRC still needs further in-depth research. CONCLUSION This study provides a new treatment strategy for treating CRC by affecting the expression of SR-B1 in intestine.
Collapse
Affiliation(s)
- Qijun Chen
- School of Pharmaceutical SciencesCapital Medical UniversityBeijingChina
| | - Lixue Wang
- School of Pharmaceutical SciencesCapital Medical UniversityBeijingChina
| | - Hui Song
- School of Pharmaceutical SciencesCapital Medical UniversityBeijingChina
| | - Wen Xing
- School of Pharmaceutical SciencesCapital Medical UniversityBeijingChina
| | - Junfeng Shi
- School of Pharmaceutical SciencesCapital Medical UniversityBeijingChina
| | - Yudi Li
- School of Pharmaceutical SciencesCapital Medical UniversityBeijingChina
| | - Ziqian Wang
- School of Pharmaceutical SciencesCapital Medical UniversityBeijingChina
| | - Jinlong Chen
- School of Basic Medical SciencesCapital Medical UniversityBeijingChina
| | - Nan Xie
- School of Pharmaceutical SciencesCapital Medical UniversityBeijingChina
| | - Wenhua Zhao
- School of Pharmaceutical SciencesCapital Medical UniversityBeijingChina
| |
Collapse
|
8
|
Jiang C, Feng D, Zhang Y, Yang K, Hu X, Xie Q. SCAT8/miR-125b-5p axis triggers malignant progression of nasopharyngeal carcinoma through SCARB1. BMC Mol Cell Biol 2023; 24:15. [PMID: 37009875 PMCID: PMC10069050 DOI: 10.1186/s12860-023-00477-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/20/2023] [Indexed: 04/04/2023] Open
Abstract
Nasopharyngeal carcinoma is a tumor with high malignancy and poor prognosis, which severely affects the health of the patients. LncRNAs and microRNAs are crucial for the occurrence and development of nasopharyngeal carcinoma, which regulate the progression of nasopharyngeal carcinoma through the ceRNA network. SCARB1 plays an essential role in nasopharyngeal carcinoma. However, the mechanism underlying the regulation of SCARB1 in nasopharyngeal carcinoma through non-coding RNAs remains unclear. Our findings indicated that the SCAT8/miR-125b-5p axis promoted the malignant progression of nasopharyngeal carcinoma by driving the expression of SCARB1. Mechanistically, the expression of SCARB1 could be regulated by the lncRNA, SCAT8 and the microRNA, miR-125b-5p. Moreover, as a ceRNA of miR-125b-5p, SCAT8 can not only regulate the expression of SCARB1, but also regulate the malignant progression of nasopharyngeal carcinoma. Notably, our results reveal a novel ceRNA regulatory network in nasopharyngeal carcinoma, which could serve as a potential target for the diagnosis and treatment of nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Chunmao Jiang
- Department of Health Management, Daping Hospital, Army Medical University, Chongqing, 400010, China
| | - Dandan Feng
- Department of Otolaryngology Head and Neck Surgery, Daping Hospital, Army Medical University, Chongqing, 400010, China
| | - Yu Zhang
- Department of Otolaryngology Head and Neck Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Kun Yang
- Department of Health Management Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Xiaotong Hu
- Department of Health Management, Daping Hospital, Army Medical University, Chongqing, 400010, China
| | - Qian Xie
- Department of Health Management, Daping Hospital, Army Medical University, Chongqing, 400010, China.
| |
Collapse
|
9
|
Tan M, Yang S, Xu X. High-density lipoprotein cholesterol and carcinogenesis. Trends Endocrinol Metab 2023; 34:303-313. [PMID: 36973155 DOI: 10.1016/j.tem.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/27/2023] [Accepted: 02/27/2023] [Indexed: 03/29/2023]
Abstract
High-density lipoprotein cholesterol (HDLC) has been recognized to be associated with atherosclerosis. In the past few years many studies have found that HDLC is also related to tumor development and progression. Despite some opposing views, a large number of studies support a negative association between HDLC and tumor incidence. Measuring serum HDLC concentrations may facilitate assessment of the prognosis of cancer patients and provide a biomarker for tumors. However, there is a lack of molecular mechanism studies on the link between HDLC and tumors. In this review we discuss the impact of HDLC on the incidence and prognosis of cancer in different systems, as well as prospects for the prediction and treatment of cancer in the future.
Collapse
Affiliation(s)
- Meijuan Tan
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shijie Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiequn Xu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
10
|
Kazakova E, Iamshchikov P, Larionova I, Kzhyshkowska J. Macrophage scavenger receptors: Tumor support and tumor inhibition. Front Oncol 2023; 12:1096897. [PMID: 36686729 PMCID: PMC9853406 DOI: 10.3389/fonc.2022.1096897] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 12/13/2022] [Indexed: 01/08/2023] Open
Abstract
Tumor-associated macrophages (TAMs) are a heterogeneous population of myeloid cells that constitute up to 50% of the cell mass of human tumors. TAMs interact with the components of the tumor microenvironment (TME) by using scavenger receptors (SRs), a large superfamily of multifunctional receptors that recognize, internalize and transport to the endosomal/lysosomal pathway apoptotic cells, cytokines, matrix molecules, lipid modified lipoproteins and other unwanted-self ligands. In our review, we summarized state-of-the art for the role of macrophage scavenger receptors in tumor development and their significance as cancer biomarkers. In this review we focused on functional activity of TAM-expressing SRs in animal models and in patients, and summarized the data for different human cancer types about the prognostic significance of TAM-expressed SRs. We discussed the role of SRs in the regulation of cancer cell biology, cell-cell and cell-matrix interaction in TME, immune status in TME, angiogenesis, and intratumoral metabolism. Targeting of tumor-promoting SRs can be a promising therapeutic approach in anti-cancer therapy. In our review we provide evidence for both tumor supporting and tumor inhibiting functions of scavenger receptors expressed on TAMs. We focused on the key differences in the prognostic and functional roles of SRs that are specific for cancer types. We highlighted perspectives for inhibition of tumor-promoting SRs in anti-cancer therapy.
Collapse
Affiliation(s)
- Elena Kazakova
- Laboratory of translational cellular and molecular biomedicine, National Research Tomsk State University, Tomsk, Russia,Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Pavel Iamshchikov
- Laboratory of translational cellular and molecular biomedicine, National Research Tomsk State University, Tomsk, Russia,Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Irina Larionova
- Laboratory of translational cellular and molecular biomedicine, National Research Tomsk State University, Tomsk, Russia,Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia,Laboratory of Genetic Technologies, Siberian State Medical University, Tomsk, Russia
| | - Julia Kzhyshkowska
- Laboratory of translational cellular and molecular biomedicine, National Research Tomsk State University, Tomsk, Russia,Laboratory of Genetic Technologies, Siberian State Medical University, Tomsk, Russia,Institute of Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany,German Red Cross Blood Service Baden-Württemberg – Hessen, Mannheim, Germany,*Correspondence: Julia Kzhyshkowska,
| |
Collapse
|
11
|
Huang W, He Z, Cai X, Zhang J, Li W, Wang K, Zhang S. The Dual-Targeted Peptide Conjugated Probe for Depicting Residual Nasopharyngeal Carcinoma and Guiding Surgery. BIOSENSORS 2022; 12:bios12090729. [PMID: 36140113 PMCID: PMC9496553 DOI: 10.3390/bios12090729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/20/2022]
Abstract
Detecting residual nasopharyngeal carcinoma (rNPC) can be difficult because of the coexistence of occult tumours and post-chemoradiation changes, which poses a challenge for both radiologists and surgeons using current imaging methods. Currently, molecular imaging that precisely targets and visualises particular biomarkers in tumours may exceed the specificity and sensitivity of traditional imaging techniques, providing the potential to distinguish tumours from non-neoplastic lesions. Here, we synthesised a HER2/SR-BI-targeted tracer to efficiently position NPC and guide surgery in living mice. This bispecific tracer contained the following two parts: IRDye 800 CW, as an imaging reagent for both optical and optoacoustic imaging, and a fusion peptide (FY-35), as the targeting reagent. Both in vitro and in vivo tests demonstrated that the tracer had higher accumulation and longer retention (up to 48 h) in tumours than a single-targeted probe, and realised sensitive detection of tumours with a minimum size of 3.9 mm. By visualising the vascular network via a customised handheld optoacoustic scan, our intraoperative fluorescence molecular imaging system provides accurate guidance for intraoperative tumour resection. Integrating the advantages of both optical and optoacoustic scanning in an intraoperative image-guided system, this method holds promise for depicting rNPC and guiding salvage surgery.
Collapse
Affiliation(s)
- Wenhui Huang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110167, China
- Medical Imaging Center, the First Affiliated Hospital, Jinan University, Guangzhou 510630, China
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Zicong He
- Medical Imaging Center, the First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Xuekang Cai
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Jingming Zhang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Wei Li
- Medical Imaging Center, the First Affiliated Hospital, Jinan University, Guangzhou 510630, China
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Kun Wang
- CAS Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- Correspondence: (K.W.); (S.Z.); Tel.: +86-135-4459-7585 (S.Z.); +86-186-1296-5656 (K.W.)
| | - Shuixing Zhang
- Medical Imaging Center, the First Affiliated Hospital, Jinan University, Guangzhou 510630, China
- Correspondence: (K.W.); (S.Z.); Tel.: +86-135-4459-7585 (S.Z.); +86-186-1296-5656 (K.W.)
| |
Collapse
|
12
|
Pereira ELR, Feio DCA, Tavares JPL, Morikawa NM, Deus DF, Vital CG, Tavares ER, Maranhão RC. Uptake of lipid core nanoparticles by fragments of tissues collected during cerebral tumor excision surgeries: hypotheses for use in drug targeting therapy. J Neurooncol 2022; 158:413-421. [PMID: 35612697 DOI: 10.1007/s11060-022-04028-1] [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: 03/25/2022] [Accepted: 05/05/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE Malignant cerebral tumors have poor prognosis and the blood-brain barrier is a major hindrance for most drugs to reach those tumors. Lipid nanoparticles (LDE) that bind to lipoprotein receptors may carry anticancer drugs and penetrate the cells through those receptors that are overexpressed in gliomas. The aim was to investigate the in vivo uptake of LDE by human cerebral tumors. METHODS Twelve consecutive patients (4 with glioblastomas, 1 meduloblastoma, 1 primary lymphoma, 2 with non-cerebral metastases and 4 with benign tumors) scheduled for tumor excision surgery were injected intravenously, 12 h before surgery, with LDE labeled 14C-cholesterol oleate. Fragments of tumors and of normal head tissues (muscle, periosteum, dura mater) discarded by the surgeon were submitted to lipid extraction and radioactive counting. RESULTS Tumor LDE uptake (range: 10-283 d.p.m./g of tissue) was not lower than that of normal tissues (range: 20-263 d.p.m./g). Malignant tumor uptake was threefold greater than benign tumor uptake (140 ± 93 vs 46 ± 18 d.p.m./g, p < 0.05). Results show that LDE can concentrate in brain malignant tumors and may be used to carry drugs directed against those tumors. CONCLUSION As LDE was previously shown to markedly decrease drug toxicity this new therapeutic strategy should be tested in future trials.
Collapse
Affiliation(s)
- Edmundo Luís Rodrigues Pereira
- Servico de Cirurgia, Nucleo de Pesquisas em Neurooncologia, Hospital Universitario Joao de Barros Barreto, Universidade Federal do Para, Belem, Para, Brazil
| | | | - João Pojucan Lobo Tavares
- Servico de Cirurgia, Nucleo de Pesquisas em Neurooncologia, Hospital Universitario Joao de Barros Barreto, Universidade Federal do Para, Belem, Para, Brazil
| | - Natalia Megumi Morikawa
- Servico de Cirurgia, Nucleo de Pesquisas em Neurooncologia, Hospital Universitario Joao de Barros Barreto, Universidade Federal do Para, Belem, Para, Brazil
| | - Debora Fernandes Deus
- Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, Brazil
| | - Carolina Graziani Vital
- Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, Brazil
| | - Elaine Rufo Tavares
- Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, Brazil
| | - Raul Cavalcante Maranhão
- Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, Brazil. .,Faculdade de Ciencias Farmaceuticas, Universidade de Sao Paulo, São Paulo, Brazil. .,Instituto Nacional de Ciencias e Tecnologia em Fluidos Complexos (INCT-FCx), São Paulo, São Paulo, Brazil. .,Laboratório de Metabolismo e Lípides, Instituto do Coração (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), Av. Dr. Enéas de Carvalho Aguiar, 44, bloco 2, 1º subsolo, São Paulo, SP, Brazil.
| |
Collapse
|
13
|
HDL as Bidirectional Lipid Vectors: Time for New Paradigms. Biomedicines 2022; 10:biomedicines10051180. [PMID: 35625916 PMCID: PMC9138557 DOI: 10.3390/biomedicines10051180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 02/06/2023] Open
Abstract
The anti-atherogenic properties of high-density lipoproteins (HDL) have been explained mainly by reverse cholesterol transport (RCT) from peripheral tissues to the liver. The RCT seems to agree with most of the negative epidemiological correlations between HDL cholesterol levels and coronary artery disease. However, therapies designed to increase HDL cholesterol failed to reduce cardiovascular risk, despite their capacity to improve cholesterol efflux, the first stage of RCT. Therefore, the cardioprotective role of HDL may not be explained by RCT, and it is time for new paradigms about the physiological function of these lipoproteins. It should be considered that the main HDL apolipoprotein, apo AI, has been highly conserved throughout evolution. Consequently, these lipoproteins play an essential physiological role beyond their capacity to protect against atherosclerosis. We propose HDL as bidirectional lipid vectors carrying lipids from and to tissues according to their local context. Lipid influx mediated by HDL appears to be particularly important for tissue repair right on site where the damage occurs, including arteries during the first stages of atherosclerosis. In contrast, the HDL-lipid efflux is relevant for secretory cells where the fusion of intracellular vesicles drastically enlarges the cytoplasmic membrane with the potential consequence of impairment of cell function. In such circumstances, HDL could deliver some functional lipids and pick up not only cholesterol but an integral part of the membrane in excess, restoring the viability of the secretory cells. This hypothesis is congruent with the beneficial effects of HDL against atherosclerosis as well as with their capacity to induce insulin secretion and merits experimental exploration.
Collapse
|
14
|
Yu H. HDL and Scavenger Receptor Class B Type I (SRBI). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1377:79-93. [DOI: 10.1007/978-981-19-1592-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
15
|
Pandey M, Cuddihy G, Gordon JA, Cox ME, Wasan KM. Inhibition of Scavenger Receptor Class B Type 1 (SR-B1) Expression and Activity as a Potential Novel Target to Disrupt Cholesterol Availability in Castration-Resistant Prostate Cancer. Pharmaceutics 2021; 13:1509. [PMID: 34575583 PMCID: PMC8467449 DOI: 10.3390/pharmaceutics13091509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023] Open
Abstract
There have been several studies that have linked elevated scavenger receptor class b type 1 (SR-B1) expression and activity to the development and progression of castration-resistant prostate cancer (CRPC). SR-B1 facilitates the influx of cholesterol to the cell from lipoproteins in systemic circulation. This influx of cholesterol may be important for many cellular functions, including the synthesis of androgens. Castration-resistant prostate cancer tumors can synthesize androgens de novo to supplement the loss of exogenous sources often induced by androgen deprivation therapy. Silencing of SR-B1 may impact the ability of prostate cancer cells, particularly those of the castration-resistant state, to maintain the intracellular supply of androgens by removing a supply of cholesterol. SR-B1 expression is elevated in CRPC models and has been linked to poor survival of patients. The overarching belief has been that cholesterol modulation, through either synthesis or uptake inhibition, will impact essential signaling processes, impeding the proliferation of prostate cancer. The reduction in cellular cholesterol availability can impede prostate cancer proliferation through both decreased steroid synthesis and steroid-independent mechanisms, providing a potential therapeutic target for the treatment of prostate cancer. In this article, we discuss and highlight the work on SR-B1 as a potential novel drug target for CRPC management.
Collapse
Affiliation(s)
- Mitali Pandey
- Department of Urological Sciences, Faculty of Medicine, University of British Columbia, Vancouver Prostate Centre, Vancouver, BC V6T 1Z3, Canada; (M.P.); (M.E.C.)
| | - Grace Cuddihy
- College of Pharmacy and Nutrition, University of Saskatchewan, 104 Clinic Place, Saskatoon, SK S7N 2Z4, Canada;
| | - Jacob A. Gordon
- Oncology Bioscience, Oncology R&D, AstraZeneca, Boston, MA 02451, USA;
| | - Michael E. Cox
- Department of Urological Sciences, Faculty of Medicine, University of British Columbia, Vancouver Prostate Centre, Vancouver, BC V6T 1Z3, Canada; (M.P.); (M.E.C.)
| | - Kishor M. Wasan
- Department of Urological Sciences, Faculty of Medicine, University of British Columbia, Vancouver Prostate Centre, Vancouver, BC V6T 1Z3, Canada; (M.P.); (M.E.C.)
| |
Collapse
|
16
|
Luo H, Lu L, Liu N, Li Q, Yang X, Zhang Z. Curcumin loaded sub-30 nm targeting therapeutic lipid nanoparticles for synergistically blocking nasopharyngeal cancer growth and metastasis. J Nanobiotechnology 2021; 19:224. [PMID: 34320999 PMCID: PMC8317404 DOI: 10.1186/s12951-021-00966-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/19/2021] [Indexed: 01/07/2023] Open
Abstract
Systemic chemotherapy is still the primary treatment for advanced-stage nasopharyngeal carcinoma (NPC), but only limited therapeutic success has been achieved in the past decade because of drug resistance and systemic toxicity. Curcumin (Cur) is an effective alternative to chemotherapeutics because it showed remarkable therapeutic potential in the treatment of NPC. However, lack of tissue specificity and poor penetration in solid tumors are the major obstacles to effective therapy. Therefore, in this work, a self-assembled sub-30 nm therapeutic lipid nanoparticle loaded with Cur, named as Cur@α-NTP-LN, was constructed, specifically targeting scavenger receptor class B member 1 (SR-B1) and enhancing its therapeutic effects on NPC in vivo. Our results showed that Cur@α-NTP-LNs were effective and superior to free Cur on NPC cell-specific targeting, suppressing cell proliferation and inducing cell apoptosis. In vivo and ex vivo optical imaging revealed that Cur@α-NTP-LNs exerted high targeting efficiency, specifically accumulating in NPC xenograft tumors and delivering Cur into the tumor center after systemic administration. Furthermore, Cur@α-NTP-LNs exhibited a remarkable inhibitory effect on the growth of NPC subcutaneous tumors, with over 71 and 47% inhibition compared to Cur- and α-NTP-LNs-treated groups, respectively. In addition, Cur@α-NTP-LNs almost blocked NPC metastasis in a lung metastasis model of NPC and significantly improved the survival rate. Thus, the sub-30 nm Cur@α-NTP-LNs enhanced the solubility of Cur and demonstrated the ability of targeted Cur delivery into the center of the solid NPC tumor, performing synergistic inhibitory effects on the growth of NPC tumor and its metastasis with high efficiency. ![]()
Collapse
Affiliation(s)
- Haiming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China. .,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Lisen Lu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Ni Liu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Qingqing Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xiaoquan Yang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhihong Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, China. .,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
17
|
Gupta A, Sharma R, Kuche K, Jain S. Exploring the therapeutic potential of the bioinspired reconstituted high density lipoprotein nanostructures. Int J Pharm 2021; 596:120272. [DOI: 10.1016/j.ijpharm.2021.120272] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/20/2020] [Accepted: 12/26/2020] [Indexed: 12/17/2022]
|
18
|
Role of cholesterol metabolism in the anticancer pharmacology of selective estrogen receptor modulators. Semin Cancer Biol 2020; 73:101-115. [PMID: 32931953 DOI: 10.1016/j.semcancer.2020.08.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/13/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022]
Abstract
Selective estrogen receptor modulators (SERMs) are a class of compounds that bind to estrogen receptors (ERs) and possess estrogen agonist or antagonist actions in different tissues. As such, they are widely used drugs. For instance, tamoxifen, the most prescribed SERM, is used to treat ERα-positive breast cancer. Aside from their therapeutic targets, SERMs have the capacity to broadly affect cellular cholesterol metabolism and handling, mainly through ER-independent mechanisms. Cholesterol metabolism reprogramming is crucial to meet the needs of cancer cells, and different key processes involved in cholesterol homeostasis have been associated with cancer progression. Therefore, the effects of SERMs on cholesterol homeostasis may be relevant to carcinogenesis, either by contributing to the anticancer efficacy of these compounds or, conversely, by promoting resistance to treatment. Understanding these aspects of SERMs actions could help to design more efficacious therapies. Herein we review the effects of SERMs on cellular cholesterol metabolism and handling and discuss their potential in anticancer pharmacology.
Collapse
|
19
|
Lu L, Qi S, Chen Y, Luo H, Huang S, Yu X, Luo Q, Zhang Z. Targeted immunomodulation of inflammatory monocytes across the blood-brain barrier by curcumin-loaded nanoparticles delays the progression of experimental autoimmune encephalomyelitis. Biomaterials 2020; 245:119987. [DOI: 10.1016/j.biomaterials.2020.119987] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 02/08/2023]
|
20
|
Chuang ST, Cruz S, Narayanaswami V. Reconfiguring Nature's Cholesterol Accepting Lipoproteins as Nanoparticle Platforms for Transport and Delivery of Therapeutic and Imaging Agents. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E906. [PMID: 32397159 PMCID: PMC7279153 DOI: 10.3390/nano10050906] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 12/13/2022]
Abstract
Apolipoproteins are critical structural and functional components of lipoproteins, which are large supramolecular assemblies composed predominantly of lipids and proteins, and other biomolecules such as nucleic acids. A signature feature of apolipoproteins is the preponderance of amphipathic α-helical motifs that dictate their ability to make extensive non-covalent inter- or intra-molecular helix-helix interactions in lipid-free states or helix-lipid interactions with hydrophobic biomolecules in lipid-associated states. This review focuses on the latter ability of apolipoproteins, which has been capitalized on to reconstitute synthetic nanoscale binary/ternary lipoprotein complexes composed of apolipoproteins/peptides and lipids that mimic native high-density lipoproteins (HDLs) with the goal to transport drugs. It traces the historical development of our understanding of these nanostructures and how the cholesterol accepting property of HDL has been reconfigured to develop them as drug-loading platforms. The review provides the structural perspective of these platforms with different types of apolipoproteins and an overview of their synthesis. It also examines the cargo that have been loaded into the core for therapeutic and imaging purposes. Finally, it lays out the merits and challenges associated with apolipoprotein-based nanostructures with a future perspective calling for a need to develop "zip-code"-based delivery for therapeutic and diagnostic applications.
Collapse
Affiliation(s)
| | | | - Vasanthy Narayanaswami
- Department of Chemistry and Biochemistry, California State University, Long Beach, 1250 Bellflower Blvd, Long Beach, CA 90840, USA; (S.T.C.); (S.C.)
| |
Collapse
|
21
|
Scheetz LM, Yu M, Li D, Castro MG, Moon JJ, Schwendeman A. Synthetic HDL Nanoparticles Delivering Docetaxel and CpG for Chemoimmunotherapy of Colon Adenocarcinoma. Int J Mol Sci 2020; 21:ijms21051777. [PMID: 32150841 PMCID: PMC7084365 DOI: 10.3390/ijms21051777] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/19/2020] [Accepted: 03/03/2020] [Indexed: 12/13/2022] Open
Abstract
Colon carcinomas comprise over two-thirds of all colorectal cancers with an overall 5-year survival rate of 64%, which rapidly decreases to 14% when the cancer becomes metastatic. Depending on the stage of colon carcinoma at diagnosis, patients can undergo surgery to attempt complete tumor resection or move directly to chemotherapy with one or a combination of drugs. As with most cancers, colon carcinomas do not always respond to chemotherapies, so targeted therapies and immunotherapies have been developed to aid chemotherapy. We report the development of a local combination therapy for colon carcinoma whereby chemo- and immunotherapeutic entities are delivered intratumorally to maximize efficacy and minimize off-target side effects. A hydrophobic chemotherapeutic agent, docetaxel (DTX), and cholesterol-modified Toll-like receptor 9 (TLR9) agonist CpG (cho-CpG) oligonucleotide are co-loaded in synthetic HDL (sHDL) nanodiscs. In vivo survival analysis of MC-38 tumor-bearing mice treated intratumorally with DTX-sHDL/CpG (median survival; MS = 43 days) showed significant improvement in overall survival compared to mice treated with single agents, free DTX (MS = 23 days, p < 0.0001) or DTX-sHDL (MS = 28 days, p < 0.0001). Two of seven mice treated with DTX-sHDL/CpG experienced complete tumor regression. None of the mice experienced any systemic toxicity as indicated by body weight maintenance and normal serum enzyme and protein levels. In summary, we have demonstrated that chemo- and immunotherapies can be co-loaded into sHDLs, delivered locally to the tumor, and can be used to improve survival outcomes significantly compared to chemotherapy alone.
Collapse
Affiliation(s)
- Lindsay M. Scheetz
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA; (L.M.S.); (M.Y.); (D.L.)
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Minzhi Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA; (L.M.S.); (M.Y.); (D.L.)
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dan Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA; (L.M.S.); (M.Y.); (D.L.)
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - María G. Castro
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA;
- Department of Cell and Developmental Biology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - James J. Moon
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA; (L.M.S.); (M.Y.); (D.L.)
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Correspondence: (J.J.M.); (A.S.); Tel.: +734-763-4056 (A.S.)
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA; (L.M.S.); (M.Y.); (D.L.)
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Correspondence: (J.J.M.); (A.S.); Tel.: +734-763-4056 (A.S.)
| |
Collapse
|
22
|
Wang D, Huang J, Gui T, Yang Y, Feng T, Tzvetkov NT, Xu T, Gai Z, Zhou Y, Zhang J, Atanasov AG. SR-BI as a target of natural products and its significance in cancer. Semin Cancer Biol 2020; 80:18-38. [PMID: 31935456 DOI: 10.1016/j.semcancer.2019.12.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/25/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023]
Abstract
Scavenger receptor class B type I (SR-BI) protein is an integral membrane glycoprotein. SR-BI is emerging as a multifunctional protein, which regulates autophagy, efferocytosis, cell survival and inflammation. It is well known that SR-BI plays a critical role in lipoprotein metabolism by mediating cholesteryl esters selective uptake and the bi-directional flux of free cholesterol. Recently, SR-BI has also been identified as a potential marker for cancer diagnosis, prognosis, or even a treatment target. Natural products are a promising source for the discovery of new drug leads. Multiple natural products were identified to regulate SR-BI protein expression. There are still a number of challenges in modulating SR-BI expression in cancer and in using natural products for modulation of such protein expression. In this review, our purpose is to discuss the relationship between SR-BI protein and cancer, and the molecular mechanisms regulating SR-BI expression, as well as to provide an overview of natural products that regulate SR-BI expression.
Collapse
Affiliation(s)
- Dongdong Wang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Fei Shan Jie 32, 550003, Guiyang, China
| | - Jiansheng Huang
- Department of Medicine, Vanderbilt University Medical Center, 318 Preston Research Building, 2200 Pierce Avenue, Nashville, Tennessee, 37232, USA
| | - Ting Gui
- Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yaxin Yang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Fei Shan Jie 32, 550003, Guiyang, China
| | - Tingting Feng
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Huaxi university town, 550025, Guiyang, China
| | - Nikolay T Tzvetkov
- Department of Biochemical Pharmacology and Drug Design, Institute of Molecular Biology "Roumen Tsanev", Bulgarian Academy of Sciences, 21 Acad. G. Bonchev Str., 1113 Sofia, Bulgaria
| | - Tao Xu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Fei Shan Jie 32, 550003, Guiyang, China
| | - Zhibo Gai
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ying Zhou
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Huaxi university town, 550025, Guiyang, China.
| | - Jingjie Zhang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Fei Shan Jie 32, 550003, Guiyang, China.
| | - Atanas G Atanasov
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, 05-552, Jastrzębiec, Poland; Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090, Vienna, Austria; Institute of Neurobiology, Bulgarian Academy of Sciences, 23 Acad. G. Bonchev Str., 1113 Sofia, Bulgaria; Ludwig Boltzmann Institute for Digital Health and Patient Safety, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria.
| |
Collapse
|
23
|
|
24
|
Scheetz L, Park KS, Li Q, Lowenstein PR, Castro MG, Schwendeman A, Moon JJ. Engineering patient-specific cancer immunotherapies. Nat Biomed Eng 2019; 3:768-782. [PMID: 31406259 PMCID: PMC6783331 DOI: 10.1038/s41551-019-0436-x] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 07/03/2019] [Indexed: 02/06/2023]
Abstract
Research into the immunological processes implicated in cancer has yielded a basis for the range of immunotherapies that are now considered the fourth pillar of cancer treatment (alongside surgery, radiotherapy and chemotherapy). For some aggressive cancers, such as advanced non-small-cell lung carcinoma, combination immunotherapies have resulted in unprecedented treatment efficacy for responding patients, and have become frontline therapies. Individualized immunotherapy, enabled by the identification of patient-specific mutations, neoantigens and biomarkers, and facilitated by advances in genomics and proteomics, promises to broaden the responder patient population. In this Perspective, we give an overview of immunotherapies leveraging engineering approaches, including the design of biomaterials, delivery strategies and nanotechnology solutions, for the realization of individualized cancer treatments such as nanoparticle vaccines customized with neoantigens, cell therapies based on patient-derived dendritic cells and T cells, and combinations of theranostic strategies. Developments in precision cancer immunotherapy will increasingly rely on the adoption of engineering principles.
Collapse
Affiliation(s)
- Lindsay Scheetz
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Kyung Soo Park
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Qiao Li
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
25
|
Apolipoprotein A-I (ApoA-I), Immunity, Inflammation and Cancer. Cancers (Basel) 2019; 11:cancers11081097. [PMID: 31374929 PMCID: PMC6721368 DOI: 10.3390/cancers11081097] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 07/25/2019] [Accepted: 07/30/2019] [Indexed: 12/21/2022] Open
Abstract
Apolipoprotein A-I (ApoA-I), the major protein component of high-density lipoproteins (HDL) is a multifunctional protein, involved in cholesterol traffic and inflammatory and immune response regulation. Many studies revealing alterations of ApoA-I during the development and progression of various types of cancer suggest that serum ApoA-I levels may represent a useful biomarker contributing to better estimation of cancer risk, early cancer diagnosis, follow up, and prognosis stratification of cancer patients. In addition, recent in vitro and animal studies disclose a more direct, tumor suppressive role of ApoA-I in cancer pathogenesis, which involves anti-inflammatory and immune-modulatory mechanisms. Herein, we review recent epidemiologic, clinicopathologic, and mechanistic studies investigating the role of ApoA-I in cancer biology, which suggest that enhancing the tumor suppressive activity of ApoA-I may contribute to better cancer prevention and treatment.
Collapse
|
26
|
Mimetic sHDL nanoparticles: A novel drug-delivery strategy to target triple-negative breast cancer. Surgery 2019; 166:1168-1175. [PMID: 31371177 DOI: 10.1016/j.surg.2019.06.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 05/31/2019] [Accepted: 06/10/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Withanolides are naturally derived heat shock protein 90 inhibitors that are potent in preclinical models of triple negative breast cancers. Conjugation to synthetic high-density lipoprotein nanoparticles improves solubility and targets delivery to the scavenger receptor B1. Triple negative breast cancers highly overexpress the scavenger receptor B1, and we hypothesize that encapsulation of the novel withalongolide A 4,19,27-triacetate by synthetic high-density lipoprotein will have enhanced efficacy against triple negative breast cancers in vivo. METHODS Validated human triple negative breast cancer cell lines were evaluated for the scavenger receptor B1 expression by quantitative polymerase chain reaction and Western blot. Withalongolide A 4,19,27-triacetate inhibitory concentration50 values were obtained using CellTiter-Glo assays (Promega, Madison, WI, USA). The scavenger receptor B1-mediated drug uptake was evaluated in vitro under fluorescence microscopy and in vivo with IVIS imaging of mouse xenografts (MD-MBA-468LN). To evaluate drug efficacy, mice were treated with synthetic high-density lipoprotein alone, withalongolide A 4,19,27-triacetate alone, withalongolide A 4,19,27-triacetate synthetic high-density lipoprotein, and chemotherapy or Prussian blue stain (control). RESULTS Triple negative breast cancer cell lines had greater scavenger receptor B1 expression by quantitative polymerase chain reaction and Western blot versus controls. Fluorescent-labeled synthetic high-density lipoprotein uptake was scavenger receptor B1-mediated in vitro, and in vivo tumor uptake using IVIS imaging demonstrated significantly increased tumor radiant efficiency versus control. Inhibitory concentration50 for withalongolide A 4,19,27-triacetate-treated cells with or without synthetic high-density lipoprotein encapsulation were 70-fold to 200-fold more potent than synthetic high-density lipoprotein alone. In triple negative breast cancer mouse xenografts, treatment with synthetic high-density lipoprotein withalongolide A 4,19,27-triacetate resulted in a 54% decrease in tumor volume compared with the control or with synthetic high-density lipoprotein alone. CONCLUSION The synthetic high-density lipoprotein withalongolide A 4,19,27-triacetate nanoconjugates are potent against triple negative breast cancers and show improved scavenger receptor B1-mediated targeting. Treatment with synthetic high-density lipoprotein-encapsulated withalongolide A 4,19,27-triacetate is able to significantly decrease the growth of tumor in mice compared with the control and has better efficacy than the current standard of care, warranting further evaluation as a novel therapeutic agent.
Collapse
|
27
|
Liu Q, Li Z, Shang H, Zhang Q, Wang X, Zhang Y, Wang Y, Li Q, Li C, Liu C, Li F. Scavenger Receptor Class B Type 1 (SR-B1) being a Potential Biomarker for the Diagnosis of Liposarcoma and Associated with the Degree of Differentiation of Liposarcomas. J Cancer 2019; 10:4326-4332. [PMID: 31413752 PMCID: PMC6691705 DOI: 10.7150/jca.31730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 05/05/2019] [Indexed: 01/01/2023] Open
Abstract
Background: Soft tissue sarcomas include multiple histological subtypes and are highly aggressive. Moreover, SR-B1 is associated with malignant behavior and poor prognosis in a variety of cancers. However, there have been no attempts to assess whether SR-B1 expression in soft tissue sarcoma. We aimed to detect the expression levels of the SR-B1 protein in soft tissue sarcoma. Methods: We assessed SR-B1 expression via immunohistochemistry and tissue microarrays in 107 soft tissue sarcomas with 4 phenotypes: 26 liposarcomas, 18 Ewing's sarcomas, 20 rhabdomyosarcomas and 43 leiomyosarcomas. Results: Tumor cell SR-B1 expression was seen in 18/26 (69.2%) liposarcomas, 1/18 (5.55%) Ewing's sarcomas, 1/20 (5.00%) rhabdomyosarcomas, 2/43 (4.70%) leiomyosarcomas and was stained in the cell membrane. We found that SR-B1 expression in liposarcomas (18/26) was significantly higher than that in non-lipomatous sarcomas (4/77) (χ2 = 49.811, p = 0.000) and in well-differentiated liposarcoma (13/15) was significantly higher than that in dedifferentiated liposarcoma (5/11) (p = 0.038). No significant correlation was found between SR-B1 and gender, nationality, size and tumor location (p > 0.05), but it was significantly associated with ages (χ2 = 11.426, p = 0.001) and sarcoma phenotypes (χ2 = 49.817, p = 0.000). Conclusion: Our findings highlight the highly expression of SR-B1 in liposarcomas. SR-B1 may be a potential biomarker for the diagnosis of liposarcoma and may indicate the degree of differentiation of liposarcomas.
Collapse
Affiliation(s)
- Qianqian Liu
- Shihezi University, Shihezi 832002, Xinjiang, P.R. China. a. Department of Pathology, School of Medicine. b. The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education. c. The First Affiliated Hospital, School of Medicine
| | - Zhenzhen Li
- Shihezi University, Shihezi 832002, Xinjiang, P.R. China. a. Department of Pathology, School of Medicine. b. The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education. c. The First Affiliated Hospital, School of Medicine
| | - Hao Shang
- Shihezi University, Shihezi 832002, Xinjiang, P.R. China. a. Department of Pathology, School of Medicine. b. The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education. c. The First Affiliated Hospital, School of Medicine
| | - Qiaochu Zhang
- Shihezi University, Shihezi 832002, Xinjiang, P.R. China. a. Department of Pathology, School of Medicine. b. The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education. c. The First Affiliated Hospital, School of Medicine
| | - Xiaomeng Wang
- Shihezi University, Shihezi 832002, Xinjiang, P.R. China. a. Department of Pathology, School of Medicine. b. The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education. c. The First Affiliated Hospital, School of Medicine
| | - Yangyang Zhang
- Shihezi University, Shihezi 832002, Xinjiang, P.R. China. a. Department of Pathology, School of Medicine. b. The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education. c. The First Affiliated Hospital, School of Medicine
| | - Yang Wang
- Shihezi University, Shihezi 832002, Xinjiang, P.R. China. a. Department of Pathology, School of Medicine. b. The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education. c. The First Affiliated Hospital, School of Medicine
| | - Qianru Li
- Shihezi University, Shihezi 832002, Xinjiang, P.R. China. a. Department of Pathology, School of Medicine. b. The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education. c. The First Affiliated Hospital, School of Medicine
| | - Chunsen Li
- Shihezi University, Shihezi 832002, Xinjiang, P.R. China. a. Department of Pathology, School of Medicine. b. The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education. c. The First Affiliated Hospital, School of Medicine
| | - Chunxia Liu
- Shihezi University, Shihezi 832002, Xinjiang, P.R. China. a. Department of Pathology, School of Medicine. b. The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education. c. The First Affiliated Hospital, School of Medicine
| | - Feng Li
- Shihezi University, Shihezi 832002, Xinjiang, P.R. China. a. Department of Pathology, School of Medicine. b. The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education. c. The First Affiliated Hospital, School of Medicine.,Department of Pathology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P. R. China
| |
Collapse
|
28
|
Sharma B, Agnihotri N. Role of cholesterol homeostasis and its efflux pathways in cancer progression. J Steroid Biochem Mol Biol 2019; 191:105377. [PMID: 31063804 DOI: 10.1016/j.jsbmb.2019.105377] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/09/2019] [Accepted: 05/04/2019] [Indexed: 12/27/2022]
Abstract
Tumor cells show high avidity for cholesterol in order to support their inherent nature to divide and proliferate. This results in the rewiring of cholesterol homeostatic pathways by influencing not only de novo synthesis but also uptake or efflux pathways of cholesterol. Recent findings have pointed towards the importance of cholesterol efflux in tumor pathogenesis. Cholesterol efflux is the first and foremost step in reverse cholesterol transport and any perturbation in this pathway may lead to the accumulation of intracellular cholesterol, thereby altering the cellular equilibrium. This review addresses the different mechanisms of cholesterol efflux from the cell and highlights their role and regulation in context to tumor development. There are four different routes by which cholesterol can be effluxed from the cell namely, 1) passive diffusion of cholesterol to mature HDL particles, 2) SR-B1 mediated facilitated diffusion, 3) Active efflux to apo A1 via ABCA1 and 4) ABCG1 mediated efflux to mature HDL. These molecular players facilitating cholesterol efflux are engaged in a complex interplay with different signaling pathways. Thus, an understanding of the efflux pathways, their regulation and cross-talk with signaling molecules may provide novel prognostic markers and therapeutic targets to combat the onset of carcinogenesis.
Collapse
Affiliation(s)
- Bhoomika Sharma
- Department of Biochemistry, BMS-Block II, Panjab University, Sector-25, Chandigarh, 160014, India.
| | - Navneet Agnihotri
- Department of Biochemistry, BMS-Block II, Panjab University, Sector-25, Chandigarh, 160014, India.
| |
Collapse
|
29
|
Sharma B, Gupta V, Dahiya D, Kumar H, Vaiphei K, Agnihotri N. Clinical relevance of cholesterol homeostasis genes in colorectal cancer. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1314-1327. [PMID: 31202724 DOI: 10.1016/j.bbalip.2019.06.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/31/2019] [Accepted: 06/10/2019] [Indexed: 12/12/2022]
Abstract
Accumulation of cholesterol is a well-known feature in cancer. Preclinical studies suggest the contribution of various cholesterol regulators in CRC. However, their clinical relevance remains poorly understood. The aim of the present study is to evaluate the expression of these modulators in CRC and elucidate their diagnostic and prognostic value. mRNA levels of HMGCR, SREBF2, NR1H3 and NR1H2 were downregulated in tumors in local and TCGA cohort. The expression of LDLR, ABCA1 and SCARB1 was not consistent in the two cohorts. Western Blot analysis showed the increased levels of LDLR and reduced levels of LXR in early stage patients. Tumoral SREBP2 levels were enhanced in early stage whereas decreased in late stage. The individual expression of HMGCR, SREBF2, NR1H3 and NR1H2 did not have the potential to be used as independent prognostic marker, however, the combined expression of these genes associated with poor clinical outcome independent of lymph node metastasis, distant metastasis and advanced stage. This work sheds light on deregulation of cholesterol uptake and efflux pathways and provides novel leads in the development of biomarkers and therapeutic regimens that can detect and target CRC at initial stages.
Collapse
Affiliation(s)
- Bhoomika Sharma
- Department of Biochemistry, Panjab University, Sector-25, Chandigarh 160014, India
| | - Vikas Gupta
- Department of General Surgery, Post Graduate Institute of Medical Education & Research (PGIMER),Sector 12, Chandigarh 160012, India
| | - Divya Dahiya
- Department of General Surgery, Post Graduate Institute of Medical Education & Research (PGIMER),Sector 12, Chandigarh 160012, India
| | - Hemanth Kumar
- Department of General Surgery, Post Graduate Institute of Medical Education & Research (PGIMER),Sector 12, Chandigarh 160012, India
| | - Kim Vaiphei
- Department of Histopathology, Post Graduate Institute of Medical Education & Research (PGIMER), Sector 12, Chandigarh 160012, India.
| | - Navneet Agnihotri
- Department of Biochemistry, Panjab University, Sector-25, Chandigarh 160014, India.
| |
Collapse
|
30
|
Xu G, Qian Y, Zheng H, Qiao S, Yan D, Lu L, Wu L, Yang X, Luo Q, Zhang Z. Long-Distance Tracing of the Lymphatic System with a Computed Tomography/Fluorescence Dual-Modality Nanoprobe for Surveying Tumor Lymphatic Metastasis. Bioconjug Chem 2019; 30:1199-1209. [DOI: 10.1021/acs.bioconjchem.9b00144] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Guoqiang Xu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Yuan Qian
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Hao Zheng
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Sha Qiao
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Dongmei Yan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Lisen Lu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Liujuan Wu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Xiaoquan Yang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Qingming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Zhihong Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| |
Collapse
|
31
|
Kinslechner K, Schütz B, Pistek M, Rapolter P, Weitzenböck HP, Hundsberger H, Mikulits W, Grillari J, Röhrl C, Hengstschläger M, Stangl H, Mikula M. Loss of SR-BI Down-Regulates MITF and Suppresses Extracellular Vesicle Release in Human Melanoma. Int J Mol Sci 2019; 20:E1063. [PMID: 30823658 PMCID: PMC6429474 DOI: 10.3390/ijms20051063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 02/22/2019] [Accepted: 02/26/2019] [Indexed: 02/06/2023] Open
Abstract
Melanoma is a skin tumor with a high tendency for metastasis and thus is one of the deadliest cancers worldwide. Here, we investigated the expression of the scavenger receptor class B type 1 (SR-BI), a high-density lipoprotein (HDL) receptor, and tested for its role in melanoma pigmentation as well as extracellular vesicle release. We first analyzed the expression of SR-BI in patient samples and found a strong correlation with MITF expression as well as with the melanin synthesis pathway. Hence, we asked whether SR-BI could also play a role for the secretory pathway in metastatic melanoma cells. Interestingly, gain- and loss-of-function of SR-BI revealed regulation of the proto-oncogene MET. In line, SR-BI knockdown reduced expression of the small GTPase RABB22A, the ESCRT-II protein VPS25, and SNAP25, a member of the SNARE complex. Accordingly, reduced overall extracellular vesicle generation was detected upon loss of SR-BI. In summary, SR-BI expression in human melanoma enhances the formation and transport of extracellular vesicles, thereby contributing to the metastatic phenotype. Therapeutic targeting of SR-BI would not only interfere with cholesterol uptake, but also with the secretory pathway, therefore suppressing a key hallmark of the metastatic program.
Collapse
Affiliation(s)
- Katharina Kinslechner
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, 1090 Vienna, Austria.
| | - Birgit Schütz
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, 1090 Vienna, Austria.
| | - Martina Pistek
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, 1090 Vienna, Austria.
| | - Philipp Rapolter
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, 1090 Vienna, Austria.
| | - Hans P Weitzenböck
- Medical and Pharmaceutical Biotechnology, IMC University of Applied Sciences, 3500 Krems, Austria.
| | - Harald Hundsberger
- Medical and Pharmaceutical Biotechnology, IMC University of Applied Sciences, 3500 Krems, Austria.
| | - Wolfgang Mikulits
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria.
| | - Johannes Grillari
- Department of Biotechnology, BOKU -University of Natural Resources and Life Sciences, 1190 Vienna, Austria.
| | - Clemens Röhrl
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, 1090 Vienna, Austria.
| | - Markus Hengstschläger
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, 1090 Vienna, Austria.
| | - Herbert Stangl
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, 1090 Vienna, Austria.
| | - Mario Mikula
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|
32
|
Ceres K, Fitzgerald H, Quiznon KS, McDonough S, Behling-Kelly E. Immunohistochemical Labeling of Low-Density Lipoprotein Receptor and Scavenger Receptor Class B Type 1 Are Increased in Canine Lymphoma. Front Vet Sci 2019; 5:340. [PMID: 30687727 PMCID: PMC6336922 DOI: 10.3389/fvets.2018.00340] [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: 10/16/2018] [Accepted: 12/18/2018] [Indexed: 12/11/2022] Open
Abstract
Altered lipid metabolism is a well-documented hallmark of neoplastic transformation and impacts disease progression. Two major lipoprotein receptors, the low-density lipoprotein receptor (LDL-R) and scavenger receptor class B, type 1 (SR-BI) are overexpressed in a number of cancer types in people. These receptors serve to deliver cholesterol to the tumor cells and have been used to target drug therapies. In this study, we performed a retrospective analysis of LDL-R and SR-B1 expression in canine lymphoma using archived formalin-fixed tissue samples. Cases were immunophenotyped and classified according to World Health Organization (WHO) standards prior to immunostaining for the LDL_R and SR-B1. A total of 45 cases were evaluated; 21 high grade B (HGB), 11 low grade B (LGB), 7 high grade T (HGT), and 6 low grade T (LGT) lymphomas. One sided Wilcoxon rank sum tests were used to compare staining intensity between neoplastic and hyperplastic lymphoid tissue. The relationships between histological score and tumor grade and score and stage at presentation were assessed using non-parametric Kruskal-Wallis tests. Neoplastic lymphoid tissue expressed higher levels of both receptors compared to reactive lymph nodes. Median LDL-R score was 85.0 (interquartile range = 101.7), Median SR-B1 score was 209.0 (interquartile range 105.2). No relationship between LDL-R or SR-B1 staining score and tumor grade or phenotype was found. Serum cholesterol concentration was compared between dogs with high and low grade tumors using a two sample T-test, and correlations between cholesterol concentration and histological score, and between the score for the two receptors were determined using a Spearman correlation. The high expression level of these lipoprotein receptors on most of the tumors could underlie the lack of relationship between score and tumor grade. The overexpression of LDL-R and SR-B1 in canine lymphoma holds therapeutic potential particularly in dogs that overexpress one or both of these receptors, and this warrants further investigation.
Collapse
Affiliation(s)
- Kristina Ceres
- Department of Population Medicine and Diagnostic Services, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Halle Fitzgerald
- College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, United States
| | | | - Sean McDonough
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Erica Behling-Kelly
- Department of Population Medicine and Diagnostic Services, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| |
Collapse
|
33
|
Ganjali S, Ricciuti B, Pirro M, Butler AE, Atkin SL, Banach M, Sahebkar A. High-Density Lipoprotein Components and Functionality in Cancer: State-of-the-Art. Trends Endocrinol Metab 2019; 30:12-24. [PMID: 30473465 DOI: 10.1016/j.tem.2018.10.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 01/05/2023]
Abstract
Cancer is the second leading cause of death in western countries, and thus represents a major global public health issue. Whilst it is well-recognized that diet, obesity, and smoking are risk factors for cancer, the role of low levels of high-density lipoprotein cholesterol (HDL-C) in cancer is less well appreciated. Conflicting evidence suggests that serum HDL-C levels may be either positively or negatively associated with cancer incidence and mortality. Such disparate associations are supported in part by the multitude of high-density lipoprotein (HDL) functions that can all have an impact on cancer cell biology. The aim of this review is to provide a comprehensive overview of the crosstalk between HDLs and cancer, focusing on the molecular mechanisms underlying this association.
Collapse
Affiliation(s)
- Shiva Ganjali
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Biagio Ricciuti
- Department of Medical Oncology, S. Maria della Misericordia Hospital, Perugia, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Perugia, Italy
| | - Alexandra E Butler
- Diabetes Research Center, Qatar Biomedical Research Institute, Doha, Qatar
| | | | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, Lodz, Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
34
|
Morin EE, Li XA, Schwendeman A. HDL in Endocrine Carcinomas: Biomarker, Drug Carrier, and Potential Therapeutic. Front Endocrinol (Lausanne) 2018; 9:715. [PMID: 30555417 PMCID: PMC6283888 DOI: 10.3389/fendo.2018.00715] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 11/12/2018] [Indexed: 12/14/2022] Open
Abstract
High-density lipoprotein (HDL) have long been studied for their protective role against cardiovascular diseases, however recently relationship between HDL and cancer came into focus. Several epidemiological studies have shown an inverse correlation between HDL-cholesterol (HDL-C) and cancer risk, and some have even implied that HDL-C can be used as a predictive measure for survival prognosis in for specific sub-population of certain types of cancer. HDL itself is an endogenous nanoparticle capable of removing excess cholesterol from the periphery and returning it to the liver for excretion. One of the main receptors for HDL, scavenger receptor type B-I (SR-BI), is highly upregulated in endocrine cancers, notably due to the high demand for cholesterol by cancer cells. Thus, the potential to exploit administration of cholesterol-free reconstituted or synthetic HDL (sHDL) to deplete cholesterol in endocrine cancer cell and stunt their growth of use chemotherapeutic drug loaded sHDL to target payload delivery to cancer cell has become increasingly attractive. This review focuses on the role of HDL and HDL-C in cancer and application of sHDLs as endocrine cancer therapeutics.
Collapse
Affiliation(s)
- Emily E. Morin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, United States
| | - Xiang-An Li
- Department of Physiology, Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
35
|
Raut S, Mooberry L, Sabnis N, Garud A, Dossou AS, Lacko A. Reconstituted HDL: Drug Delivery Platform for Overcoming Biological Barriers to Cancer Therapy. Front Pharmacol 2018; 9:1154. [PMID: 30374303 PMCID: PMC6196266 DOI: 10.3389/fphar.2018.01154] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022] Open
Abstract
Drug delivery to malignant tumors is limited by several factors, including off-target toxicities and suboptimal benefits to cancer patient. Major research efforts have been directed toward developing novel technologies involving nanoparticles (NPs) to overcome these challenges. Major obstacles, however, including, opsonization, transport across cancer cell membranes, multidrug-resistant proteins, and endosomal sequestration of the therapeutic agent continue to limit the efficiency of cancer chemotherapy. Lipoprotein-based drug delivery technology, "nature's drug delivery system," while exhibits highly desirable characteristics, it still needs substantial investment from private/government stakeholders to promote its eventual advance to the bedside. Consequently, this review focuses specifically on the synthetic (reconstituted) high-density lipoprotein rHDL NPs, evaluating their potential to overcome specific biological barriers and the challenges of translation toward clinical utilization and commercialization. This highly robust drug transport system provides site-specific, tumor-selective delivery of anti-cancer agents while reducing harmful off-target effects. Utilizing rHDL NPs for anti-cancer therapeutics and tumor imaging revolutionizes the future strategy for the management of a broad range of cancers and other diseases.
Collapse
Affiliation(s)
- Sangram Raut
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Linda Mooberry
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Nirupama Sabnis
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Ashwini Garud
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Akpedje Serena Dossou
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Andras Lacko
- Lipoprotein Drug Delivery Research Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| |
Collapse
|
36
|
Muresan XM, Sticozzi C, Belmonte G, Cervellati F, Ferrara F, Lila MA, Valacchi G. SR-B1 involvement in keratinocytes in vitro wound closure. Arch Biochem Biophys 2018; 658:1-6. [PMID: 30240595 DOI: 10.1016/j.abb.2018.09.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/16/2018] [Accepted: 09/17/2018] [Indexed: 12/17/2022]
Abstract
Skin represents the most extended organ of human body, having as main function the protection of our body from outdoor stressors. Its protective ability is compromised when the skin is disrupted as a consequence of mechanical insults. For this purpose, cutaneous tissue is equipped with an efficient and fine mechanism involved in repairing the wounded area. Among the numerous players that take part in the wound healing process, SR-B1 has been recently shown to have a role in keratinocyte re-epithelialization. SR-B1 is a mediator of cholesterol uptake from HDLs, whereas it is implicated in other cellular processes such as vitamins absorption, vesicle trafficking or pathogen identification. The aim of this study was to investigate the mechanisms involved in SR-B1 role in skin wound closure. Our in vitro data demonstrated that SR-B1 influenced keratinocyte proliferation and migration through a downregulation of nuclear cyclin D1 levels and active MMP9 expression respectively possibly in an NF-kB-dependent mechanism. In addition, SR-B1 was also able to modulate keratinocyte morphology into a pro-migratory cytoskeleton rearrangement. The present in vitro study suggests a new role of SRB1 as a possible new key player in cutaneous wound healing mechanism.
Collapse
Affiliation(s)
- Ximena M Muresan
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Claudia Sticozzi
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Giuseppe Belmonte
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Franco Cervellati
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Francesca Ferrara
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Mary Ann Lila
- Plants for Human Health Institute, Animal Sciences Dept., NC Research Campus, NC State University, NC, USA
| | - Giuseppe Valacchi
- Dept. Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy; Plants for Human Health Institute, Animal Sciences Dept., NC Research Campus, NC State University, NC, USA.
| |
Collapse
|
37
|
Celhay O, Bousset L, Guy L, Kemeny JL, Leoni V, Caccia C, Trousson A, Damon-Soubeyrant C, De Haze A, Sabourin L, Godfraind C, de Joussineau C, Pereira B, Morel L, Lobaccaro JM, Baron S. Individual Comparison of Cholesterol Metabolism in Normal and Tumour Areas in Radical Prostatectomy Specimens from Patients with Prostate Cancer: Results of the CHOMECAP Study. Eur Urol Oncol 2018; 2:198-206. [PMID: 31017097 DOI: 10.1016/j.euo.2018.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/13/2018] [Accepted: 08/01/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND Deregulation of cholesterol metabolism represents a hallmark of prostate cancer (PCa) and promotes its development. OBJECTIVE To compare cholesterol metabolism on individual paired normal and tumour prostate tissues obtained from patients with PCa. DESIGN, SETTING, AND PARTICIPANTS Between 2008 and 2012, normal and tumour paired tissue samples were collected from radical prostatectomy specimens from a cohort of 69 patients treated for localised PCa. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Tumour and normal tissues were subjected to gene analysis, sterol measurement, and immunohistochemistry. The Wilcoxon paired test and Spearman test were applied for comparison and correlation analyses, respectively. Principal component analysis was also carried out to investigate relationships between quantitative variables. RESULTS AND LIMITATIONS Overall, cholesterol concentrations were not significantly different between tissue pairs. However, tumour samples were significantly associated with downregulated de novo cholesterol synthesis, but exhibited 54.7% overexpression of SCARB1 that could increase high-density lipoprotein uptake in PCa. Tumour tissues showed different trafficking of available cholesterol, with significantly lower ACAT1, and an altered efflux via APOE. Furthermore, cholesterol metabolism in tumour tissues was characterised by higher accumulation of 7α-hydroxycholesterol (OHC), 7βOHC, and 7-ketosterol, and a lower level of 27OHC. CONCLUSIONS Focusing on individually paired prostate tissues, our results highlighted several differences between normal and tumour samples linked to a metabolic shift in cholesterol flux. PCa samples exhibited a specific tissue signature characterised by higher SCARB1 expression, higher accumulation of OHC species, and clear downregulation of de novo cholesterol synthesis. PATIENT SUMMARY Comparing normal and tumour tissues from the same prostates, our study identified a set of alterations in prostate cancer samples in terms of their use of cholesterol. These included higher cholesterol uptake, accumulation of oxidised cholesterol derivatives, and autonomous cellular production of cholesterol. Together, these data provide promising clinical targets to fight prostate cancer.
Collapse
Affiliation(s)
- Olivier Celhay
- Laboratoire Génétique, Reproduction et Développement, Université Clermont Auvergne, Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France; Urologie Bordeaux Saint-Gatien, Clinique Tivoli-Ducos, Bordeaux, France; Service d'Urologie, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Laura Bousset
- Laboratoire Génétique, Reproduction et Développement, Université Clermont Auvergne, Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Laurent Guy
- Service d'Urologie, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Jean-Louis Kemeny
- Service d'Anatomie Pathologique, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Valerio Leoni
- Laboratory of Clinical Chemistry, Hospital of Varese, ASST-Settelaghi, Varese, Italy
| | - Claudio Caccia
- Laboratory of Clinical Chemistry, Hospital of Varese, ASST-Settelaghi, Varese, Italy
| | - Amalia Trousson
- Laboratoire Génétique, Reproduction et Développement, Université Clermont Auvergne, Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Christelle Damon-Soubeyrant
- Laboratoire Génétique, Reproduction et Développement, Université Clermont Auvergne, Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Angélique De Haze
- Laboratoire Génétique, Reproduction et Développement, Université Clermont Auvergne, Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Laura Sabourin
- Urologie Bordeaux Saint-Gatien, Clinique Tivoli-Ducos, Bordeaux, France
| | - Catherine Godfraind
- Service d'Anatomie Pathologique, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Cyrille de Joussineau
- Laboratoire Génétique, Reproduction et Développement, Université Clermont Auvergne, Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Bruno Pereira
- Unité de biostatistiques, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Laurent Morel
- Laboratoire Génétique, Reproduction et Développement, Université Clermont Auvergne, Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Jean Marc Lobaccaro
- Laboratoire Génétique, Reproduction et Développement, Université Clermont Auvergne, Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Silvère Baron
- Laboratoire Génétique, Reproduction et Développement, Université Clermont Auvergne, Clermont-Ferrand, France; Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France.
| |
Collapse
|
38
|
Biomimetic nano-surfactant stabilizes sub-50 nanometer phospholipid particles enabling high paclitaxel payload and deep tumor penetration. Biomaterials 2018; 181:240-251. [PMID: 30096559 DOI: 10.1016/j.biomaterials.2018.07.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/05/2018] [Accepted: 07/23/2018] [Indexed: 11/21/2022]
Abstract
Sub-50 nm nanoparticles feature long circulation and deep tumor penetration. However, at high volume fractions needed for intravenous injection, safe, highly biocompatible phospholipids cannot form such nanoparticles due to the fluidity of phospholipid shells. Here we overcome this challenge using a nano-surfactant, a sterilized 18-amino-acid biomimetic of the amphipathic helical motif abundant in HDL-apolipoproteins. As it induces a nanoscale phase (glass) transition in the phospholipid monolayer, the peptide stabilizes 5-7 nm phospholipid micelles that do not fuse at high concentrations but aggregate into stable micellesomes exhibiting size-dependent penetration into tumors. In mice bearing human Her-2-positive breast cancer xenografts, high-payload paclitaxel encapsulated in 25 nm (diameter) micellesomes kills more cancer cells than paclitaxel in standard clinical formulation, as evidenced by the enhanced apparent diffusion coefficient of water determined by in vivo MR imaging. Importantly, the bio-inertness of this biomimetic nano-surfactant spares the nanoparticles from being absorbed by liver hepatocytes, making them more generally available for drug delivery.
Collapse
|
39
|
Pirro M, Ricciuti B, Rader DJ, Catapano AL, Sahebkar A, Banach M. High density lipoprotein cholesterol and cancer: Marker or causative? Prog Lipid Res 2018; 71:54-69. [DOI: 10.1016/j.plipres.2018.06.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 05/15/2018] [Accepted: 06/02/2018] [Indexed: 12/11/2022]
|
40
|
Experimental challenges regarding the in vitro investigation of the nanoparticle-biocorona in disease states. Toxicol In Vitro 2018; 51:40-49. [PMID: 29738787 DOI: 10.1016/j.tiv.2018.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/11/2018] [Accepted: 05/03/2018] [Indexed: 11/20/2022]
Abstract
Toxicological evaluation of nanoparticles (NPs) requires the utilization of in vitro techniques due to their number and diverse properties. Cell culture systems are often lacking in their ability to perform comparative toxicity assessment due to dosimetry issues and capacity to simulate in vivo environments. Upon encountering a physiological environment, NPs become coated with biomolecules forming a biocorona (BC), influencing function, biodistribution, and toxicity. Disease-induced alterations in the biological milieu can alter BC formation. This study evaluates the role of low-density lipoprotein (LDL) in altering macrophage responses to iron oxide (Fe3O4) NPs. BCs were formed by incubating Fe3O4 NPs in serum-free media, or 10% fetal bovine serum with or without LDL present. Following exposures to a normalized dose (25 μg/mL), macrophage association of Fe3O4 NPs with a LDL-BC was enhanced. TNF-α mRNA expression and protein levels were differentially induced due to BCs. Cell surface expression of SR-B1 was reduced following all Fe3O4 NPs exposures, while only NPs with an LDL-BC enhanced mitochondrial membrane potential. These findings suggest that elevations in LDL may contribute to distinct BC formation thereby influencing NP-cellular interactions and response. Further, our study highlights challenges that may arise during the in vitro evaluation of disease-related variations in the NP-BC.
Collapse
|
41
|
Wang X, Wu C, Yuan B, Wang D, Liu H, Feng H, Sun S. Low scavenger receptor class B type I expression is associated with gastric adenocarcinoma tumor aggressiveness. Oncol Lett 2018. [PMID: 29541232 DOI: 10.3892/ol.2018.7889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Scavenger receptor class B type I (SR-BI), a well-documented high-density lipoprotein receptor, has been implicated in the development and progression of human cancer. However, little is known regarding the expression profile and clinical value of SR-BI in gastric adenocarcinoma. In the present study immunohistochemistry analysis was performed on a well-annotated gastric adenocarcinoma tissue microarray to investigate the association between SR-BI expression and clinicopathological parameters or patient outcome. The results revealed that SR-BI expression was detected in 69% of the 84 gastric adenocarcinomas. Moreover, a significant association was observed between low SR-BI expression and poor histological grade, higher Tumor-Node-Metastasis T stage, higher N stage and diffuse type carcinoma. Low SR-BI expression was also significantly associated with a shorter overall survival time in patients with gastric adenocarcinoma, although it was not an independent prognostic factor. Overall, the results of the present study demonstrated that SR-BI was possibly involved in gastric carcinogenesis and could be used as a biomarker to predict malignancy of gastric adenocarcinoma.
Collapse
Affiliation(s)
- Xingwen Wang
- Cancer Center, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Changshun Wu
- Department of Bone and Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Baoying Yuan
- Cancer Center, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Dan Wang
- Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Huiling Liu
- Cancer Center, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Hong Feng
- Cancer Center, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Shui Sun
- Department of Bone and Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
42
|
Feng H, Wang M, Wu C, Yu J, Wang D, Ma J, Han J. High scavenger receptor class B type I expression is related to tumor aggressiveness and poor prognosis in lung adenocarcinoma: A STROBE compliant article. Medicine (Baltimore) 2018; 97:e0203. [PMID: 29595658 PMCID: PMC5895397 DOI: 10.1097/md.0000000000010203] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Scavenger receptor class B type I (SR-B1) is highly expressed in a variety of cancers, including prostate, breast and ovarian. However, the relationship between SR-B1 and lung adenocarcinoma is unknown. We analyzed the expression of SR-B1 in a well-characterized lung adenocarcinoma tissue microarray by immunohistochemistry, in 90 cancerous and 90 adjacent normal lung tissues. Results showed that the positive expression rate of SR-B1 in cancer tissues (86/90, 96%) was significantly higher than that of adjacent tissues (50/90, 56%) (P < .001). And SR-B1 overexpression in lung adenocarcinoma tissue was significantly higher than that of adjacent normal tissue (P < .001), accounting for 67% of cases. This elevated SR-B1 expression was associated with AJCC stage (P < .001), T stage (P = .012), N stage (P = .002), and lymph node positivity (P < .001). The Kaplan-Meier survival analysis indicated that patients with high SR-B1 expression had a shorter overall survival (P < .001). On the multivariate analysis, SR-B1 was an independent prognostic factor for outcomes after adjustment for other prognostic factors (P = .038). In conclusion, high SR-B1 expression is associated with conventional pathologic parameters that represent tumor aggressiveness and may purport a poor clinical prognosis in lung adenocarcinoma.
Collapse
Affiliation(s)
| | | | - Changshun Wu
- Department of Orthopedics, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong
| | - Jinyu Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing
| | - Dan Wang
- Central Laboratory, Shandong Provincial Hospital affiliated to Shandong University, 544 Jingsi Road, Jinan
| | - Jian Ma
- Department of General Surgery, Yangxin County People's Hospital, Yangxin, Shandong, China
| | | |
Collapse
|
43
|
Lipoproteins for therapeutic delivery: recent advances and future opportunities. Ther Deliv 2018; 9:257-268. [DOI: 10.4155/tde-2017-0122] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The physiological role(s) of mammalian plasma lipoproteins is to transport hydrophobic molecules (primarily cholesterol and triacylglycerols) to their respective destinations. Lipoproteins have also been studied as drug-delivery agents due to their advantageous payload capacity, long residence time in the circulation and biocompatibility. The purpose of this review is to briefly discuss current findings with the focus on each type of formulation's potential for clinical applications. Regarding utilizing lipoprotein type formulation for cancer therapeutics, their potential for tumor-selective delivery is also discussed.
Collapse
|
44
|
Xu GH, Lou N, Shi HC, Xu YC, Ruan HL, Xiao W, Liu L, Li X, Xiao HB, Qiu B, Bao L, Yuan CF, Zhou YL, Hu WJ, Chen K, Yang HM, Zhang XP. Up-regulation of SR-BI promotes progression and serves as a prognostic biomarker in clear cell renal cell carcinoma. BMC Cancer 2018; 18:88. [PMID: 29357836 PMCID: PMC5778766 DOI: 10.1186/s12885-017-3761-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/06/2017] [Indexed: 01/05/2023] Open
Abstract
Background Scavenger receptor class B type I (SR-BI) has been reported to be involved in carcinogenesis of several human cancers. However, it is currently unknown whether SR-BI plays a role in clear cell renal cell carcinoma (ccRCC). Here, we aimed to evaluate a tumor promotive mechanism for SR-BI in ccRCC. Methods The expression of SR-BI was evaluated by real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR), Western blot and immunohistochemistry (IHC) in ccRCC tissues and cell lines. Lipid droplets in ccRCC tissues and normal kidney tissues were examined by Oil Red O (ORO) and hematoxylin-eosin (HE) staining. The correlation between SR-BI mRNA levels and clinicopathological features was analyzed by Pearson’s chi-square test or Fisher’s exact test. Kaplan-Meier analysis and Cox model were used to evaluate the difference in progression-free survival (PFS) associated with expression of SR-BI. Inhibition of SR-BI was conducted by using small interfering RNA (siRNA). In vitro assays were performed to assess the impact of SR-BI knockdown on cell biological behaviors. High density lipoprotein (HDL)-cholesterol content in ccRCC cells and extracellular media was also measured after transfection with siRNA. Results The expression of SR-BI was markedly up-regulated in ccRCC tissues and tumor cell lines. ORO and HE staining revealed huge amounts of lipid droplets accumulation in ccRCC. Clinical analysis showed that over-expression of SR-BI was positively associated with tumor size, grade, distant metastasis and inversely correlated with PFS. Furthermore, SR-BI was proved to be an independent prognostic marker in ccRCC patients. The inhibition of SR-BI attenuated the tumorous behaviors of ccRCC cells, expression of metastasis and AKT pathway related proteins. The content of HDL-cholesterol was reduced in cells while increased in extracellular media after transfection with si-SR-BI. Conclusions Our results demonstrate that SR-BI functions as an oncogene and promotes progression of ccRCC. SR-BI may serve as a potential prognostic biomarker and therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Guang-Hua Xu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Ning Lou
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Hang-Chuan Shi
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Yu-Chen Xu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Hai-Long Ruan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Wen Xiao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Lei Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Xiang Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Hai-Bing Xiao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Bin Qiu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Lin Bao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Chang-Fei Yuan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Ya-Li Zhou
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No.13, Hangkong Road, Wuhan, Hubei, 430030, China
| | - Wen-Jun Hu
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No.13, Hangkong Road, Wuhan, Hubei, 430030, China
| | - Ke Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, Hubei, 430022, China
| | - Hong-Mei Yang
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No.13, Hangkong Road, Wuhan, Hubei, 430030, China.
| | - Xiao-Ping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, Hubei, 430022, China.
| |
Collapse
|
45
|
Chang H, Wei JW, Chen K, Zhang S, Han F, Lu LX, Xiao WW, Gao YH. Apolipoprotein A-I Is a Prognosticator of Nasopharyngeal Carcinoma in the Era of Intensity-modulated Radiotherapy. J Cancer 2018; 9:702-710. [PMID: 29556328 PMCID: PMC5858492 DOI: 10.7150/jca.22836] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/08/2017] [Indexed: 02/07/2023] Open
Abstract
Background: In the era of intensity-modulated radiotherapy (IMRT), distant metastasis remains the major cause of death from nasopharyngeal carcinoma (NPC). This study aimed to evaluate the clinical value of pretreatment serum lipid profiles in predicting clinical outcome of NPC. Methodology / Principal Findings: A total of 1927 consecutive patients who had untreated NPC and completed radical IMRT between Jan. 2010 and Dec. 2011 were retrospectively reviewed. Pretreatment serum lipid indexes including total cholesterol, triglyceride, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, apolipoprotein A-I (apoAI) and apolipoprotein B were analyzed for their association with survivals, together with the clinical features (age, sex, pathological type, anemia, chemotherapy sequence and Epstein-Barr virus deoxyribonucleic acid). Hazard ratio (HR) and 95% confidence interval (CI) were calculated for each independent prognosticator. After univariate and multivariate survival analysis, low apoAI level (< 1.125 mmol/L) appeared to predict poor 5-year overall survival (OS), disease-free survival (DFS) and distant-metastasis-free survival (DMFS).The HRs were 1.549 (95% CI, 1.137-2.109), 1.293 (95% CI, 1.047-1.597) and 1.288 (95% CI, 1.022-1.623), respectively. Subgroup survival analysis showed that the apoAI maintained predicting independence for OS, DFS and DMFS in patients with locally advanced NPC, even in those treated with concurrent chemoradiotherapy. Conclusions / Significance: NPC patient with low serum level of pretreatment apoAI might be at risk of distant metastasis. Treatment aiming to eradicate distant metastasis might improve survival of these patients.
Collapse
Affiliation(s)
- Hui Chang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine; Guangzhou, Guangdong 510060, China
| | - Jia-Wang Wei
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine; Guangzhou, Guangdong 510060, China
| | - Kai Chen
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine; Guangzhou, Guangdong 510060, China
| | - Shu Zhang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine; Guangzhou, Guangdong 510060, China
| | - Fei Han
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine; Guangzhou, Guangdong 510060, China
| | - Li-Xia Lu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine; Guangzhou, Guangdong 510060, China
| | - Wei-Wei Xiao
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine; Guangzhou, Guangdong 510060, China
| | - Yuan-Hong Gao
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine; Guangzhou, Guangdong 510060, China
| |
Collapse
|
46
|
Liu YY, Lin SJ, Chen YY, Liu LN, Bao LB, Tang LQ, Ou JS, Liu ZG, Chen XZ, Xu Y, Ma J, Chan AT, Chen M, Xia YF, Liu WL, Zeng YX, Mai HQ, Zeng MS, Pan JJ, Zhang X. High-density lipoprotein cholesterol as a predictor of poor survival in patients with nasopharyngeal carcinoma. Oncotarget 2018; 7:42978-42987. [PMID: 27304186 PMCID: PMC5190001 DOI: 10.18632/oncotarget.7160] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 01/19/2016] [Indexed: 01/26/2023] Open
Abstract
Purpose We aimed to assess the prognostic value of pretreatment high density lipoprotein cholesterol (HDL-C) levels in patients with nasopharyngeal carcinoma (NPC) and investigate the possible biological effects of these lipoproteins on NPC cells in vitro. Experimental Design We examined the prognostic value of pretreatment HDL-C levels in 2443 patients with non-metastatic NPC from three independent institutions. The Cox proportional hazard model and log-rank test were used to analyze the correlation between HDL-C levels and overall survival (OS). Cell growth, colony formation, and apoptotic assays were used to determine the biological functions of HDL on NPC cells in vitro. All of the statistical tests were two-sided. Results OS was decreased in patients with high pretreatment HDL-C levels compared with those with low HDL-C levels (P < 0.05). Similarly, a decreased OS was noted in advanced stage (stage III-IV), NPC patients with high pretreatment HDL-C levels (P < 0.01). Multivariate analyses indicated that HDL-C was an independent prognostic factor associated with shorter OS in training cohorts. These findings were confirmed in both independent validation cohorts (P < 0.01). In vitro experiments demonstrated that HDL could increase cell proliferation, invasion, and colony formation, which were largely dependent on the expression of its receptor SR-B1. Finally, HDL could enhance chemoresistance by protecting cancer cells from apoptosis. Conclusions Pretreatment HDL-C is a poor prognostic factor for patients with NPC. This effect may be associated with the ability of HDL to enhance proliferation, colony formation, migration, and chemoresistance in NPC cells.
Collapse
Affiliation(s)
- Yan-Yan Liu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shao-Jun Lin
- Department of Radiation Oncology, Fujian Provincial Cancer Hospital Affiliated to Fujian Medical University, Fuzhou, China
| | - Yuan-Yuan Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital/Zhejiang Key Laboratory, Hangzhou, China
| | - Li-Na Liu
- Department of Oncology, The Second Affiliated Hospital of Guangzhou Medical College, Guangzhou, China
| | - Liu-Bin Bao
- Department of Oncology, The Second Affiliated Hospital of Guangzhou Medical College, Guangzhou, China
| | - Lin-Quan Tang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jing-Song Ou
- Division of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhi-Gang Liu
- Department of Radiation Oncology, Hunan Provincial Tumor Hospital, Tumor Hospital Xiangya School of Medicine of Central South University, Changsha, China
| | - Xiao-Zhong Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital/Zhejiang Key Laboratory, Hangzhou, China
| | - Yan Xu
- Department of Radiation Oncology, Zhejiang Cancer Hospital/Zhejiang Key Laboratory, Hangzhou, China
| | - Jun Ma
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Anthony T Chan
- Partner State Key Laboratory of Oncology in South China at Sir YK Pao Centre for Cancer, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ming Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital/Zhejiang Key Laboratory, Hangzhou, China
| | - Yun-Fei Xia
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wan-Li Liu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yi-Xin Zeng
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Hai-Qiang Mai
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Mu-Sheng Zeng
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jian-Ji Pan
- Department of Radiation Oncology, Fujian Provincial Cancer Hospital Affiliated to Fujian Medical University, Fuzhou, China
| | - Xing Zhang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
47
|
Panchoo M, Lacko A. Scavenger receptor class B type 1 regulates neuroblastoma cell proliferation, migration and invasion. Biochem Biophys Res Commun 2018; 495:614-620. [DOI: 10.1016/j.bbrc.2017.10.154] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 10/29/2017] [Indexed: 12/18/2022]
|
48
|
Shen WJ, Azhar S, Kraemer FB. SR-B1: A Unique Multifunctional Receptor for Cholesterol Influx and Efflux. Annu Rev Physiol 2017; 80:95-116. [PMID: 29125794 DOI: 10.1146/annurev-physiol-021317-121550] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The scavenger receptor, class B type 1 (SR-B1), is a multiligand membrane receptor protein that functions as a physiologically relevant high-density lipoprotein (HDL) receptor whose primary role is to mediate selective uptake or influx of HDL-derived cholesteryl esters into cells and tissues. SR-B1 also facilitates the efflux of cholesterol from peripheral tissues, including macrophages, back to liver. As a regulator of plasma membrane cholesterol content, SR-B1 promotes the uptake of lipid soluble vitamins as well as viral entry into host cells. These collective functions of SR-B1 ultimately affect programmed cell death, female fertility, platelet function, vasculature inflammation, and diet-induced atherosclerosis and myocardial infarction. SR-B1 has also been identified as a potential marker for cancer diagnosis and prognosis. Finally, the SR-B1-linked selective HDL-cholesteryl ester uptake pathway is now being evaluated as a gateway for the delivery of therapeutic and diagnostic agents. In this review, we focus on the regulation and functional significance of SR-B1 in mediating cholesterol movement into and out of cells.
Collapse
Affiliation(s)
- Wen-Jun Shen
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, California 94305; .,VA Palo Alto Health Care System, Palo Alto, California 94304
| | - Salman Azhar
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, California 94305; .,VA Palo Alto Health Care System, Palo Alto, California 94304
| | - Fredric B Kraemer
- Division of Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, California 94305; .,VA Palo Alto Health Care System, Palo Alto, California 94304
| |
Collapse
|
49
|
Chu Y, Lao W, Jin G, Dai D, Chen L, Kang H. Evaluation of the relationship between CD36 and MARCO single-nucleotide polymorphisms and susceptibility to carotid atherosclerosis in a Chinese Han population. Gene 2017; 633:66-70. [PMID: 28866086 DOI: 10.1016/j.gene.2017.08.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 08/29/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE This study analyzed the genetic association between two scavenger receptors single nucleotide polymorphisms (CD36 rs1761667, MARCO rs12998782) and carotid atherosclerosis in a Chinese Han population. METHODS Samples of genomic DNA collected from patients (n=215) and healthy control subjects (n=252) were analyzed by the polymerase chain reaction with high-resolution melting analysis. Odds ratios and 95% confidence intervals were used to evaluate the association between the two SNPs and carotid atherosclerosis. RESULTS There was no difference between the SNPs regarding their association with the frequency of carotid atherosclerosis in the case and control groups or in the male case group and control group. Female patients of genotype GA for CD36 rs1761667 and CT for MARCO rs12998782 were at an increased risk for carotid atherosclerosis. The presence of rs1761667 GA and rs12998782 CT may increase the risk for carotid atherosclerosis among postmenopausal females. CONCLUSIONS CD36 and MARCO are associated with the susceptibility of Chinese Han females to carotid atherosclerosis. Menopausal status may affect the association between gene polymorphisms and carotid atherosclerosis in the female Chinese Han population.
Collapse
Affiliation(s)
- Yang Chu
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Wenting Lao
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Guojiang Jin
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Di Dai
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Li Chen
- Department of Blood Transfusion, Shanghai East Hospital, Tongji University, Shanghai 200120, China
| | - Hui Kang
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China.
| |
Collapse
|
50
|
Tyagi A, Semwal M, Sharma A. A database of breast oncogenic specific siRNAs. Sci Rep 2017; 7:8706. [PMID: 28821760 PMCID: PMC5562753 DOI: 10.1038/s41598-017-08948-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 07/20/2017] [Indexed: 01/12/2023] Open
Abstract
Breast cancer is a serious problem causing the death of women across the world. At present, one of the major challenges is to design drugs to target breast cancer specific gene(s). RNA interference (RNAi) is an important technique for targeted gene silencing that may lead to promising novel therapeutic strategies for breast cancer. Therefore, identification of such molecules having high oncogene specificity is the need of the hour. Here, we have developed a database named as Breast Oncogenic Specific siRNAs (BOSS, http://bioinformatics.cimap.res.in/sharma/boss/) on the basis of the current research status on siRNA-mediated repression of oncogenes in different breast cancer cell lines. BOSS is a resource of experimentally validated breast oncogenic siRNAs, collected from research articles and patents published yet. The present database contains information on 865 breast oncogenic siRNA entries. Each entry provides comprehensive information of an siRNA that includes its name, sequence, target gene, type of cells, and inhibition value, etc. Additionally, some useful tools like siRNAMAP and BOSS BLAST were also developed and linked with the database. siRNAMAP can be used for the selection of best siRNA against a target gene while BOSS BLAST tool helps to locate the siRNA sequences in deferent oncogenes.
Collapse
Affiliation(s)
- Atul Tyagi
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O.-CIMAP, Near Kukrail Picnic Spot, Lucknow, 226 015, Uttar Pradesh, India.
| | - Manoj Semwal
- ICT Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O.-CIMAP, Near Kukrail Picnic Spot, Lucknow, 226 015, Uttar Pradesh, India.
| | - Ashok Sharma
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O.-CIMAP, Near Kukrail Picnic Spot, Lucknow, 226 015, Uttar Pradesh, India.
| |
Collapse
|