1
|
Hagen MW, Setiawan NJ, Woodruff KA, Termini CM. Syndecans in hematopoietic cells and their niches. Am J Physiol Cell Physiol 2024; 327:C372-C378. [PMID: 38912739 PMCID: PMC11427021 DOI: 10.1152/ajpcell.00326.2024] [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: 05/15/2024] [Revised: 06/13/2024] [Accepted: 06/13/2024] [Indexed: 06/25/2024]
Abstract
Heparan sulfate proteoglycans are a family of glycoproteins that modulate cell signaling by binding growth factors and changing their bioavailability. Syndecans are a specific family of transmembrane heparan sulfate proteoglycans that regulate cell adhesion, migration, and signaling. In this review, we will summarize emerging evidence for the functions of syndecans in the normal and malignant blood systems and their microenvironments. More specifically, we detail the known functions of syndecans within normal hematopoietic stem cells. Furthermore, we discuss the functions of syndecans in hematological malignancies, including myeloid malignancies, lymphomas, and bleeding disorders. As normal and malignant hematopoietic cells require cues from their microenvironments to function, we also summarize the roles of syndecans in cells of the stromal, endothelial, and osteolineage compartments. Syndecan biology is a rapidly evolving field; a comprehensive understanding of these molecules and their place in the hematopoietic system promises to improve our grasp on disease processes and better predict the efficacies of growth factor-targeting therapies.
Collapse
Affiliation(s)
- Matthew W Hagen
- Translational Science & Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States
| | - Nicollette J Setiawan
- Translational Science & Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States
| | - Kelsey A Woodruff
- Translational Science & Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States
| | - Christina M Termini
- Translational Science & Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States
| |
Collapse
|
2
|
Adamowski M, Sharma Y, Molcan T, Wołodko K, Kelsey G, Galvão AM. Leptin signalling regulates transcriptional differences in granulosa cells from genetically obese mice but not the activation of NLRP3 inflammasome. Sci Rep 2024; 14:8070. [PMID: 38580672 PMCID: PMC10997671 DOI: 10.1038/s41598-024-58181-w] [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: 09/04/2023] [Accepted: 03/26/2024] [Indexed: 04/07/2024] Open
Abstract
Obesity is associated with increased ovarian inflammation and the establishment of leptin resistance. We presently investigated the role of impaired leptin signalling on transcriptional regulation in granulosa cells (GCs) collected from genetically obese mice. Furthermore, we characterised the association between ovarian leptin signalling, the activation of the NOD-like receptor protein 3 (NLRP3) inflammasome and macrophage infiltration in obese mice. After phenotype characterisation, ovaries were collected from distinct group of animals for protein and mRNA expression analysis: (i) mice subjected to a diet-induced obesity (DIO) protocol, where one group was fed a high-fat diet (HFD) and another a standard chow diet (CD) for durations of 4 or 16 weeks; (ii) mice genetically deficient in the long isoform of the leptin receptor (ObRb; db/db); (iii) mice genetically deficient in leptin (ob/ob); and (iv) mice rendered pharmacologically hyperleptinemic (LEPT). Next, GCs from antral follicles isolated from db/db and ob/ob mice were subjected to transcriptome analysis. Transcriptional analysis revealed opposing profiles in genes associated with steroidogenesis and prostaglandin action between the genetic models, despite the similarities in body weight. Furthermore, we observed no changes in the mRNA and protein levels of NLRP3 inflammasome components in the ovaries of db/db mice or in markers of M1 and M2 macrophage infiltration. This contrasted with the downregulation of NLRP3 inflammasome components and M1 markers in ob/ob and 16-wk HFD-fed mice. We concluded that leptin signalling regulates NLRP3 inflammasome activation and the expression of M1 markers in the ovaries of obese mice in an ObRb-dependent and ObRb-independent manner. Furthermore, we found no changes in the expression of leptin signalling and NLRP3 inflammasome genes in GCs from db/db and ob/ob mice, which was associated with no effects on macrophage infiltration genes, despite the dysregulation of genes associated with steroidogenesis in homozygous obese db/db. Our results suggest that: (i) the crosstalk between leptin signalling, NLRP3 inflammasome and macrophage infiltration takes place in ovarian components other than the GC compartment; and (ii) transcriptional changes in GCs from homozygous obese ob/ob mice suggest structural rearrangement and organisation, whereas in db/db mice the impairment in steroidogenesis and secretory activity.
Collapse
Affiliation(s)
- Marek Adamowski
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Yashaswi Sharma
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Tomasz Molcan
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Karolina Wołodko
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Gavin Kelsey
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK
| | - António M Galvão
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland.
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK.
- Department of Comparative Biomedical Sciences, Royal Veterinary College, 4 Royal College Street, London, NW1 0TU, UK.
| |
Collapse
|
3
|
Zhai S, Li X, Lin T. Obese Mouse Fat Cell-derived Extracellular Vesicles Transport miR-99a-5p to Mitigate the Proliferation and Migration of Non-small Cell Lung Cancer Cells. Comb Chem High Throughput Screen 2024; 27:214-226. [PMID: 36927435 DOI: 10.2174/1386207326666230316103604] [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: 08/17/2022] [Revised: 12/29/2022] [Accepted: 01/20/2023] [Indexed: 03/18/2023]
Abstract
OBJECTIVE Fat cells-derived extracellular vesicles (FC-EVs) play a role in regulating the tumor microenvironment in cancers by transporting RNAs. MicroRNAs (miRNAs) are vital regulators of cancer development. This study was conducted to explore the role of FC-EVs in the proliferation and migration of non-small cell lung cancer (NSCLC) cells, providing targets for NSCLC treatment. METHODS The obese mouse model was established via high-fat diet (HFD), followed by separation and characterization of FC-EVs (HFD-EVs). The levels of miR-99a-5p, precursor-miR-99a-5p, and heparan sulfate-glucosamine 3-sulfotransferase 3B1 (HS3ST3B1) were measured by RT-qPCR or Western blot assay. Cell proliferation and migration were evaluated by 3-(4, 5-dimethylthiazol- 2-yl)-2, 5-diphenyltetrazolium bromide and wound healing assays. The expression of Cy3-labeled miR-99a-5p in A549 cells (one NSCLC cell line) was observed via confocal microscopy. The binding of miR-99a-5p to HS3ST3B1 was analyzed by the dual luciferase assay. Rescue experiments were performed to confirm the role of HS3ST3B1 in NSCLC cells. RESULTS miR-99a-5p was upregulated in adipose tissues, FCs, and HFD-EVs. HFD-EVs mitigated the proliferation and migration of NSCLC cells. HFD-EVs transported miR-99a-5p into A549 cells, which upregulated miR-99a-5p expression and inhibited HS3ST3B1 expression in A549 cells. HS3ST3B1 overexpression reversed the inhibition of HFD-EVs on the proliferation and migration of NSCLC cells. CONCLUSION HFD-EVs transported miR-99a-5p into NSCLC cells and inhibited HS3ST3B1, thereby inhibiting proliferation and migration of NSCLC cells.
Collapse
Affiliation(s)
- Shengping Zhai
- Department of General Practice, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Xiaoping Li
- Department of Pulmonary and Critical Care Medicine, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Tiantian Lin
- Respiratory Intensive Care Unit, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| |
Collapse
|
4
|
Five EMT-Related Gene Signatures Predict Acute Myeloid Leukemia Patient Outcome. DISEASE MARKERS 2022; 2022:7826393. [PMID: 36246561 PMCID: PMC9568336 DOI: 10.1155/2022/7826393] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/07/2022] [Accepted: 08/23/2022] [Indexed: 11/18/2022]
Abstract
Background The epithelial mesenchymal transition (EMT) gene has been shown to be significantly associated with the prognosis of solid tumors; however, there is a lack of models for the EMT gene to predict the prognosis of AML patients. Methods First, we downloaded clinical data and raw transcriptome sequencing data from the TCGA database of acute myeloid leukemia (AML) patients. All currently confirmed EMT-related genes were obtained from the dbEMT 2.0 database, and 30% of the TCGA data were randomly selected as the test set. Univariate Cox regression analysis, random forest, and lasso regression were used to optimize the number of genes for model construction, and multivariate Cox regression was used for model construction. Area under the ROC curve was used to assess the efficacy of the model application, and the internal validation set was used to assess the stability of the model. Results A total of 173 AML samples were downloaded, and a total of 1184 EMT-related genes were downloaded. The results of univariate batch Cox regression analysis suggested that 212 genes were associated with patient prognosis, random forest and lasso regression yielded 18 and 8 prognosis-related EMT genes, respectively, and the results of multifactorial COX regression model suggested that 5 genes, CBR1, HS3ST3B1, LIMA1, MIR573, and PTP4A3, were considered as independent risk factors affecting patient prognosis. The model ROC results suggested that the area under the curve was 0.868 and the internal validation results showed that the area under the curve was 0.815. Conclusion During this study, we constructed a signature model of five EMT-related genes to predict overall survival in patients with AML; it will provide a useful tool for clinical decision making.
Collapse
|
5
|
Marques C, Reis CA, Vivès RR, Magalhães A. Heparan Sulfate Biosynthesis and Sulfation Profiles as Modulators of Cancer Signalling and Progression. Front Oncol 2021; 11:778752. [PMID: 34858858 PMCID: PMC8632541 DOI: 10.3389/fonc.2021.778752] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/15/2021] [Indexed: 12/17/2022] Open
Abstract
Heparan Sulfate Proteoglycans (HSPGs) are important cell surface and Extracellular Matrix (ECM) maestros involved in the orchestration of multiple cellular events in physiology and pathology. These glycoconjugates bind to various bioactive proteins via their Heparan Sulfate (HS) chains, but also through the protein backbone, and function as scaffolds for protein-protein interactions, modulating extracellular ligand gradients, cell signalling networks and cell-cell/cell-ECM interactions. The structural features of HS chains, including length and sulfation patterns, are crucial for the biological roles displayed by HSPGs, as these features determine HS chains binding affinities and selectivity. The large HS structural diversity results from a tightly controlled biosynthetic pathway that is differently regulated in different organs, stages of development and pathologies, including cancer. This review addresses the regulatory mechanisms underlying HS biosynthesis, with a particular focus on the catalytic activity of the enzymes responsible for HS glycan sequences and sulfation motifs, namely D-Glucuronyl C5-Epimerase, N- and O-Sulfotransferases. Moreover, we provide insights on the impact of different HS structural epitopes over HSPG-protein interactions and cell signalling, as well as on the effects of deregulated expression of HS modifying enzymes in the development and progression of cancer. Finally, we discuss the clinical potential of HS biosynthetic enzymes as novel targets for therapy, and highlight the importance of developing new HS-based tools for better patients' stratification and cancer treatment.
Collapse
Affiliation(s)
- Catarina Marques
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Porto, Portugal.,Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Celso A Reis
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
| | | | - Ana Magalhães
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
6
|
Li J, Su G, Xu Y, Arnold K, Pagadala V, Wang C, Liu J. Synthesis of 3- O-Sulfated Heparan Sulfate Oligosaccharides Using 3- O-Sulfotransferase Isoform 4. ACS Chem Biol 2021; 16:2026-2035. [PMID: 34351732 DOI: 10.1021/acschembio.1c00474] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Heparan sulfate (HS) 3-O-sulfotransferase isoform 4 (3-OST-4) is a specialized carbohydrate sulfotransferase participating in the biosynthesis of heparan sulfate. Here, we report the expression and purification of the recombinant 3-OST-4 enzyme and use it for the synthesis of a library of 3-O-sulfated hexasaccharides and 3-O-sulfated octasaccharides. The unique structural feature of the library is that each oligosaccharide contains a disaccharide domain with a 2-O-sulfated glucuronic acid (GlcA2S) and 3-O-sulfated glucosamine (GlcNS3S). By rearranging the order of the enzymatic modification steps, we demonstrate the synthesis of oligosaccharides with different saccharide sequences. The structural characterization was completed by electrospray ionization mass spectrometry and NMR. These 3-O-sulfated oligosaccharides show weak to very weak anti-Factor Xa activity, a measurement of anticoagulant activity. We discovered that HSoligo 7 (HS oligosaccharide 7), a 3-O-sulfated octasaccharide, binds to high mobility group box 1 protein (HMGB1) and tau protein, both believed to be involved in the process of inflammation. Access to the recombinant 3-OST-4 expands the capability of the chemoenzymatic method to synthesize novel 3-O-sulfated oligosaccharides. The oligosaccharides will become valuable reagents to probe the biological functions of 3-O-sulfated HS and to develop HS-based therapeutic agents.
Collapse
Affiliation(s)
- Jine Li
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Guowei Su
- Glycan Therapeutics Corporation, 617 Hutton Street, Raleigh, North Carolina 27606, United States
| | - Yongmei Xu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Katelyn Arnold
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Vijayakanth Pagadala
- Glycan Therapeutics Corporation, 617 Hutton Street, Raleigh, North Carolina 27606, United States
| | - Chunyu Wang
- Department of Biological Sciences, Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, New York 12180, United States
| | - Jian Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
7
|
Shi H, Li XY, Chen Y, Zhang X, Wu Y, Wang ZX, Chen PH, Dai HQ, Feng J, Chatterjee S, Li ZJ, Huang XW, Wei HQ, Wang J, Lu GD, Zhou J. Quercetin Induces Apoptosis via Downregulation of Vascular Endothelial Growth Factor/Akt Signaling Pathway in Acute Myeloid Leukemia Cells. Front Pharmacol 2020; 11:534171. [PMID: 33362534 PMCID: PMC7758733 DOI: 10.3389/fphar.2020.534171] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 10/29/2020] [Indexed: 12/20/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive haematological malignancy characterized by highly proliferative accumulation of immature and dysfunctional myeloid cells. Quercetin (Qu), one kind of flavonoid, exhibits anti-cancer property in multiple types of solid tumor, but its effect on acute myeloid leukemia is less studied, and the underlying mechanisms still largely unknown. This study aimed to explore the specific target and potential mechanism of quercetin-induced cell death in AML. First, we found that quercetin induces cell death in the form of apoptosis, which was caspase dependent. Second, we found that quercetin-induced apoptosis depends on the decrease of mitochondria membrane potential (MMP) and Bcl-2 proteins. With quantitative chemical proteomics, we observed the downregulation of VEGFR2 and PI3K/Akt signaling in quercetin-treated cells. Consistently, cell studies also identified that VEGFR2 and PI3K/Akt signaling pathways are involved in the action of quercetin on mitochondria and Bcl-2 proteins. The decrease of MMP and cell death could be rescued when PI3K/Akt signaling is activated, suggesting that VEGFR2 and PI3K/Akt exert as upstream regulators for quercetin effect on apoptosis induction in AML cells. In conclusion, our findings from this study provide convincing evidence that quercetin induces cell death via downregulation of VEGF/Akt signaling pathways and mitochondria-mediated apoptosis in AML cells.
Collapse
Affiliation(s)
- Huan Shi
- Department of Physiology, School of Preclinical Medicine, Guangxi Medical University, Nanning, China.,Department of Physiology, School of Medicine, Hunan University of Medicine, Huaihua, China
| | - Xin-Yu Li
- Department of Physiology, School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Yao Chen
- Department of Physiology, School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Xing Zhang
- Artemisinin Research Center and the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yong Wu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China
| | - Zi-Xuan Wang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China
| | - Pan-Hong Chen
- Department of Physiology, School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Hui-Qi Dai
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China
| | - Ji Feng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China
| | - Sayantan Chatterjee
- Department of Physiology, School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Zhong-Jie Li
- Department of Physiology, School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Xiao-Wei Huang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China
| | - Hong-Qiao Wei
- Department of Physiology, School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Jigang Wang
- Artemisinin Research Center and the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guo-Dong Lu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China.,Key Laboratory of High-incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education of China, Nanning, China.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Jing Zhou
- Department of Physiology, School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| |
Collapse
|
8
|
Heparan Sulfate Proteoglycan Signaling in Tumor Microenvironment. Int J Mol Sci 2020; 21:ijms21186588. [PMID: 32916872 PMCID: PMC7554799 DOI: 10.3390/ijms21186588] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 12/18/2022] Open
Abstract
In the last few decades, heparan sulfate (HS) proteoglycans (HSPGs) have been an intriguing subject of study for their complex structural characteristics, their finely regulated biosynthetic machinery, and the wide range of functions they perform in living organisms from development to adulthood. From these studies, key roles of HSPGs in tumor initiation and progression have emerged, so that they are currently being explored as potential biomarkers and therapeutic targets for cancers. The multifaceted nature of HSPG structure/activity translates in their capacity to act either as inhibitors or promoters of tumor growth and invasion depending on the tumor type. Deregulation of HSPGs resulting in malignancy may be due to either their abnormal expression levels or changes in their structure and functions as a result of the altered activity of their biosynthetic or remodeling enzymes. Indeed, in the tumor microenvironment, HSPGs undergo structural alterations, through the shedding of proteoglycan ectodomain from the cell surface or the fragmentation and/or desulfation of HS chains, affecting HSPG function with significant impact on the molecular interactions between cancer cells and their microenvironment, and tumor cell behavior. Here, we overview the structural and functional features of HSPGs and their signaling in the tumor environment which contributes to tumorigenesis and cancer progression.
Collapse
|
9
|
LncRNA KCNQ1OT1 facilitates the progression of bladder cancer by targeting MiR-218-5p/HS3ST3B1. Cancer Gene Ther 2020; 28:212-220. [PMID: 32820233 DOI: 10.1038/s41417-020-00211-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/23/2020] [Accepted: 08/05/2020] [Indexed: 11/09/2022]
Abstract
Long non-coding RNA (lncRNA) is characterized by biological function in diverse cancers. LncRNA KCNQ1 opposite strand/antisense transcript 1 (KCNQ1OT1) is well acknowledged to regulate various cancers, while its role in bladder cancer remains unclear. In the present study, we aimed at probing into the impact and detailed mechanisms of KCNQ1OT1 in bladder cancer progression. In this study, we demonstrated that KCNQ1OT1 expression in bladder cancer tissues was notably up-regulated compared with in normal adjacent tissues, and KCNQ1OT1 modulated the malignant phenotypes of bladder cancer cells. Moreover, it was validated that KCNQ1OT1 could specifically bind to miR-218-5p and reduce its expression. Overexpressed miR-218-5p would inhibit the proliferation and metastasis of bladder cancer cells while facilitating apoptosis. In terms of Mechanism, Heparan Sulfate-Glucosamine 3-Sulfotransferase 3B1 (HS3ST3B1) was validated as a target gene of miR-218-5p, and could be regulated by KCNQ1OT1 indirectly. In conclusion, KCNQ1OT1 can promote the progression of bladder cancer through regulation of miR-218-5p/HS3ST3B1, which is expected to serve as a new therapeutic target for bladder cancer.
Collapse
|
10
|
Gulberti S, Mao X, Bui C, Fournel-Gigleux S. The role of heparan sulfate maturation in cancer: A focus on the 3O-sulfation and the enigmatic 3O-sulfotransferases (HS3STs). Semin Cancer Biol 2020; 62:68-85. [DOI: 10.1016/j.semcancer.2019.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 01/05/2023]
|
11
|
Xiang X, Li L, Bo P, Kuang T, Liu S, Xie X, Guo S, Fu X, Zhang Y. 7‑Difluoromethyl‑5,4'‑dimethoxygenistein exerts anti‑angiogenic effects on acute promyelocytic leukemia HL‑60 cells by inhibiting the TLR4/NF‑κB signaling pathway. Mol Med Rep 2020; 21:2251-2259. [PMID: 32186776 PMCID: PMC7115195 DOI: 10.3892/mmr.2020.11029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 02/18/2020] [Indexed: 01/17/2023] Open
Abstract
Angiogenesis plays an important role in the development and metastasis of tumors, and anti-angiogenesis agents are used to treat tumors. For example, the acute promyelocytic leukemia (APL) may be treated with arsenic trioxide. Angiogenesis in APL is a multi-step dynamic equilibrium process coordinated by various angiogenic stimulators and inhibitors, which play key roles in the occurrence, progression and chemosensitivity of this disease. Our research group previously synthesized 7-difluoromethyl-5,4′-dimethoxygenistein (DFMG), and found that it inhibits angiogenesis during atherosclerotic plaque formation. In the present study, the effect and mechanism of DFMG in angiogenesis induced by APL HL-60 cells was investigated using a chick embryo chorioallantoic membrane model and Matrigel tubule formation assays. The results obtained revealed an anti-angiogenesis effect of DFMG towards HL-60 cells. When the Toll-like receptor 4/nuclear factor-κB (TLR4/NF-κB) signaling pathway was inhibited, the anti-angiogenic effect of DFMG was further enhanced. However, when the TLR4/NF-κB signaling pathway was activated, the anti-angiogenic effect of DFMG was attenuated. These results demonstrated that DFMG inhibits angiogenesis induced by APL HL-60 cells, and provides insights into the mechanism by which DFMG inhibits the TLR4/NF-κB signaling pathway. In conclusion, in the present study, the anti-angiogenesis effect of DFMG on APL has been reported, and the mechanism by which DFMG induced the anti-angiogenesis effect was explored. These findings have provided a potential new drug candidate for the treatment of patients with APL.
Collapse
Affiliation(s)
- Xueping Xiang
- Department of Internal Medicine, Medical College of Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | - Lesai Li
- Department of Gynecologic Oncology, Hunan Cancer Hospital, Changsha, Hunan 410013, P.R. China
| | - Pingjuan Bo
- Department of Internal Medicine, Medical College of Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | - Ting Kuang
- Department of Gynecologic Oncology, Hunan Cancer Hospital, Changsha, Hunan 410013, P.R. China
| | - Sujuan Liu
- Department of Internal Medicine, Medical College of Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | - Xiaolin Xie
- Department of Internal Medicine, Medical College of Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | - Sihui Guo
- Department of Internal Medicine, Medical College of Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | - Xiaohua Fu
- Department of Internal Medicine, Medical College of Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | - Yong Zhang
- Department of Internal Medicine, Medical College of Hunan Normal University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
12
|
Lesur D, Duhirwe G, Kovensky J. High resolution MALDI-TOF-MS and MS/MS: Application for the structural characterization of sulfated oligosaccharides. EUROPEAN JOURNAL OF MASS SPECTROMETRY (CHICHESTER, ENGLAND) 2019; 25:428-436. [PMID: 31109177 DOI: 10.1177/1469066719851438] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Sulfated oligosaccharides are involved in important biological events that are often modulated by specific sequences and sulfation patterns, but their structural analysis remains challenging. Matrix-assisted laser desorption/ionization-mass spectrometry (MALDI-MS) analysis of three different sulfated oligosaccharides (Fondaparinux, the octasulfated pentasaccharide, a disulfated heparin-derived tetrasaccharide 1, and an octasulfated maltoheptaose) 2 was performed using the 2-(4-hydroxyphenylazo)benzoic acid-tetramethylguanidinium (HABA-TMG2) matrix. High resolution mass spectrometry of the main ions observed was successful, and this was complemented by tandem mass spectrometry (MS/MS) analysis for structural assessment. Despite sulfate losses, fully sulfated molecular ions were observed and these allowed the determination of oligosaccharide structures: UA-GlcNAc-UA(2S)-AnhMan(6S) for compound 1 and (Glc6S)6-Glc (1S,6S) for compound 2.
Collapse
Affiliation(s)
- David Lesur
- Laboratoire de Glycochimie, des Antimicrobiens et des Agroressources (LG2A) CNRS UMR 7378, Institut de Chimie de Picardie FR 3085, Université de Picardie Jules Verne, Amiens, France
| | - Gilbert Duhirwe
- Laboratoire de Glycochimie, des Antimicrobiens et des Agroressources (LG2A) CNRS UMR 7378, Institut de Chimie de Picardie FR 3085, Université de Picardie Jules Verne, Amiens, France
| | - José Kovensky
- Laboratoire de Glycochimie, des Antimicrobiens et des Agroressources (LG2A) CNRS UMR 7378, Institut de Chimie de Picardie FR 3085, Université de Picardie Jules Verne, Amiens, France
| |
Collapse
|
13
|
Denys A, Allain F. The Emerging Roles of Heparan Sulfate 3- O-Sulfotransferases in Cancer. Front Oncol 2019; 9:507. [PMID: 31249810 PMCID: PMC6582251 DOI: 10.3389/fonc.2019.00507] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 05/28/2019] [Indexed: 12/14/2022] Open
Abstract
Alteration in the expression of heparan sulfate (HS)-modifying enzymes has been frequently observed in cancer. Consequently, dysregulation of the HS biosynthetic machinery results in dramatic changes in the HS structure, thereby impacting a range of pivotal cellular processes involved in tumorigenesis and cancer progression including proliferation, migration, apoptosis, and immune escape. HS 3-O-sulfotransferases (HS3STs) catalyse the maturation step of glucosaminyl 3-O-sulfation within HS chains. Although seven HS3ST isozymes have been described in human, 3-O-sulfation is a rare modification and only a few biological processes have been described to be influenced by 3-O-sulfated HS. An aberrant expression of HS3STs has been reported in a variety of cancers. Thus, it was suggested that changes in the expression of these enzymes as a result of tumorigenesis or tumor growth may critically influence cancer cell behavior. In accordance with this assumption, a number of studies have documented the epigenetic repression of HS3ST2 and HS3ST3A in many cancers. However, the situation is not so clear, and there is accumulating evidence that HS3ST2, HS3ST3A, HS3ST3B, and HS3ST4 may also act as tumor-promoting enzymes in a number of cancer cells depending on their phenotypes and molecular signatures. In this mini-review, we focus on the recent insights regarding the abnormal expression of HS3STs in cancer and discuss the functional consequences on tumor cell behavior. In term of clinical outcome, further investigations are needed to explore the potential value of HS3STs and/or their 3-O-sulfated products as targets for therapeutic strategies in cancer treatment.
Collapse
Affiliation(s)
- Agnès Denys
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Fabrice Allain
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| |
Collapse
|
14
|
Li Y, Shi B, Dong F, Zhu X, Liu B, Liu Y. Long Non-coding RNA DLEU1 Promotes Cell Proliferation, Invasion, and Confers Cisplatin Resistance in Bladder Cancer by Regulating the miR-99b/HS3ST3B1 Axis. Front Genet 2019; 10:280. [PMID: 30984249 PMCID: PMC6449426 DOI: 10.3389/fgene.2019.00280] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 03/14/2019] [Indexed: 12/12/2022] Open
Abstract
Although accumulating evidence has shown the important function of long non-coding RNAs (lncRNAs) in tumor progression and chemotherapy resistance, the role of lncRNA DLEU1 in regulating proliferation, invasion, and chemoresistance of bladder cancer (BCA) cells remains largely unknown. Here, we found that DLEU1 was upregulated in BLCA tissues and BCA patients with high DLEU1 expression exhibited a shorter survival time. Furthermore, mechanistic analysis and functional assays validated that DLEU1 induced cell proliferation, invasion, and cisplatin resistance of BCA cells by de-repressing the expression of HS3ST3B1 through sponging miR-99b. Low miR-99b and high HS3ST3B1 levels were correlated with worse prognosis in patients with BCA. Ectopic expression of HS3ST3B1 or inhibition of miR-99b reversed DLEU1 knockdown-mediated suppression of cell proliferation, invasion, and cisplatin resistance. Thus, our study revealed a novel role for the DLEU1/miR-99b/HS3ST3B1 axis in regulating proliferation, invasion, and cisplatin resistance of BCA cells.
Collapse
Affiliation(s)
- Yongzhi Li
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Benkang Shi
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Fengming Dong
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Xingwang Zhu
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Bing Liu
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yili Liu
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
15
|
Xie M, Li JP. Heparan sulfate proteoglycan - A common receptor for diverse cytokines. Cell Signal 2018; 54:115-121. [PMID: 30500378 DOI: 10.1016/j.cellsig.2018.11.022] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 01/04/2023]
Abstract
Heparan sulfate proteoglycans (HSPG) are macromolecular glyco-conjugates expressed ubiquitously on the cell surface and in the extracellular matrix where they interact with a wide range of ligands to regulate many aspects of cellular function. The capacity of the side glycosaminoglycan chain heparan sulfate (HS) being able to interact with diverse protein ligands relies on its complex structure that is generated by a controlled biosynthesis process, involving the actions of glycosyl-transferases, sulfotransferases and the glucuronyl C5-epimerase. It is believed that activities of the modification enzymes control the HS structures that are designed to serve the biological functions in a given cell or biological status. In this review, we briefly discuss recent understandings on the roles of HSPG in cytokine stimulated cellular signaling, focusing on FGF, TGF-β, Wnt, Hh, HGF and VEGF.
Collapse
Affiliation(s)
- Meng Xie
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology, SciLifeLab Uppsala, The Biomedical Center, University of Uppsala, Uppsala, Sweden.
| |
Collapse
|
16
|
Kaltenbach DD, Jaishankar D, Hao M, Beer JC, Volin MV, Desai UR, Tiwari V. Sulfotransferase and Heparanase: Remodeling Engines in Promoting Virus Infection and Disease Development. Front Pharmacol 2018; 9:1315. [PMID: 30555321 PMCID: PMC6282075 DOI: 10.3389/fphar.2018.01315] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 10/29/2018] [Indexed: 01/08/2023] Open
Abstract
An extraordinary binding site generated in heparan sulfate (HS) structures, during its biosynthesis, provides a unique opportunity to interact with multiple protein ligands including viral proteins, and therefore adds tremendous value to this master molecule. An example of such a moiety is the sulfation at the C3 position of glucosamine residues in HS chain via 3-O sulfotransferase (3-OST) enzymes, which generates a unique virus-cell fusion receptor during herpes simplex virus (HSV) entry and spread. Emerging evidence now suggests that the unique patterns in HS sulfation assist multiple viruses in invading host cells at various steps of their life cycles. In addition, sulfated-HS structures are known to assist in invading host defense mechanisms and initiating multiple inflammatory processes; a critical event in the disease development. All these processes are detrimental for the host and therefore raise the question of how HS-sulfation is regulated. Epigenetic modulations have been shown to be implicated in these reactions during HSV infection as well as in HS modifying enzyme sulfotransferases, and therefore pose a critical component in answering it. Interestingly, heparanase (HPSE) activity is shown to be upregulated during virus infection and multiple other diseases assisting in virus replication to promote cell and tissue damage. These phenomena suggest that sulfotransferases and HPSE serve as key players in extracellular matrix remodeling and possibly generating unique signatures in a given disease. Therefore, identifying the epigenetic regulation of OST genes, and HPSE resulting in altered yet specific sulfation patterns in HS chain during virus infection, will be a significant a step toward developing potential diagnostic markers and designing novel therapies.
Collapse
Affiliation(s)
- Dominik D Kaltenbach
- Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, Downers Grove, IL, United States
| | - Dinesh Jaishankar
- Department of Ophthalmology & Visual Sciences, University of Illinois at Chicago, Chicago, IL, United States
| | - Meng Hao
- Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, United States
| | - Jacob C Beer
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL, United States
| | - Michael V Volin
- Department of Microbiology & Immunology, College of Graduate Studies, Midwestern University, Downers Grove, IL, United States
| | - Umesh R Desai
- Department of Medicinal Chemistry and Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA, United States
| | - Vaibhav Tiwari
- Department of Microbiology & Immunology, College of Graduate Studies, Midwestern University, Downers Grove, IL, United States
| |
Collapse
|
17
|
The Pro-Tumoral Activity of Heparan Sulfate 3- O-Sulfotransferase 3B (HS3ST3B) in Breast Cancer MDA-MB-231 Cells Is Dependent on the Expression of Neuropilin-1. Molecules 2018; 23:molecules23102718. [PMID: 30360368 PMCID: PMC6222811 DOI: 10.3390/molecules23102718] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/17/2018] [Accepted: 10/19/2018] [Indexed: 01/13/2023] Open
Abstract
Heparan sulfate 3-O-sulfotransferases (HS3STs) catalyze the maturation step of heparan sulfate (HS) 3-O-sulfation. This modification is relatively rare. Moreover, only a few biological processes have been described to be influenced by 3-O-sulfated HS, and few ligands have been identified so far. Among them, neuropilin-1 (Nrp1) was reported to exhibit tumor-promoting properties by enhancing the action of various growth factors. We recently demonstrated that transient overexpression of HS3ST2, 3B or 4 enhanced the proliferation of breast cancer MDA-MB-231 cells and promote efficient protection against pro-apoptotic stimuli. Hence, we hypothesized that the pro-tumoral activity of these HS3STs could depend on the expression of Nrp1. To test this, MDA-MB-231 cells were stably transfected with a construct encoding HS3ST3B and the expression of Nrp1 was down-regulated by RNA interference. First, we confirmed that stable expression of HS3ST3B effectively increased cell proliferation and viability. Silencing the expression of Nrp1 markedly attenuated the promoting effects of HS3ST3B, while the same treatment had only a moderate effect on the behavior of the parental cells. Altogether, our findings support the idea that the tumor-promoting effects of HS3ST3B could be dependent on the expression of Nrp1 in cancer cells.
Collapse
|
18
|
Hellec C, Delos M, Carpentier M, Denys A, Allain F. The heparan sulfate 3-O-sulfotransferases (HS3ST) 2, 3B and 4 enhance proliferation and survival in breast cancer MDA-MB-231 cells. PLoS One 2018; 13:e0194676. [PMID: 29547633 PMCID: PMC5856405 DOI: 10.1371/journal.pone.0194676] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 03/07/2018] [Indexed: 01/03/2023] Open
Abstract
Heparan sulfate 3-O-sulfotransferases (HS3STs) catalyze the final maturation step of heparan sulfates. Although seven HS3ST isozymes have been described in human, 3-O-sulfation is a relatively rare modification, and only a few biological processes have been described to be influenced by 3-O-sulfated motifs. A conflicting literature has recently reported that HS3ST2, 3A, 3B and 4 may exhibit either tumor-promoting or anti-oncogenic properties, depending on the model used and cancer cell phenotype. Hence, we decided to compare the consequences of the overexpression of each of these HS3STs in the same cellular model. We demonstrated that, unlike HS3ST3A, the other three isozymes enhanced the proliferation of breast cancer MDA-MB-231 and BT-20 cells. Moreover, the colony forming capacity of MDA-MB-231 cells was markedly increased by the expression of HS3ST2, 3B and 4. No notable difference was observed between the three isozymes, meaning that the modifications catalyzed by each HS3ST had the same functional impact on cell behavior. We then demonstrated that overexpression of HS3ST2, 3B and 4 was accompanied by increased activation of c-Src, Akt and NF-κB and up-regulation of the anti-apoptotic proteins survivin and XIAP. In line with these findings, we showed that HS3ST-transfected cells are more resistant to cell death induction by pro-apoptotic stimuli or NK cells. Altogether, our findings demonstrate that HS3ST2, 3B and 4 share the same pro-tumoral activity and support the idea that these HS3STs could compensate each other for loss of their expression depending on the molecular signature of cancer cells and/or changes in the tumor environment.
Collapse
Affiliation(s)
- Charles Hellec
- University of Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Maxime Delos
- University of Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Mathieu Carpentier
- University of Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Agnès Denys
- University of Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Fabrice Allain
- University of Lille, CNRS, UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
- * E-mail:
| |
Collapse
|
19
|
Nagarajan A, Malvi P, Wajapeyee N. Heparan Sulfate and Heparan Sulfate Proteoglycans in Cancer Initiation and Progression. Front Endocrinol (Lausanne) 2018; 9:483. [PMID: 30197623 PMCID: PMC6118229 DOI: 10.3389/fendo.2018.00483] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 08/03/2018] [Indexed: 12/28/2022] Open
Abstract
Heparan sulfate (HS) are complex unbranched carbohydrate chains that are heavily modified by sulfate and exist either conjugated to proteins or as free, unconjugated chains. Proteins with covalently bound Heparan sulfate chains are termed Heparan Sulfate Proteoglycans (HSPGs). Both HS and HSPGs bind to various growth factors and act as co-receptors for different cell surface receptors. They also modulate the dynamics and kinetics of various ligand-receptor interactions, which in turn can influence the duration and potency of the signaling. HS and HSPGs have also been shown to exert a structural role as a component of the extracellular matrix, thereby altering processes such as cell adhesion, immune cell infiltration and angiogenesis. Previous studies have shown that HS are deregulated in a variety of solid tumors and hematological malignancies and regulate key aspects of cancer initiation and progression. HS deregulation in cancer can occur as a result of changes in the level of HSPGs or due to changes in the levels of HS biosynthesis and remodeling enzymes. Here, we describe the major cell-autonomous (proliferation, apoptosis/senescence and differentiation) and cell-non-autonomous (angiogenesis, immune evasion, and matrix remodeling) roles of HS and HSPGs in cancer. Finally, we discuss therapeutic opportunities for targeting deregulated HS biosynthesis and HSPGs as a strategy for cancer treatment.
Collapse
Affiliation(s)
- Arvindhan Nagarajan
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Parmanand Malvi
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Narendra Wajapeyee
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, United States
- *Correspondence: Narendra Wajapeyee
| |
Collapse
|
20
|
Antia IU, Mathew K, Yagnik DR, Hills FA, Shah AJ. Analysis of procainamide-derivatised heparan sulphate disaccharides in biological samples using hydrophilic interaction liquid chromatography mass spectrometry. Anal Bioanal Chem 2017; 410:131-143. [DOI: 10.1007/s00216-017-0703-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/08/2017] [Accepted: 10/11/2017] [Indexed: 12/31/2022]
|
21
|
Lymphangiogenesis in rat asthma model. Angiogenesis 2016; 20:73-84. [PMID: 27787629 DOI: 10.1007/s10456-016-9529-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 10/20/2016] [Indexed: 01/04/2023]
Abstract
Although bronchial angiogenesis has been well documented in allergic asthma, lymphangiogenesis has not been widely studied. Therefore, we evaluated changes in lung lymphatics in a rat model of allergen-induced asthma using house dust mite (Der p 1; 100 μg/challenge). Additionally, properties of isolated lung lymphatic endothelial cells (CD45-, CD141+, LYVE-1+, Prox-1+) were studied in vitro. Three weeks after the onset of intranasal allergen exposure (twice-weekly), an increase in the number of lung lymphatic vessels was measured (34% increase) by lung morphometry. New lymphatic structures were seen predominantly in the peribronchial and periarterial interstitial space but also surrounding large airways. Isolated lymphatic endothelial cells from sensitized lungs showed enhanced proliferation (% Ki67+), chemotaxis, and tube formation (number and length) compared to lymphatic endothelial cells isolated from naive rat lungs. This hyper-proliferative lymphangiogenic phenotype was preserved through multiple cell passages (2-8). Lymphatic endothelial cells isolated from naive and HDM-sensitized rats produced similar in vitro levels of VEGF-C, VEGF-D, and VEGFR3 protein, each recognized as critical lymphangiogenic factors. Inhibition with anti-VEGFR (axitinib, 0.1 μM) blocked proliferation and chemotaxis. Results suggest that in vivo sensitization causes fundamental changes to lymphatic endothelium, which are retained in vitro, and may relate to VEGFR downstream signaling.
Collapse
|
22
|
Han Y, Wang X, Wang B, Jiang G. The progress of angiogenic factors in the development of leukemias. Intractable Rare Dis Res 2016; 5:6-16. [PMID: 26989643 PMCID: PMC4761589 DOI: 10.5582/irdr.2015.01048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Angiogenic factors have been demonstrated to play important roles in modulating angiogenesis of solid tumors. Recently, accumulating studies extensively indicated that some angiogenic factors widely exist in malignant cells of hematologic malignancy, which regulated the expression of a number of genes that were involved in abnormal proliferation, differentiation and apoptosis of these cells. With deep research of angiogenic factors, its expression, function and regulatory mechanism were gradually elucidated, and some of them were related to the development and prognosis of leukemia, or provide more possible strategies for treatment of patients with leukemia. Herein, we summarize the progress in study of some important angiogenic factors and hematological malignancies.
Collapse
Affiliation(s)
- Yang Han
- Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
- School of Medicine and Life Sciences, Ji'nan University, Ji'nan, Shandong, China
| | - Xidi Wang
- Laboratory Department, People's Hospital of Zhangqiu City, Zhangqiu, Shandong, China
| | - Bingping Wang
- Department of Hematology, Shengli Oilfield Central Hospital, Dongying, China
| | - Guosheng Jiang
- Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
- Address correspondence to: Dr. Guosheng Jiang, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, NO.18877 of Jingshi Road, Ji'nan, Shandong, China. E-mail:
| |
Collapse
|
23
|
A role for 3-O-sulfated heparan sulfate in promoting human cytomegalovirus infection in human iris cells. J Virol 2015; 89:5185-92. [PMID: 25717110 DOI: 10.1128/jvi.00109-15] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 02/18/2015] [Indexed: 01/17/2023] Open
Abstract
Human cytomegalovirus (HCMV) has emerged as a clinically opportunistic pathogen that targets multiple types of ocular cells and tissues, including the iris region of the uveal tract during anterior uveitis. In this report, we used primary cultures of human iris stroma (HIS) cells derived from human eye donors to investigate HCMV entry. The following lines of evidence suggested the role of 3-O-sulfated heparan sulfate (3-OS HS) during HCMV-mediated entry and cell-to-cell fusion in HIS cells. First, 3-O-sulfotransferase-3 (3-OST-3) expression in HIS cells promoted HCMV internalization, while pretreatment of HIS cells with heparinase enzyme or with anti-3-OS HS (G2) peptide significantly reduced the HCMV-mediated formation of plaques/foci. Second, coculture of the HCMV-infected HIS cells with CHO-K1 cells expressing 3-OS HS significantly enhanced cell fusion. Finally, a similar trend of enhanced fusion was observed with cells expressing HCMV glycoproteins (gB, gO, and gH-gL) cocultured with 3-OS HS cells. Taken together, these results highlight the role of 3-OS HS during HCMV plaque formation and cell-to-cell fusion and identify a novel target for future therapeutic interventions.
Collapse
|