1
|
Wu C, Zhai Y, Ji J, Yang X, Ye L, Lu G, Shi X, Zhai G. Advances in tumor stroma-based targeted delivery. Int J Pharm 2024; 664:124580. [PMID: 39142464 DOI: 10.1016/j.ijpharm.2024.124580] [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: 05/13/2024] [Revised: 08/06/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
The tumor stroma plays a crucial role in tumor progression, and the interactions between the extracellular matrix, tumor cells, and stromal cells collectively influence tumor progression and the efficacy of therapeutic agents. Currently, utilizing components of the tumor stroma for drug delivery is a noteworthy strategy. A number of targeted drug delivery systems designed based on tumor stromal components are entering clinical trials. Therefore, this paper provides a thorough examination of the function of tumor stroma in the advancement of targeted drug delivery systems. One approach is to use tumor stromal components for targeted drug delivery, which includes certain stromal components possessing inherent targeting capabilities like HA, laminin, along with targeting stromal cells homologously. Another method entails directly focusing on tumor stromal components to reshape the tumor stroma and facilitate drug delivery. These drug delivery systems exhibit great potential in more effective cancer therapy strategies, such as precise targeting, enhanced penetration, improved safety profile, and biocompatibility. Ultimately, the deployment of these drug delivery systems can deepen our comprehension of tumor stroma and the advanced development of corresponding drug delivery systems.
Collapse
Affiliation(s)
- Chunyan Wu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84124, United States
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Guoliang Lu
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Xiaoqun Shi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China.
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China.
| |
Collapse
|
2
|
Song Y, Zhou D, Zhang P, Zhu N, Guo R, Wang T, Zhuang F, Sun D. Heparanase accelerates the angiogenesis and inhibits the ferroptosis of p53-mutant non-small cell cancers in VEGF-dependent manner. Cytotechnology 2024; 76:503-517. [PMID: 39188651 PMCID: PMC11344742 DOI: 10.1007/s10616-024-00632-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/17/2024] [Indexed: 08/28/2024] Open
Abstract
The aim of this study is to explore the effects and specific mechanisms of heparanase on angiogenesis and iron deficiency anemia in TP53 mutant cancer. For this purpose, we conducted in vitro cell experiments and in vivo animal experiments respectively. In this study, we first analyzed the differential expression of heparanase in TP53 wild-type and mutant cells, and analyzed its effects on iron removal and angiogenesis in two types of CALU-1 and NCI-H358 cells. Secondly, we validated whether the mechanism of action of heparanase on TP53 mutant cells for iron removal and angiogenesis is related to VEGF. We applied the iron removal agonist erastin and VEGF inhibitor bevacizumab in both in vitro and in vivo experiments to validate the relationship between heparanase and VEGF in the mechanisms of iron removal and angiogenesis. The experimental results show that heparanase is highly expressed in TP53 mutated cancer cells, and has anti-ferroptosis and pro-angiogenic effects. Our experiment also confirmed that the effect of heparanase on TP53 mutant cancer's iron removal and angiogenesis is related to VEGF. In short, heparanase is highly expressed in p53 mutated lung cancer, and the mechanism of ferroptosis tolerance to TP53 mutated cancer is related to VEGF.
Collapse
Affiliation(s)
- Yaobo Song
- Department of Medical Oncology, Yantaishan Hospital, Yantai, China
| | - Dongmei Zhou
- Department of Medical Oncology, Yantaishan Hospital, Yantai, China
| | - Ping Zhang
- Department of Medical Oncology, Yantaishan Hospital, Yantai, China
| | - Na Zhu
- Department of Medical Oncology, Yantaishan Hospital, Yantai, China
| | - Ruijuan Guo
- Department of Medical Oncology, Yantaishan Hospital, Yantai, China
| | - Tian Wang
- Department of Medical Oncology, Yantaishan Hospital, Yantai, China
| | - Feifei Zhuang
- Department of Medical Oncology, Yantaishan Hospital, Yantai, China
| | - Dengjun Sun
- Department of Medical Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, 20 Yuhuangding East Road, Yantai, 264000 Shandong Province China
| |
Collapse
|
3
|
Chen D, Wang LJ, Li HL, Feng F, Li JC, Liu L. Progress of heparanase in septic cardiomyopathy: A review. Medicine (Baltimore) 2024; 103:e38901. [PMID: 39151539 PMCID: PMC11332786 DOI: 10.1097/md.0000000000038901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 08/19/2024] Open
Abstract
Septic cardiomyopathy (SCM) is a severe complication caused by sepsis, resulting in a high mortality rate. The current understanding of the pathogenic mechanism of SCM primarily involves endocardial injury, microcirculation disturbance, mitochondrial dysfunction and fibrosis. Heparanase (HPA), an endo-β-D-glucuronidase, has been implicated in inflammation, immune response, coagulation promotion, microcirculation disturbance, mitochondrial dysfunction and fibrosis. Therefore, it was hypothesized that HPA may play an important role in the pathogenesis of SCM. The present study provides a summary of various pathophysiological changes and mechanisms behind the involvement of HPA in SCM. It also presents a novel perspective on the pathogenic mechanism, diagnosis and treatment of SCM.
Collapse
Affiliation(s)
- Di Chen
- The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu, P. R. China
| | - Lin-Jun Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu, P. R. China
| | - Hong-Lei Li
- The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu, P. R. China
| | - Fei Feng
- The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu, P. R. China
| | - Jian-Chun Li
- The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu, P. R. China
| | - Liping Liu
- The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu, P. R. China
- Departments of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, P. R. China
| |
Collapse
|
4
|
Yuan F, Zhou H, Liu C, Wang Y, Quan J, Liu J, Li H, von Itzstein M, Yu X. Heparanase interacting BCLAF1 to promote the development and drug resistance of ICC through the PERK/eIF2α pathway. Cancer Gene Ther 2024; 31:904-916. [PMID: 38467765 DOI: 10.1038/s41417-024-00754-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/13/2024]
Abstract
Intrahepatic cholangiocarcinoma (ICC) is a primary epithelial carcinoma known for its aggressive nature, high metastatic potential, frequent recurrence, and poor prognosis. Heparanase (HPSE) is the only known endogenous β-glucuronidase in mammals. In addition to its well-established enzymatic roles, HPSE critically exerts non-catalytic function in tumor biology. This study herein aimed to investigate the non-enzymatic roles of HPSE as well as relevant regulatory mechanisms in ICC. Our results demonstrated that HPSE was highly expressed in ICC and promoted the proliferation of ICC cells, with elevated HPSE levels implicating a poor overall survival of ICC patients. Notably, HPSE interacted with Bcl-2-associated factor 1 (BCLAF1) to upregulate the expression of Bcl-2, which subsequently activated the PERK/eIF2α-mediated endoplasmic reticulum (ER) stress pathway to promote anti-apoptotic effect of ICC. Moreover, our in vivo experiments revealed that concomitant administration of gemcitabine and the Bcl-2 inhibitor navitoclax enhanced the sensitivity of ICC cells with highly expressed HPSE to chemotherapy. In summary, our findings revealed that HPSE promoted the development and drug resistance of ICC via its non-enzymatic function. Bcl-2 may be considered as an effective target with therapeutic potential to overcome ICC chemotherapy resistance induced by HPSE, presenting valuable insights into the development of novel therapeutic strategies against ICC.
Collapse
Affiliation(s)
- Fengyan Yuan
- Department of Basic Medical Sciences, School of Medicine, Hunan Normal University, Changsha, China
- Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
- Research Center of Reproduction and Translational Medicine of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Huiqin Zhou
- Department of Basic Medical Sciences, School of Medicine, Hunan Normal University, Changsha, China
- Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
- Research Center of Reproduction and Translational Medicine of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Chongyang Liu
- Department of Basic Medical Sciences, School of Medicine, Hunan Normal University, Changsha, China
- Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
- Research Center of Reproduction and Translational Medicine of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Yi Wang
- Department of Basic Medical Sciences, School of Medicine, Hunan Normal University, Changsha, China
- Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
- Research Center of Reproduction and Translational Medicine of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Jing Quan
- Department of Basic Medical Sciences, School of Medicine, Hunan Normal University, Changsha, China
- Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
- Research Center of Reproduction and Translational Medicine of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Jie Liu
- Department of Basic Medical Sciences, School of Medicine, Hunan Normal University, Changsha, China
- Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
- Research Center of Reproduction and Translational Medicine of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Hao Li
- Biliary Tract Surgery Laboratory, Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, China.
- Hunan Research Center of Biliary Disease, the First Affiliated Hospital of Hunan Normal University, Changsha, China.
- Key Laboratory of Biliary Disease Prevention and treatment, the First Affiliated Hospital of Hunan Normal University,, Changsha, China.
| | - Mark von Itzstein
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, Australia.
| | - Xing Yu
- Department of Basic Medical Sciences, School of Medicine, Hunan Normal University, Changsha, China.
- Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China.
- Research Center of Reproduction and Translational Medicine of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China.
| |
Collapse
|
5
|
Vlodavsky I, Hilwi M, Kayal Y, Soboh S, Ilan N. Impact of heparanase-2 (Hpa2) on cancer and inflammation: Advances and paradigms. FASEB J 2024; 38:e23670. [PMID: 38747803 DOI: 10.1096/fj.202400286r] [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: 02/05/2024] [Revised: 04/09/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024]
Abstract
HPSE2, the gene-encoding heparanase 2 (Hpa2), is mutated in urofacial syndrome (UFS), a rare autosomal recessive congenital disease attributed to peripheral neuropathy. Hpa2 lacks intrinsic heparan sulfate (HS)-degrading activity, the hallmark of heparanase (Hpa1), yet it exhibits a high affinity toward HS, thereby inhibiting Hpa1 enzymatic activity. Hpa2 regulates selected genes that promote normal differentiation, tissue homeostasis, and endoplasmic reticulum (ER) stress, resulting in antitumor, antiangiogenic, and anti-inflammatory effects. Importantly, stress conditions induce the expression of Hpa2, thus establishing a feedback loop, where Hpa2 enhances ER stress which, in turn, induces Hpa2 expression. In most cases, cancer patients who retain high levels of Hpa2 survive longer than patients bearing Hpa2-low tumors. Experimentally, overexpression of Hpa2 attenuates the growth of tumor xenografts, whereas Hpa2 gene silencing results in aggressive tumors. Studies applying conditional Hpa2 knockout (cHpa2-KO) mice revealed an essential involvement of Hpa2 contributed by the host in protecting against cancer and inflammation. This was best reflected by the distorted morphology of the Hpa2-null pancreas, including massive infiltration of immune cells, acinar to adipocyte trans-differentiation, and acinar to ductal metaplasia. Moreover, orthotopic inoculation of pancreatic ductal adenocarcinoma (PDAC) cells into the pancreas of Hpa2-null vs. wild-type mice yielded tumors that were by far more aggressive. Likewise, intravenous inoculation of cancer cells into cHpa2-KO mice resulted in a dramatically increased lung colonization reflecting the involvement of Hpa2 in restricting the formation of a premetastatic niche. Elucidating Hpa2 structure-activity-relationships is expected to support the development of Hpa2-based therapies against cancer and inflammation.
Collapse
Affiliation(s)
- Israel Vlodavsky
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Maram Hilwi
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Yasmin Kayal
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Soaad Soboh
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Neta Ilan
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| |
Collapse
|
6
|
Riccardi F, Tangredi C, Dal Bo M, Toffoli G. Targeted therapy for multiple myeloma: an overview on CD138-based strategies. Front Oncol 2024; 14:1370854. [PMID: 38655136 PMCID: PMC11035824 DOI: 10.3389/fonc.2024.1370854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/28/2024] [Indexed: 04/26/2024] Open
Abstract
Multiple myeloma (MM) is an incurable hematological disease characterized by the uncontrolled growth of plasma cells primarily in the bone marrow. Although its treatment consists of the administration of combined therapy regimens mainly based on immunomodulators and proteosome inhibitors, MM remains incurable, and most patients suffer from relapsed/refractory disease with poor prognosis and survival. The robust results achieved by immunotherapy targeting MM-associated antigens CD38 and CD319 (also known as SLAMF7) have drawn attention to the development of new immune-based strategies and different innovative compounds in the treatment of MM, including new monoclonal antibodies, antibody-drug conjugates, recombinant proteins, synthetic peptides, and adaptive cellular therapies. In this context, Syndecan1 (CD138 or SDC1), a transmembrane heparan sulfate proteoglycan that is upregulated in malignant plasma cells, has gained increasing attention in the panorama of MM target antigens, since its key role in MM tumorigenesis, progression and aggressiveness has been largely reported. Here, our aim is to provide an overview of the most important aspects of MM disease and to investigate the molecular functions of CD138 in physiologic and malignant cell states. In addition, we will shed light on the CD138-based therapeutic approaches currently being tested in preclinical and/or clinical phases in MM and discuss their properties, mechanisms of action and clinical applications.
Collapse
Affiliation(s)
- Federico Riccardi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Carmela Tangredi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| |
Collapse
|
7
|
Kocabay S, Alagöz MA, Akkaya B. Investigation of inhibitory effect of sulfated chitosan oligomer on human heparanase enzyme: in silico and in vitro studies. J Biomol Struct Dyn 2024:1-9. [PMID: 38410992 DOI: 10.1080/07391102.2024.2317421] [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: 10/16/2023] [Accepted: 02/04/2024] [Indexed: 02/28/2024]
Abstract
Deaths from cancer are widespread worldwide and the numbers continue to increase day by day. During the disease progression of cancer in cells, many of its metabolic activities change. Increased heparanase enzyme release is just one example. Following heparanase enzyme activity, many molecules interact with the remodeling of glycosaminoglycan structures, which triggers the release of different enzymes, cytokines, and growth factors, including fibroblast growth factors (FGF1 and FGF2), vascular endothelial growth factor (VEGF), hepatocyte growth factor, transforming growth factor β and platelet-derived growth factor. These are the most important factors in metastasis due to the formation of new vascular structures caused by those elements. To reduce tumor growth and metastasis, various drugs have been designed by modifying chitosan and its derivatives. In this study, we used chitosan oligomer (A), sulfated chitosan oligomer (ShCsO) (B), heparin (C), phosphate monomer (D1) of PI-88 and sulfate monomer (D2) of PI-88 as heparanase inhibitors. We modified the chitosan oligomer with chlorosulfonic acid to synthesize ShCsO to investigate its inhibitory effects on human serum heparanase. Also examined were molecular docking; molecular dynamics (MD); adsorption, distribution, metabolism, elimination and toxicity (ADMET); and target prediction. ShCsO decreased enzyme activity at a concentration of 0.0001 mg/mL. The docking scores of A, B and C from in silico studies were -6.254, -6.936 and -6.980 kcal/mol, respectively, and the scores for the two different PI-88 monomers were -5.741 and -5.824 kcal/mol. These results show that ShCsO may be a potential drug candidate for treating cancer.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Samet Kocabay
- Department of Molecular Biology and Genetics, Faculty of Science and Literature, Inonu University, Malatya, Türkiye
| | - M Abdullah Alagöz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Inonu University, Malatya, Türkiye
| | - Birnur Akkaya
- Department of Molecular Biology and Genetics, Faculty of Science, Sivas Cumhuriyet University, Sivas, Türkiye
| |
Collapse
|
8
|
Xie C, Schaefer L, Iozzo RV. Global impact of proteoglycan science on human diseases. iScience 2023; 26:108095. [PMID: 37867945 PMCID: PMC10589900 DOI: 10.1016/j.isci.2023.108095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023] Open
Abstract
In this comprehensive review, we will dissect the impact of research on proteoglycans focusing on recent developments involved in their synthesis, degradation, and interactions, while critically assessing their usefulness in various biological processes. The emerging roles of proteoglycans in global infections, specifically the SARS-CoV-2 pandemic, and their rising functions in regenerative medicine and biomaterial science have significantly affected our current view of proteoglycans and related compounds. The roles of proteoglycans in cancer biology and their potential use as a next-generation protein-based adjuvant therapy to combat cancer is also emerging as a constructive and potentially beneficial therapeutic strategy. We will discuss the role of proteoglycans in selected and emerging areas of proteoglycan science, such as neurodegenerative diseases, autophagy, angiogenesis, cancer, infections and their impact on mammalian diseases.
Collapse
Affiliation(s)
- Christopher Xie
- Department of Pathology and Genomic Medicine, the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Liliana Schaefer
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | - Renato V. Iozzo
- Department of Pathology and Genomic Medicine, the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
9
|
McDonald B, Barth K, Schmidt MHH. The origin of brain malignancies at the blood-brain barrier. Cell Mol Life Sci 2023; 80:282. [PMID: 37688612 PMCID: PMC10492883 DOI: 10.1007/s00018-023-04934-1] [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: 05/17/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/11/2023]
Abstract
Despite improvements in extracranial therapy, survival rate for patients suffering from brain metastases remains very poor. This is coupled with the incidence of brain metastases continuing to rise. In this review, we focus on core contributions of the blood-brain barrier to the origin of brain metastases. We first provide an overview of the structure and function of the blood-brain barrier under physiological conditions. Next, we discuss the emerging idea of a pre-metastatic niche, namely that secreted factors and extracellular vesicles from a primary tumor site are able to travel through the circulation and prime the neurovasculature for metastatic invasion. We then consider the neurotropic mechanisms that circulating tumor cells possess or develop that facilitate disruption of the blood-brain barrier and survival in the brain's parenchyma. Finally, we compare and contrast brain metastases at the blood-brain barrier to the primary brain tumor, glioma, examining the process of vessel co-option that favors the survival and outgrowth of brain malignancies.
Collapse
Affiliation(s)
- Brennan McDonald
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Dresden, Germany.
| | - Kathrin Barth
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Dresden, Germany
| | - Mirko H H Schmidt
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Dresden, Germany
| |
Collapse
|
10
|
Cai H, Chen Y, Zhang Q, Liu Y, Jia H. High preoperative CEA and systemic inflammation response index (C-SIRI) predict unfavorable survival of resectable colorectal cancer. World J Surg Oncol 2023; 21:178. [PMID: 37291634 DOI: 10.1186/s12957-023-03056-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/28/2023] [Indexed: 06/10/2023] Open
Abstract
BACKGROUND CEA and systemic inflammation were reported to correlate with proliferation, invasion, and metastasis of colorectal cancer. This study investigated the prognostic significance of the preoperative CEA and systemic inflammation response index (C-SIRI) in patients with resectable colorectal cancer. METHODS Two hundred seventeen CRC patients were recruited from Chongqing Medical University, the first affiliated hospital, between January 2015 and December 2017. Baseline characteristics, preoperative CEA level, and peripheral monocyte, neutrophil, and lymphocyte counts were retrospectively reviewed. The optimal cutoff value for SIRI was defined as 1.1, and for CEA, the best cutoff values were 4.1 ng/l and 13.0 ng/l. Patients with low levels of CEA (< 4.1 ng/l) and SIRI (< 1.1) were assigned a value of 0, those with high levels of CEA (≥ 13.0 ng/l) and SIRI (≥ 1.1) were assigned a value of 3, and those with CEA (4.1-13.0 ng/l) and SIRI (≥ 1.1), CEA (≥ 13.0 ng/l), and SIRI (< 1.1) were assigned a value of 2. Those with CEA (< 4.1 ng/l) and SIRI (≥ 1.1) and CEA (4.1-13.0 ng/l) and SIRI (< 1.1) were assigned a value of 1. The prognostic value was assessed based on univariate and multivariate survival analysis. RESULTS Preoperative C-SIRI was statistically correlated with gender, site, stage, CEA, OPNI, NLR, PLR, and MLR. However, no difference was observed between C-SIRI and age, BMI, family history of cancer, adjuvant therapy, and AGR groups. Among these indicators, the correlation between PLR and NLR is the strongest. In addition, high preoperative C-SIRI was significantly correlated with poorer overall survival (OS) (HR: 2.782, 95% CI: 1.630-4.746, P < 0.001) based on univariate survival analysis. Moreover, it remained an independent predictor for OS (HR: 2.563, 95% CI: 1.419-4.628, p = 0.002) in multivariate Cox regression analysis. CONCLUSION Our study showed that preoperative C-SIRI could serve as a significant prognostic biomarker in patients with resectable colorectal cancer.
Collapse
Affiliation(s)
- Hao Cai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1, Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Yu Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1, Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Qiao Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1, Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Yang Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1, Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - HouJun Jia
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, No.1, Medical College Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
11
|
Liang X, Yang Y, Huang C, Ye Z, Lai W, Luo J, Li X, Yi X, Fan JB, Wang Y, Wang Y. cRGD-targeted heparin nanoparticles for effective dual drug treatment of cisplatin-resistant ovarian cancer. J Control Release 2023; 356:691-701. [PMID: 36933699 DOI: 10.1016/j.jconrel.2023.03.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/23/2023] [Accepted: 03/10/2023] [Indexed: 03/20/2023]
Abstract
Resistance to the chemotherapeutic agent cisplatin (DDP) is the primary reason for invalid chemotherapy of ovarian cancer. Given the complex mechanisms underlying chemo-resistance, the design of combination therapies based on blocking multiple mechanisms is a rationale to synergistically elevate therapeutic effect for effectively overcoming cancer chemo-resistance. Herein, we demonstrated a multifunctional nanoparticle (DDP-Ola@HR), which could simultaneously co-deliver DDP and Olaparib (Ola, DNA damage repair inhibitor) using targeted ligand cRGD peptide modified with heparin (HR) as nanocarrier, enabling the concurrent tackling of multiple resistance mechanisms to effectively inhibit the growth and metastasis of DDP-resistant ovarian cancer. In combination strategy, heparin could suppress the function of multidrug resistance-associated protein 2 (MRP2) and P-glycoprotein (P-gp) to promote the intracellular accumulation of DDP and Ola by specifically binding with heparanase (HPSE) to down-regulate PI3K/AKT/mTOR signaling pathway, and simultaneously served as a carrier combined with Ola to synergistically enhance the anti-proliferation ability of DDP for resistant ovarian cancer, thus achieving great therapeutic efficacy. Our DDP-Ola@HR could provide a simple and multifunctional combination strategy to trigger an anticipated cascading effect, thus effectively overcoming the chemo-resistance of ovarian cancer.
Collapse
Affiliation(s)
- Xiaomei Liang
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Yulu Yang
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Chuanqing Huang
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Zhibin Ye
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Wujiang Lai
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Jiamao Luo
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Xiaoxuan Li
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Xiao Yi
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China
| | - Jun-Bing Fan
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China.
| | - Ying Wang
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China.
| | - Yifeng Wang
- Department of Obstetrics & Gynecology, Zhujiang Hospital; Cancer Research Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, Southern Medical University, Guangzhou, Cuangdong, 510515, China.
| |
Collapse
|
12
|
Wang HQ, Fu R, Man QW, Yang G, Liu B, Bu LL. Advances in CAR-T Cell Therapy in Head and Neck Squamous Cell Carcinoma. J Clin Med 2023; 12:jcm12062173. [PMID: 36983174 PMCID: PMC10052000 DOI: 10.3390/jcm12062173] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/22/2023] [Accepted: 03/02/2023] [Indexed: 03/16/2023] Open
Abstract
Surgery with the assistance of conventional radiotherapy, chemotherapy and immunotherapy is the basis for head and neck squamous cell carcinoma (HNSCC) treatment. However, with these treatment modalities, the recurrence and metastasis of tumors remain at a high level. Increasingly, the evidence indicates an excellent anti-tumor effect of chimeric antigen receptor T (CAR-T) cells in hematological malignancy treatment, and this novel immunotherapy has attracted researchers’ attention in HNSCC treatment. Although several clinical trials have been conducted, the weak anti-tumor effect and the side effects of CAR-T cell therapy against HNSCC are barriers to clinical translation. The limited choices of targeting proteins, the barriers of CAR-T cell infiltration into targeted tumors and short survival time in vivo should be solved. In this review, we introduce barriers of CAR-T cell therapy in HNSCC. The limitations and current promising strategies to overcome barriers in solid tumors, as well as the applications for HNSCC treatment, are covered. The perspectives of CAR-T cell therapy in future HNSCC treatment are also discussed.
Collapse
Affiliation(s)
- Han-Qi Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
- Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Ruxing Fu
- Department of Materials Science and Engineering, University of California, Los Angeles, CA 92093, USA
| | - Qi-Wen Man
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
- Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Guang Yang
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bing Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
- Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
- Correspondence: (B.L.); (L.-L.B.)
| | - Lin-Lin Bu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
- Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
- Correspondence: (B.L.); (L.-L.B.)
| |
Collapse
|
13
|
Gu Y, Peng L, Ding W, Wang Y, Zeng X. An ultrasensitive FRET-based fluorescent low molecular weight heparin nanoprobe for quantifying heparanase activity. Talanta 2023; 254:124207. [PMID: 36549136 DOI: 10.1016/j.talanta.2022.124207] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/13/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022]
Abstract
Heparanase (HPA) is a multifaceted endo-β-glucuronidase, and its dysregulation facilitates cancer metastasis. Developing techniques for fast and sensitively monitoring HPA enzymatic activity is crucial for searching for molecular therapies targeting HPA. Herein, we developed a novel fluorescence resonance energy transfer (FRET)-based nanoprobe AuNCs-LMWH-AuNRs, with AuNCs@GSH-cys and AuNRs/end-NH2/side-SiO2 attached to the non-reducing terminus and reducing terminus of low molecular weight heparin (LMWH), respectively. AuNCs@GSH-cys exhibited an absolute quantum yield of 1.1%. The absorption spectra of AuNRs/end-NH2/side-SiO2 (825 nm for maximum longitudinal absorption) and the emission spectra of AuNCs@GSH-cys (824 nm for maximum emission) were precisely overlapping, further enhancing the efficiency of FRET. In the presence of HPA, the LMWH nanoprobe exhibited an ultrasensitive response with excitation/emission wavelength (lambda (ex) = 560 nm, lambda (em) = 824 nm). The probe presented a wide linear dynamic detection range (LDR) of 0.125 ng/μL - 0.01 μg/μL in vitro with a limit of detection (LODs) of 82.15 pM (0.43 pg/μL). The excellent selectivity and good fluorescence turn-on efficiency of the probe made it possible for one-step detection of cellular heparanase activity. High throughput screening of HPA inhibitors also can be accomplished using the highly efficient LMWH nanoprobe.
Collapse
Affiliation(s)
- Yayun Gu
- Medical School, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province, 226001, China.
| | - Lizhong Peng
- Medical School, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province, 226001, China
| | - Weihua Ding
- Medical School, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province, 226001, China
| | - Yang Wang
- Kobilka Institute of Innovative Drug Discovery, The Chinese University of Hong Kong, 2001 Longxiang Avenue, Shenzhen, Guangdong Province, 518172, China
| | - Xuhui Zeng
- Medical School, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province, 226001, China.
| |
Collapse
|
14
|
Su X, Wang B, Zhou Z, Li Z, Tong S, Chen S, Zhang N, Liu S, Zhang M. A positive feedback loop of heparanase/syndecan1/nerve growth factor regulates cancer pain progression. Korean J Pain 2023; 36:60-71. [PMID: 36536517 PMCID: PMC9812689 DOI: 10.3344/kjp.22277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 12/24/2022] Open
Abstract
Background The purpose of this research was to assess the role of heparanase (HPSE)/syndecan1 (SDC1)/nerve growth factor (NGF) on cancer pain from melanoma. Methods The influence of HPSE on the biological function of melanoma cells and cancer pain in a mouse model was evaluated. Immunohistochemical staining was used to analyze HPSE and SDC1. HPSE, NGF, and SDC1 were detected using western blot. Inflammatory factors were detected using ELISA assay. Results HPSE promoted melanoma cell viability, proliferation, migration, invasion, and tumor growth, as well as cancer pain, while SST0001 treatment reversed the promoting effect of HPSE. HPSE up-regulated NGF, and NGF feedback promoted HPSE. High expression of NGF reversed the inhibitory effect of HPSE down-regulation on melanoma cell phenotype deterioration, including cell viability, proliferation, migration, and invasion. SST0001 down-regulated SDC1 expression. SDC1 reversed the inhibitory effect of SST0001 on cancer pain. Conclusions The results showed that HPSE promoted melanoma development and cancer pain by interacting with NGF/SDC1. It provides new insights to better understand the role of HPSE in melanoma and also provides a new direction for cancer pain treatment.
Collapse
Affiliation(s)
- Xiaohu Su
- Department of Anesthesiology, Suqian First People’s Hospital, Suqian City, Jiangsu Province, China
| | - Bingwu Wang
- Cancer Institute, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Zhaoyun Zhou
- Department of Anesthesiology, Tai’an Central Hospital, Tai’an City, Shandong Province, China
| | - Zixian Li
- Department of Anesthesiology, Graduate School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Song Tong
- Department of Anesthesiology, Graduate School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Simin Chen
- Department of Anesthesiology, Graduate School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Nan Zhang
- Department of Anesthesiology, Graduate School of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Su Liu
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Maoyin Zhang
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China,Correspondence: Maoyin Zhang Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, No. 99, Huaihai West Road, Quanshan District, Xuzhou City, Jiangsu Province 221002, China, Tel: +86-18168777315, Fax: +86-0516-85805911, E-mail:
| |
Collapse
|
15
|
Tripathi K, Bandari SK, Sanderson RD. Extracellular vesicles released during hypoxia transport heparanase and enhance macrophage migration, endothelial tube formation and cancer cell stemness. PROTEOGLYCAN RESEARCH 2023; 1:e1. [PMID: 37091070 PMCID: PMC10117102 DOI: 10.1002/pgr2.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 03/30/2023]
Abstract
Heparanase is upregulated during the progression of most cancers and via its enzyme activity promotes extracellular matrix degradation, angiogenesis and cell migration. Heparanase expression is often associated with enhanced tumor aggressiveness and chemoresistance. We previously demonstrated that increased heparanase expression in tumor cells enhances secretion and alters the composition of tumor-released exosomes. In the present study, we discovered that extracellular vesicles (EVs) secreted by human multiple myeloma cells growing in hypoxic conditions exhibited elevated levels of heparanase cargo compared to EVs from cells growing in normoxic conditions. When macrophages (RAW 264.7 monocyte/macrophage-like cells) were exposed to EVs released by tumor cells growing in either hypoxic or normoxic conditions, macrophage migration and invasion was elevated by EVs from hypoxic conditions. The elevated invasion of macrophages was blocked by a monoclonal antibody that inhibits heparanase enzyme activity. Moreover, the heparanase-bearing EVs from hypoxic cells greatly enhanced endothelial cell tube formation consistent with the known role of heparanase in promoting angiogenesis. EVs from hypoxic tumor cells when compared with EVs from normoxic cells also enhanced cancer stemness properties of both CAG and RPMI 8226 human myeloma cells. Together these data indicate that under hypoxic conditions, tumor cells secrete EVs having an elevated level of heparanase as cargo. These EVs can act on both tumor and non-tumor cells, enhancing tumor progression and tumor cell stemness that likely supports chemoresistance and relapse of tumor.
Collapse
Affiliation(s)
- Kaushlendra Tripathi
- Department of PathologyUniversity of Alabama at BirminghamBirminghamALUSA
- Present address:
Building 29B, Room 5NN Suite 22, Lab 5NN11, Molecular Pathology SectionLab of Immunogenetics, NIAID, NIH9000 Rockville PikeBethesdaMaryland20892USA
| | - Shyam K. Bandari
- Department of PathologyUniversity of Alabama at BirminghamBirminghamALUSA
- Present address:
Exelixis1851 Harbor Bay ParkwayAlamedaCalifornia94502USA
| | - Ralph D. Sanderson
- Department of PathologyUniversity of Alabama at BirminghamBirminghamALUSA
| |
Collapse
|
16
|
Hammond E, Ferro V. An Enzymatic Activity Assay for Heparanase That Is Useful for Evaluating Clinically Relevant Inhibitors and Studying Kinetics. Methods Mol Biol 2023; 2619:227-238. [PMID: 36662473 DOI: 10.1007/978-1-0716-2946-8_16] [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] [Indexed: 01/21/2023]
Abstract
The enzyme heparanase cleaves heparan sulfate and is involved in a range of human diseases including cancer, inflammation, diabetes, and viral infection. There is a need for a simple and reliable enzymatic assay to allow for the screening of compounds to find inhibitors of heparanase. We have developed an assay that uses the heparinoid fondaparinux as enzyme substrate and detects one of the products of catalysis, which contains a newly formed reducing terminus, with the tetrazolium salt WST-1. Due to the homogenous substrate and single point of cleavage therein, this assay allows for more systematic kinetic analysis of heparanase inhibitors. Here, we provide a detailed method for conducting this assay and also provide information to assist researchers in evaluating whether the assay is performing properly in their laboratories.
Collapse
Affiliation(s)
| | - Vito Ferro
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
17
|
Schleyer KA, Liu J, Chen Z, Wang Z, Zhang Y, Zuo J, Ybargollin AJ, Guo H, Cui L. A Universal and Modular Scaffold for Heparanase Activatable Probes and Drugs. Bioconjug Chem 2022; 33:2290-2298. [PMID: 36346913 PMCID: PMC10897860 DOI: 10.1021/acs.bioconjchem.2c00426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Heparanase (HPSE) is an endo-β-glucuronidase involved in extracellular matrix remodeling in rapidly healing tissues, most cancers and inflammation, and viral infection. Its importance as a therapeutic target warrants further study, but such is hampered by a lack of research tools. To expand the toolkits for probing HPSE enzymatic activity, we report the design of a substrate scaffold for HPSE comprised of a disaccharide substrate appended with a linker, capable of carrying cargo until being cleaved by HPSE. Here exemplified as a fluorogenic, coumarin-based imaging probe, this scaffold can potentially expand the availability of HPSE-responsive imaging or drug delivery tools using a variety of imaging moieties or other cargo. We show that electronic tuning of the scaffold provides a robust response to HPSE while simplifying the structural requirements of the attached cargo. Molecular docking and modeling suggest a productive probe/HPSE binding mode. These results further support the hypothesis that the reactivity of these HPSE-responsive probes is predominantly influenced by the electron density of the aglycone. This universal HPSE-activatable scaffold will greatly facilitate future development of HPSE-responsive probes and drugs.
Collapse
Affiliation(s)
- Kelton A Schleyer
- Department of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, UF Health Cancer Center, University of Florida, Gainesville, Florida 32610, United States
| | - Jun Liu
- Department of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, UF Health Cancer Center, University of Florida, Gainesville, Florida 32610, United States
| | - Zixin Chen
- Department of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, UF Health Cancer Center, University of Florida, Gainesville, Florida 32610, United States
| | - Zhishen Wang
- Department of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, UF Health Cancer Center, University of Florida, Gainesville, Florida 32610, United States
| | - Yuzhao Zhang
- Department of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, UF Health Cancer Center, University of Florida, Gainesville, Florida 32610, United States
| | - Junxiang Zuo
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Alberto Jimenez Ybargollin
- Department of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, UF Health Cancer Center, University of Florida, Gainesville, Florida 32610, United States
| | - Hua Guo
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Lina Cui
- Department of Medicinal Chemistry, College of Pharmacy, UF Health Science Center, UF Health Cancer Center, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
18
|
Franchi M, Karamanos KA, Cappadone C, Calonghi N, Greco N, Franchi L, Onisto M, Masola V. Substrate Type and Concentration Differently Affect Colon Cancer Cells Ultrastructural Morphology, EMT Markers, and Matrix Degrading Enzymes. Biomolecules 2022; 12:1786. [PMID: 36551219 PMCID: PMC9775446 DOI: 10.3390/biom12121786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Aim of the study was to understand the behavior of colon cancer LoVo-R cells (doxorubicin-resistant) vs. LoVo-S (doxorubicin sensitive) in the initial steps of extracellular matrix (ECM) invasion. We investigated how the matrix substrates Matrigel and type I collagen-mimicking the basement membrane (BM) and the normal or desmoplastic lamina propria, respectively-could affect the expression of epithelial-to-mesenchymal transition (EMT) markers, matrix-degrading enzymes, and phenotypes. Gene expression with RT-qPCR, E-cadherin protein expression using Western blot, and phenotypes using scanning electron microscopy (SEM) were analyzed. The type and different concentrations of matrix substrates differently affected colon cancer cells. In LoVo-S cells, the higher concentrated collagen, mimicking the desmoplastic lamina propria, strongly induced EMT, as also confirmed by the expression of Snail, metalloproteases (MMPs)-2, -9, -14 and heparanase (HPSE), as well as mesenchymal phenotypes. Stimulation in E-cadherin expression in LoVo-S groups suggests that these cells develop a hybrid EMT phenotype. Differently, LoVo-R cells did not increase their aggressiveness: no changes in EMT markers, matrix effectors, and phenotypes were evident. The low influence of ECM components in LoVo-R cells might be related to their intrinsic aggressiveness related to chemoresistance. These results improve understanding of the critical role of tumor microenvironment in colon cancer cell invasion, driving the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Marco Franchi
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy
| | | | - Concettina Cappadone
- Department of Pharmacy and Biotechnologies, University of Bologna, 40126 Bologna, Italy
| | - Natalia Calonghi
- Department of Pharmacy and Biotechnologies, University of Bologna, 40126 Bologna, Italy
| | - Nicola Greco
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Leonardo Franchi
- Department of Medicine, University of Bologna, 40126 Bologna, Italy
| | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Valentina Masola
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| |
Collapse
|
19
|
Horwacik I. The Extracellular Matrix and Neuroblastoma Cell Communication-A Complex Interplay and Its Therapeutic Implications. Cells 2022; 11:cells11193172. [PMID: 36231134 PMCID: PMC9564247 DOI: 10.3390/cells11193172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 11/16/2022] Open
Abstract
Neuroblastoma (NB) is a pediatric neuroendocrine neoplasm. It arises from the sympatho-adrenal lineage of neural-crest-derived multipotent progenitor cells that fail to differentiate. NB is the most common extracranial tumor in children, and it manifests undisputed heterogeneity. Unsatisfactory outcomes of high-risk (HR) NB patients call for more research to further inter-relate treatment and molecular features of the disease. In this regard, it is well established that in the tumor microenvironment (TME), malignant cells are engaged in complex and dynamic interactions with the extracellular matrix (ECM) and stromal cells. The ECM can be a source of both pro- and anti-tumorigenic factors to regulate tumor cell fate, such as survival, proliferation, and resistance to therapy. Moreover, the ECM composition, organization, and resulting signaling networks are vastly remodeled during tumor progression and metastasis. This review mainly focuses on the molecular mechanisms and effects of interactions of selected ECM components with their receptors on neuroblastoma cells. Additionally, it describes roles of enzymes modifying and degrading ECM in NB. Finally, the article gives examples on how the knowledge is exploited for prognosis and to yield new treatment options for NB patients.
Collapse
Affiliation(s)
- Irena Horwacik
- Laboratory of Molecular Genetics and Virology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| |
Collapse
|
20
|
Liu M, Xu X, Peng K, Hou P, Yuan Y, Li S, Sun X, Shi Z, Zhang J, Dong Y, Liu Q, Ai L, Liang L, Gan L, Huang Q, Yu Y, Liu T. Heparanase modulates the prognosis and development of BRAF V600E-mutant colorectal cancer by regulating AKT/p27Kip1/Cyclin E2 pathway. Oncogenesis 2022; 11:58. [PMID: 36130926 PMCID: PMC9492760 DOI: 10.1038/s41389-022-00428-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 11/09/2022] Open
Abstract
BRAF V600E-mutant colorectal cancer (CRC) is a rare subtype of colorectal cancer with poor prognosis. Compelling evidence indicates that the heparanase (HPSE) gene has multiple functions in cancer, however, its role in BRAF V600E-mutant CRC remains elusive. Differentially expressed genes between BRAF V600E-mutant and wild-type patients were explored by analyzing public data from The Cancer Genome Atlas and the Gene Expression Omnibus. Clinical samples of 172 patients with BRAF V600E-mutant CRC diagnosed at Zhongshan Hospital Fudan University were collected. Overall survival was analyzed using Kaplan-Meier curves and Cox regression models. Cell models and xenografts were utilized to investigate the effect of HPSE on tumor proliferation. HPSE was significantly highly expressed in the BRAF V600E-mutant group. High HPSE expression level was independently associated with inferior survival in the BRAF V600E-mutant cohort. HPSE knockdown impeded tumor proliferation of BRAF V600E-mutant CRC cells in vitro and in vivo. Mechanistically, HPSE silencing arrested cell cycle in G0/G1 phase by downregulating Cyclin E2 expression via the AKT/p27Kip1 pathway. These findings support a role for HPSE in promoting BRAF V600E-mutant CRC progression, which suggests it holds great promise as a prognostic biomarker and a potential therapeutic target for the aggressive CRC subtype.
Collapse
Affiliation(s)
- Mengling Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaojing Xu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ke Peng
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Pengcong Hou
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yitao Yuan
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Suyao Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xun Sun
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhongyi Shi
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jiayu Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yu Dong
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Qing Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Luoyan Ai
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Li Liang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lu Gan
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Qihong Huang
- Department of Clinical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yiyi Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Center of Evidence-based Medicine, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
21
|
Inflammation- and Metastasis-Related Proteins Expression Changes in Early Stages in Tumor and Non-Tumor Adjacent Tissues of Colorectal Cancer Samples. Cancers (Basel) 2022; 14:cancers14184487. [PMID: 36139645 PMCID: PMC9497293 DOI: 10.3390/cancers14184487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/05/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Non-tumor adjacent tissue plays a key role in colorectal cancer development, as well as chronic inflammation, but their role has not yet been dilucidated. In addition, inflammation is a process which is related to epithelial-mesenchymal transition and metastasis, but their changes across the different colorectal cancer stages are not fully studied. Understanding how these processes participate in all colorectal cancer phases can be key to a better understanding of the disease. Abstract Chronic inflammation can induce malignant cell transformation, having an important role in all colorectal cancer (CRC) phases. Non-tumor adjacent tissue plays an important role in tumor progression, but its implication in CRC has not yet been fully elucidated. The aim was to analyze the expression of inflammatory, epithelial-mesenchymal transition (EMT), and metastasis-related proteins in both tumor and non-tumor adjacent tissues from CRC patients by western blot. Tumor tissue presented an increase in metastasis and EMT-related proteins compared to non-tumor adjacent tissue, especially in stage II. Tumor tissue stage II also presented an increase in inflammatory-related proteins compared to other stages, which was also seen in non-tumor adjacent tissue stage II. Additionally, the relapse-free survival study of Vimentin and VEGF-B expression levels in stage II patients showed that the higher the expression levels of each protein, the lower 10-year relapse-free survival. These could suggest that some metastasis-related signalling pathways may be activated in stage II in tumor tissue, accompanied by an increase in inflammation. Furthermore, non-tumor adjacent tissue presented an increase of the inflammatory status that could be the basis for future tumor progression. In conclusion, these proteins could be useful as biomarkers of diagnosis for CRC at early stages.
Collapse
|
22
|
Masola V, Franchi M, Zaza G, Atsina FM, Gambaro G, Onisto M. Heparanase regulates EMT and cancer stem cell properties in prostate tumors. Front Oncol 2022; 12:918419. [PMID: 35965510 PMCID: PMC9363836 DOI: 10.3389/fonc.2022.918419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Prostate cancer displays a certain phenotypic plasticity that allows for the transition of cells from the epithelial to the mesenchymal state. This process, known as epithelial–mesenchymal transition (EMT), is one of the factors that give the tumor cells greater invasive and migratory capacity with subsequent formation of metastases. In addition, many cancers, including prostate cancer, are derived from a cell population that shows the properties of stem cells. These cells, called cancer stem cells (CSCs) or tumor-initiating cells, not only initiate the tumor process and growth but are also able to mediate metastasis and drug resistance. However, the impact of EMT and CSCs in prostate cancer progression and patient survival is still far from fully understood. Heparanase (HPSE), the sole mammalian endoglycosidase capable of degrading heparan sulfate (HS), is also involved in prostate cancer progression. We had previously proved that HPSE regulates EMT in non-cancerous pathologies. Two prostate cancer cell lines (DU145 and PC3) were silenced and overexpressed for HPSE. Expression of EMT and stemness markers was evaluated. Results showed that the expression of several EMT markers are modified by HPSE expression in both the prostate cancer cell lines analyzed. In the same way, the stemness markers and features are also modulated by HPSE expression. Taken together, the present findings seem to prove a new mechanism of action of HPSE in sustaining prostate cancer growth and diffusion. As for other tumors, these results highlight the importance of HPSE as a potential pharmacological target in prostate cancer treatment.
Collapse
Affiliation(s)
- Valentina Masola
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- *Correspondence: Maurizio Onisto, ; Valentina Masola,
| | - Marco Franchi
- Department of Life Quality Sciences, University of Bologna, Rimini, Italy
| | - Gianluigi Zaza
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | | | - Giovanni Gambaro
- Renal Unit, Department of Medicine, University Hospital of Verona, Verona, Italy
| | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- *Correspondence: Maurizio Onisto, ; Valentina Masola,
| |
Collapse
|
23
|
Xu H, Yang B, Ren Z, Wu D, Hu A, Hu J. miR-429 negatively regulates the progression of hypoxia-induced retinal neovascularization by the HPSE-VEGF pathway. Exp Eye Res 2022; 223:109196. [PMID: 35872179 DOI: 10.1016/j.exer.2022.109196] [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: 03/10/2022] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 11/26/2022]
Abstract
Heparanase (HPSE) and vascular endothelial growth factor (VEGF) are believed to play a vital role in hypoxia-induced retinal neovascularization (RNV). HPSE is a target gene of miR-429. Our study aimed to investigate the effect of the miR-429-HPSE-VEGF pathway on hypoxia-induced RNV. The gene and protein expression of miR-429, HPSE and VEGF in human retinal endothelial cells and retinas was determined by real-time PCR and Western blot assays. The effects of miR-429 on human retinal endothelial cells and retinal neovascularization under hypoxia condition were verified by in vitro and in vivo experiments. First, we studied the effect of the miR-429-HPSE-VEGF pathway in HRECs under hypoxic conditions. HREC functions such as migration and tube formation were enhanced under hypoxic conditions. Overexpression of miR-429 in HRECs reversed these changes. Then, we investigated the effect of miR-429 on hypoxia-induced RNV in vivo. When miR-429 agomirs were injected into the vitreous cavity of mice with oxygen-induced retinopathy to overexpress miR-429, the mRNA and protein expression of VEGF was significantly reduced. In addition, indicators of retinal neovascularization, such as the retinal avascular area, and morphology of vessels, were reduced significantly in the miR-429 overexpression group. In this study, our data showed that miR-429 plays an important role by inhibiting the HPSE-VEGF pathway in hypoxia-induced retinopathy.
Collapse
Affiliation(s)
- Haiyan Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Bing Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Zewen Ren
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Dongjing Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Andina Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Jie Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| |
Collapse
|
24
|
Alorda-Clara M, Torrens-Mas M, Morla-Barcelo PM, Roca P, Sastre-Serra J, Pons DG, Oliver J. High Concentrations of Genistein Decrease Cell Viability Depending on Oxidative Stress and Inflammation in Colon Cancer Cell Lines. Int J Mol Sci 2022; 23:ijms23147526. [PMID: 35886874 PMCID: PMC9323408 DOI: 10.3390/ijms23147526] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/01/2022] [Accepted: 07/06/2022] [Indexed: 12/24/2022] Open
Abstract
Genistein could play a crucial role in modulating three closely linked physiological processes altered during cancer: oxidative stress, mitochondrial biogenesis, and inflammation. However, genistein’s role in colorectal cancer remains unclear. We aimed to determine genistein’s effects in two colon cancer cells: HT29 and SW620, primary and metastatic cancer cells, respectively. After genistein treatment for 48 h, cell viability and hydrogen peroxide (H2O2) production were studied. The cell cycle was studied by flow cytometry, mRNA and protein levels were analyzed by RT-qPCR and Western blot, respectively, and finally, cytoskeleton remodeling and NF-κB translocation were determined by confocal microscopy. Genistein 100 µM decreased cell viability and produced G2/M arrest, increased H2O2, and produced filopodia in SW620 cells. In HT29 cells, genistein produced an increase of cell death, H2O2 production, and in the number of stress fibers. In HT29 cells, mitochondrial biogenesis was increased, however, in SW620 cells, it was decreased. Finally, the expression of inflammation-related genes increased in both cell lines, being greater in SW620 cells, where NF-κB translocation to the nucleus was higher. These results indicate that high concentrations of genistein could increase oxidative stress and inflammation in colon cancer cells and, ultimately, decrease cell viability.
Collapse
Affiliation(s)
- Marina Alorda-Clara
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, E-07122 Palma de Mallorca, Illes Balears, Spain; (M.A.-C.); (M.T.-M.); (P.M.M.-B.); (P.R.); (J.S.-S.); (J.O.)
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Edificio S, E-07120 Palma de Mallorca, Illes Balears, Spain
| | - Margalida Torrens-Mas
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, E-07122 Palma de Mallorca, Illes Balears, Spain; (M.A.-C.); (M.T.-M.); (P.M.M.-B.); (P.R.); (J.S.-S.); (J.O.)
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Edificio S, E-07120 Palma de Mallorca, Illes Balears, Spain
- Translational Research in Aging and Longevity (TRIAL) Group, Instituto de Investigación Sanitaria Illes Balears (IdISBa), E-07120 Palma de Mallorca, Illes Balears, Spain
| | - Pere Miquel Morla-Barcelo
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, E-07122 Palma de Mallorca, Illes Balears, Spain; (M.A.-C.); (M.T.-M.); (P.M.M.-B.); (P.R.); (J.S.-S.); (J.O.)
| | - Pilar Roca
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, E-07122 Palma de Mallorca, Illes Balears, Spain; (M.A.-C.); (M.T.-M.); (P.M.M.-B.); (P.R.); (J.S.-S.); (J.O.)
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Edificio S, E-07120 Palma de Mallorca, Illes Balears, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, E-28029 Madrid, Madrid, Spain
| | - Jorge Sastre-Serra
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, E-07122 Palma de Mallorca, Illes Balears, Spain; (M.A.-C.); (M.T.-M.); (P.M.M.-B.); (P.R.); (J.S.-S.); (J.O.)
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Edificio S, E-07120 Palma de Mallorca, Illes Balears, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, E-28029 Madrid, Madrid, Spain
| | - Daniel Gabriel Pons
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, E-07122 Palma de Mallorca, Illes Balears, Spain; (M.A.-C.); (M.T.-M.); (P.M.M.-B.); (P.R.); (J.S.-S.); (J.O.)
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Edificio S, E-07120 Palma de Mallorca, Illes Balears, Spain
- Correspondence: ; Tel.: +34-971173149
| | - Jordi Oliver
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, E-07122 Palma de Mallorca, Illes Balears, Spain; (M.A.-C.); (M.T.-M.); (P.M.M.-B.); (P.R.); (J.S.-S.); (J.O.)
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Edificio S, E-07120 Palma de Mallorca, Illes Balears, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, E-28029 Madrid, Madrid, Spain
| |
Collapse
|
25
|
Wang H, You Y, Zhu X. The Role of Exosomes in the Progression and Therapeutic Resistance of Hematological Malignancies. Front Oncol 2022; 12:887518. [PMID: 35692747 PMCID: PMC9178091 DOI: 10.3389/fonc.2022.887518] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/28/2022] [Indexed: 12/13/2022] Open
Abstract
Exosomes are membrane limited structures which derive from cell membranes and cytoplasm. When released into extracellular space, they circulate through the extracellular fluid, including the peripheral blood and tissue fluid. Exosomes surface molecules mediate their targeting to specific recipient cells and deliver their contents to recipient cells by receptor-ligand interaction and/or phagocytosis and/or endocytosis or direct fusion with cell membrane. Exosomes contain many functional molecules, including nucleic acids (DNAs, mRNAs, non-coding RNAs), proteins (transcription factors, enzymes), and lipids which have biological activity. By passing these cargos, exosomes can transfer information between cells. In this way, exosomes are extensively involved in physiological and pathological processes, such as angiogenesis, matrix reprogramming, coagulation, tumor progression. In recent years, researcher have found that exosomes from malignant tumors can mediate information exchange between tumor cells or between tumor cells and non-tumor cells, thereby promoting tumor survival, progression, and resistance to therapy. In this review, we discuss the pro-tumor and anti-therapeutic effects of exosomes in hematological malignancies, hoping to contribute to the early conquest of hematological malignancy.
Collapse
Affiliation(s)
- Haobing Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong You
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojian Zhu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Xiaojian Zhu,
| |
Collapse
|
26
|
Ginini L, Billan S, Fridman E, Gil Z. Insight into Extracellular Vesicle-Cell Communication: From Cell Recognition to Intracellular Fate. Cells 2022; 11:1375. [PMID: 35563681 PMCID: PMC9101098 DOI: 10.3390/cells11091375] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 01/27/2023] Open
Abstract
Extracellular vesicles (EVs) are heterogamous lipid bilayer-enclosed membranous structures secreted by cells. They are comprised of apoptotic bodies, microvesicles, and exosomes, and carry a range of nucleic acids and proteins that are necessary for cell-to-cell communication via interaction on the cells surface. They initiate intracellular signaling pathways or the transference of cargo molecules, which elicit pleiotropic responses in recipient cells in physiological processes, as well as pathological processes, such as cancer. It is therefore important to understand the molecular means by which EVs are taken up into cells. Accordingly, this review summarizes the underlying mechanisms involved in EV targeting and uptake. The primary method of entry by EVs appears to be endocytosis, where clathrin-mediated, caveolae-dependent, macropinocytotic, phagocytotic, and lipid raft-mediated uptake have been variously described as being prevalent. EV uptake mechanisms may depend on proteins and lipids found on the surfaces of both vesicles and target cells. As EVs have been shown to contribute to cancer growth and progression, further exploration and targeting of the gateways utilized by EVs to internalize into tumor cells may assist in the prevention or deceleration of cancer pathogenesis.
Collapse
Affiliation(s)
- Lana Ginini
- Rappaport Family Institute for Research in the Medical Sciences, Technion–Israel Institute of Technology, Haifa 31096, Israel; (L.G.); (E.F.)
| | - Salem Billan
- Head and Neck Institute, The Holy Family Hospital Nazareth, Nazareth 1641100, Israel;
- Medical Oncology and Radiation Therapy Program, Oncology Section, Rambam Health Care Campus, HaAliya HaShniya Street 8, Haifa 3109601, Israel
| | - Eran Fridman
- Rappaport Family Institute for Research in the Medical Sciences, Technion–Israel Institute of Technology, Haifa 31096, Israel; (L.G.); (E.F.)
| | - Ziv Gil
- Head and Neck Institute, The Holy Family Hospital Nazareth, Nazareth 1641100, Israel;
| |
Collapse
|
27
|
Production, characteristics and applications of microbial heparinases. Biochimie 2022; 198:109-140. [DOI: 10.1016/j.biochi.2022.03.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/03/2022] [Accepted: 03/28/2022] [Indexed: 12/26/2022]
|
28
|
Tang WQ, Hei Y, Lin J. Heparanase-1 is downregulated in chemoradiotherapy orbital rhabdomyosarcoma and relates with tumor growth as well as angiogenesis. Int J Ophthalmol 2022; 15:31-39. [PMID: 35047353 DOI: 10.18240/ijo.2022.01.05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 08/20/2021] [Indexed: 11/23/2022] Open
Abstract
AIM To determine the role of heparanase-1 (HPSE-1) in orbital rhabdomyosarcoma (RMS), and to investigate the feasibility of HPSE-1 targeted therapy for RMS. METHODS Immunohistochemistry was performed to analyze HPSE-1 expression in 51 cases of orbital RMS patients (including 28 cases of embryonal RMS and 23 cases of alveolar RMS), among whom there were 27 treated and 24 untreated with preoperative chemoradiotherapy. In vitro, studies were conducted to examine the effect of HPSE-1 silencing on RMS cell proliferation and tube formation of human umbilical vein endothelial cells (HUVECs). RD cells (an RMS cell line) and HUVECs were infected with HPSE-1 shRNA lentivirus at a multiplicity of infection (MOI) of 10 and 30 separately. Real-time PCR and Western blot were applied to detect the mRNA and protein expression levels of HPSE-1. Cell viability of treated or control RD cells was evaluated by cell counting kit-8 (CCK-8) assay. Matrigel tube formation assay was used to evaluate the effect of HPSE-1 RNAi on the tube formation of HUVECs. RESULTS Immunohistochemistry showed that the expression rate of HPSE-1 protein was 92.9% in orbital embryonal RMS and 91.3% in orbital alveolar RMS. Tissue from alveolar orbital RMS did not show relatively stronger staining than that from the embryonal orbital RMS. However, despite the types of RMS, comparing the cases treated chemoradiotherapy with those untreated, we have observed that chemoradiotherapy resulted in weaker staining in patients' tissues. The expression levels of HPSE-1 declined significantly in both the mRNA and protein levels in HPSE-1 shRNA transfected RD cells. The CCK-8 assay showed that lentivirus-mediated HPSE-1 silencing resulted in significantly reduced RD cells viability in vitro. Silencing HPSE-1 expression also inhibited VEGF-induced tube formation of HUVECs in Matrigel. CONCLUSION HPSE-1 silencing may be a promising therapy for the inhibition of orbital RMS progression.
Collapse
Affiliation(s)
- Wei-Qiang Tang
- Department of Ophthalmology, the Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
| | - Yan Hei
- Department of Ophthalmology, the Third Medical Centre, Chinese PLA General Hospital, Beijing 100039, China
| | - Jing Lin
- Department of Clinical Laboratory, the Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
| |
Collapse
|
29
|
Masola V, Greco N, Gambaro G, Franchi M, Onisto M. Heparanase as active player in endothelial glycocalyx remodeling. Matrix Biol Plus 2022; 13:100097. [PMID: 35036899 PMCID: PMC8749438 DOI: 10.1016/j.mbplus.2021.100097] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022] Open
Abstract
The surface of all animal cells is coated with a layer of carbohydrates linked in various ways to the outer side of the plasma membrane. These carbohydrates are mainly bound to proteins in the form of glycoproteins and proteoglycans and together with the glycolipids constitute the so-called glycocalyx. In particular, the endothelial glycocalyx that covers the luminal layer of the endothelium is composed of glycosaminoglycans (heparan sulphate -HS and hyaluronic acid -HA), proteoglycans (syndecans and glypicans) and adsorbed plasma proteins. Thanks to its ability to absorb water, this structure contributes to making the surface of the vessels slippery but at the same time acts by modulating the mechano-transduction of the vessels, the vascular permeability and the adhesion of leukocytes in thus regulating several physiological and pathological events. Among the various enzymes involved in the degradation of the glycocalyx, heparanase (HPSE) has been shown to be particularly involved. This enzyme is responsible for the cutting of heparan sulfate (HS) chains at the level of the proteoglycans of the endothelial glycocalyx whose dysfunction appears to have a role in organ fibrosis, sepsis and viral infection. In this mini-review, we describe the mechanisms by which HPSE contributes to glycocalyx remodeling and then examine the role of glycocalyx degradation in the development of pathological conditions and pharmacological strategies to preserve glycocalyx during disease pathogenesis.
Collapse
Affiliation(s)
- Valentina Masola
- Renal Unit, Dept. of Medicine, University Hospital of Verona, Verona, Italy.,Dept. of Biomedical Sciences, University of Padova, Padua, Italy
| | - Nicola Greco
- Dept. of Biomedical Sciences, University of Padova, Padua, Italy
| | - Giovanni Gambaro
- Renal Unit, Dept. of Medicine, University Hospital of Verona, Verona, Italy
| | - Marco Franchi
- Dept. of Life Quality Sciences, University of Bologna, Rimini, Italy
| | - Maurizio Onisto
- Dept. of Biomedical Sciences, University of Padova, Padua, Italy
| |
Collapse
|
30
|
Knani I, Singh P, Gross-Cohen M, Aviram S, Ilan N, Sanderson RD, Aronheim A, Vlodavsky I. Induction of heparanase 2 (Hpa2) expression by stress is mediated by ATF3. Matrix Biol 2022; 105:17-30. [PMID: 34808335 PMCID: PMC8821145 DOI: 10.1016/j.matbio.2021.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/10/2021] [Accepted: 11/14/2021] [Indexed: 01/03/2023]
Abstract
Activity of heparanase, endoglycosidase that cleaves heparan sulfate side chains in heparan sulfate proteoglycans, is highly implicated in tumor progression and metastasis. Heparanase inhibitors are therefore being evaluated clinically as anti-cancer therapeutics. Heparanase 2 (Hpa2) is a close homolog of heparanase that lacks HS-degrading activity and functions as an endogenous inhibitor of heparanase. As a result, Hpa2 appears to attenuate tumor growth but mechanisms that regulate Hpa2 expression and determine the ratio between heparanase and Hpa2 are largely unknown. We have recently reported that the expression of Hpa2 is induced by endoplasmic reticulum (ER) and proteotoxic stresses, but the mechanism(s) underlying Hpa2 gene regulation was obscure. Here we expand the notion that Hpa2 is regulated by conditions of stress. We report that while ER and hypoxia, each alone, resulted in a 3-7 fold increase in Hpa2 expression, combining ER stress and hypoxia resulted in a noticeable, over 40-fold increase in Hpa2 expression. A prominent induction of Hpa2 expression was also quantified in cells exposed to heat shock, proteotoxic stress, lysosomal stress, and chemotherapy (cisplatin), strongly implying that Hpa2 is regulated by conditions of stress. Furthermore, analyses of the Hpa2 gene promoter led to the identification of activating-transcription-factor 3 (ATF3) as a transcription factor that mediates Hpa2 induction by stress, thus revealing, for the first time, a molecular mechanism that underlies Hpa2 gene regulation. Induction of Hpa2 and ATF3 by conditions of stress that often accompany the rapid expansion of tumors is likely translated to improved survival of cancer patients.
Collapse
Affiliation(s)
- Ibrahim Knani
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Preeti Singh
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Miriam Gross-Cohen
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Sharon Aviram
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Neta Ilan
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Ralph D Sanderson
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ami Aronheim
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Israel Vlodavsky
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel,Correspondence should be addressed: Israel Vlodavsky, Technion Integrated Cancer Center, Bruce Rappaport Faculty of Medicine; Technion, P. O. Box 9649, Haifa 31096, Israel,
| |
Collapse
|
31
|
Whitefield C, Hong N, Mitchell JA, Jackson CJ. Computational design and experimental characterisation of a stable human heparanase variant. RSC Chem Biol 2022; 3:341-349. [PMID: 35382258 PMCID: PMC8905545 DOI: 10.1039/d1cb00239b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/11/2022] [Indexed: 11/25/2022] Open
Abstract
Heparanase is the only human enzyme known to hydrolyse heparin sulfate and is involved in many important physiological processes. However, it is also unregulated in many disease states, such as cancer, diabetes and Covid-19. It is thus an important drug target, yet the heterologous production of heparanase is challenging and only possible in mammalian or insect expression systems, which limits the ability of many laboratories to study it. Here we describe the computational redesign of heparanase to allow high yield expression in Escherchia coli. This mutated form of heparanase exhibits essentially identical kinetics, inhibition, structure and protein dynamics to the wild type protein, despite the presence of 26 mutations. This variant will facilitate wider study of this important enzyme and contributes to a growing body of literature that shows evolutionarily conserved and functionally neutral mutations can have significant effects on protein folding and expression. A mutant heparanase that exhibits wild type structure and activity but can be heterologously produced in bacterial protein expression systems.![]()
Collapse
Affiliation(s)
- Cassidy Whitefield
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
| | - Nansook Hong
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
| | - Joshua A. Mitchell
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
| | - Colin J. Jackson
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Australian National University, Canberra, ACT 2601, Australia
| |
Collapse
|
32
|
Chen CG, Iozzo RV. Extracellular matrix guidance of autophagy: a mechanism regulating cancer growth. Open Biol 2022; 12:210304. [PMID: 34982945 PMCID: PMC8727153 DOI: 10.1098/rsob.210304] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/01/2021] [Indexed: 01/09/2023] Open
Abstract
The extracellular matrix (ECM) exists as a dynamic network of biophysical and biochemical factors that maintain tissue homeostasis. Given its sensitivity to changes in the intra- and extracellular space, the plasticity of the ECM can be pathological in driving disease through aberrant matrix remodelling. In particular, cancer uses the matrix for its proliferation, angiogenesis, cellular reprogramming and metastatic spread. An emerging field of matrix biology focuses on proteoglycans that regulate autophagy, an intracellular process that plays both critical and contextual roles in cancer. Here, we review the most prominent autophagic modulators from the matrix and the current understanding of the cellular pathways and signalling cascades that mechanistically drive their autophagic function. We then critically assess how their autophagic functions influence tumorigenesis, emphasizing the complexities and stage-dependent nature of this relationship in cancer. We highlight novel emerging data on immunoglobulin-containing and proline-rich receptor-1, heparanase and thrombospondin 1 in autophagy and cancer. Finally, we further discuss the pro- and anti-autophagic modulators originating from the ECM, as well as how these proteoglycans and other matrix constituents specifically influence cancer progression.
Collapse
Affiliation(s)
- Carolyn G. Chen
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
33
|
Lanzi C, Favini E, Dal Bo L, Tortoreto M, Arrighetti N, Zaffaroni N, Cassinelli G. Upregulation of ERK-EGR1-heparanase axis by HDAC inhibitors provides targets for rational therapeutic intervention in synovial sarcoma. J Exp Clin Cancer Res 2021; 40:381. [PMID: 34857011 PMCID: PMC8638516 DOI: 10.1186/s13046-021-02150-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 10/21/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Synovial sarcoma (SS) is an aggressive soft tissue tumor with limited therapeutic options in advanced stage. SS18-SSX fusion oncogenes, which are the hallmarks of SS, cause epigenetic rewiring involving histone deacetylases (HDACs). Promising preclinical studies supporting HDAC targeting for SS treatment were not reflected in clinical trials with HDAC inhibitor (HDACi) monotherapies. We investigated pathways implicated in SS cell response to HDACi to identify vulnerabilities exploitable in combination treatments and improve the therapeutic efficacy of HDACi-based regimens. METHODS Antiproliferative and proapoptotic effects of the HDACi SAHA and FK228 were examined in SS cell lines in parallel with biochemical and molecular analyses to bring out cytoprotective pathways. Treatments combining HDACi with drugs targeting HDACi-activated prosurvival pathways were tested in functional assays in vitro and in a SS orthotopic xenograft model. Molecular mechanisms underlying synergisms were investigated in SS cells through pharmacological and gene silencing approaches and validated by qRT-PCR and Western blotting. RESULTS SS cell response to HDACi was consistently characterized by activation of a cytoprotective and auto-sustaining axis involving ERKs, EGR1, and the β-endoglycosidase heparanase, a well recognized pleiotropic player in tumorigenesis and disease progression. HDAC inhibition was shown to upregulate heparanase by inducing expression of the positive regulator EGR1 and by hampering negative regulation by p53 through its acetylation. Interception of HDACi-induced ERK-EGR1-heparanase pathway by cell co-treatment with a MEK inhibitor (trametinib) or a heparanase inhibitor (SST0001/roneparstat) enhanced antiproliferative and pro-apoptotic effects. HDAC and heparanase inhibitors had opposite effects on histone acetylation and nuclear heparanase levels. The combination of SAHA with SST0001 prevented the upregulation of ERK-EGR1-heparanase induced by the HDACi and promoted caspase-dependent cell death. In vivo, the combined treatment with SAHA and SST0001 potentiated the antitumor efficacy against the CME-1 orthotopic SS model as compared to single agent administration. CONCLUSIONS The present study provides preclinical rationale and mechanistic insights into drug combinatory strategies based on the use of ERK pathway and heparanase inhibitors to improve the efficacy of HDACi-based antitumor therapies in SS. The involvement of classes of agents already clinically available, or under clinical evaluation, indicates the transferability potential of the proposed approaches.
Collapse
Affiliation(s)
- Cinzia Lanzi
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Enrica Favini
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Laura Dal Bo
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Monica Tortoreto
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Noemi Arrighetti
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Nadia Zaffaroni
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Giuliana Cassinelli
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy.
| |
Collapse
|
34
|
Ravi Kiran AVVV, Kusuma Kumari G, Krishnamurthy PT, Khaydarov RR. Tumor microenvironment and nanotherapeutics: intruding the tumor fort. Biomater Sci 2021; 9:7667-7704. [PMID: 34673853 DOI: 10.1039/d1bm01127h] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over recent years, advancements in nanomedicine have allowed new approaches to diagnose and treat tumors. Nano drug delivery systems exploit the enhanced permeability and retention (EPR) effect and enter the tumor tissue's interstitial space. However, tumor barriers play a crucial role, and cause inefficient EPR or the homing effect. Mounting evidence supports the hypothesis that the components of the tumor microenvironment, such as the extracellular matrix, and cellular and physiological components collectively or cooperatively hinder entry and distribution of drugs, and therefore, limit the theragnostic applications of cancer nanomedicine. This abnormal tumor microenvironment plays a pivotal role in cancer nanomedicine and was recently recognized as a promising target for improving nano-drug delivery and their therapeutic outcomes. Strategies like passive or active targeting, stimuli-triggered nanocarriers, and the modulation of immune components have shown promising results in achieving anticancer efficacy. The present review focuses on the tumor microenvironment and nanoparticle-based strategies (polymeric, inorganic and organic nanoparticles) for intruding the tumor barrier and improving therapeutic effects.
Collapse
Affiliation(s)
- Ammu V V V Ravi Kiran
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Garikapati Kusuma Kumari
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Praveen T Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Renat R Khaydarov
- Institute of Nuclear Physics, Uzbekistan Academy of Sciences, Tashkent, 100047, Uzbekistan.
| |
Collapse
|
35
|
Heparanase (HPSE) Associates with the Tumor Immune Microenvironment in Colorectal Cancer. Processes (Basel) 2021. [DOI: 10.3390/pr9091605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
There is an unmet clinical need to identify potential predictive biomarkers for immunotherapy efficacy in mismatch repair proficient (pMMR) metastatic colorectal cancer (mCRC). Heparanase (HPSE) is a multifunctional molecule mediating tumor–host crosstalk. However, the function of HPSE in the tumor immune microenvironment of CRC remains unclear. Data of CRC patients from public datasets (TCGA and GSE39582) and Zhongshan Hospital (ZS cohort) were collected to perform integrative bioinformatic analyses. In total, 1036 samples from TCGA (N = 457), GSE39582 (N = 510) and ZS cohort (N = 69) were included. Samples of deficient MMR (dMMR) and consensus molecular subtypes 1 (CMS1) showed significantly higher HPSE expression. The expression of HPSE also exhibited a significantly positive association with PD-L1 expression, tumor mutation burden and the infiltration of macrophages. Immune pathways were remarkably enriched in the HPSE high-expression group, which also showed higher expressions of chemokines and immune checkpoint genes. Survival analysis suggested that high HPSE expression tended to be associated with shorter overall survival in patients with pMMR mCRC. HPSE might contribute to the immune-activated tumor microenvironment with high levels of immune checkpoint molecules, suggesting that pMMR mCRC with high HPSE expression might respond to immune checkpoint inhibitors.
Collapse
|
36
|
Gross-Cohen M, Yanku Y, Kessler O, Barash U, Boyango I, Cid-Arregui A, Neufeld G, Ilan N, Vlodavsky I. Heparanase 2 (Hpa2) attenuates tumor growth by inducing Sox2 expression. Matrix Biol 2021; 99:58-71. [PMID: 34004353 DOI: 10.1016/j.matbio.2021.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/06/2021] [Accepted: 05/06/2021] [Indexed: 12/11/2022]
Abstract
The pro-tumorigenic properties of heparanase are well documented, and heparanase inhibitors are being evaluated clinically as anti-cancer therapeutics. In contrast, the role of heparanase 2 (Hpa2), a close homolog of heparanase, in cancer is largely unknown. Previously, we have reported that in head and neck cancer, high levels of Hpa2 are associated with prolonged patient survival and decreased tumor cell dissemination to regional lymph nodes, suggesting that Hpa2 functions to restrain tumorigenesis. Also, patients with high levels of Hpa2 were diagnosed as low grade and exhibited increased expression of cytokeratins, an indication that Hpa2 promotes or maintains epithelial cell differentiation and identity. To reveal the molecular mechanism underlying the tumor suppressor properties of Hpa2, and its ability to induce the expression of cytokeratin, we employed overexpression as well as gene editing (Crispr) approaches, combined with gene array and RNAseq methodologies. At the top of the list of many genes found to be affected by Hpa2 was Sox2. Here we provide evidence that silencing of Sox2 resulted in bigger tumors endowed with reduced cytokeratin levels, whereas smaller tumors were developed by cells overexpressing Sox2, suggesting that in head and neck carcinoma, Sox2 functions to inhibit tumor growth. Notably, Hpa2-null cells engineered by Crispr/Cas 9, produced bigger tumors vs control cells, and rescue of Hpa2 attenuated tumor growth. These results strongly imply that Hpa2 functions as a tumor suppressor in head and neck cancer, involving Sox2 upregulation mediated, in part, by the high-affinity interaction of Hpa2 with heparan sulfate.
Collapse
Affiliation(s)
- Miriam Gross-Cohen
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Yifat Yanku
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Ofra Kessler
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Uri Barash
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Ilanit Boyango
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | | | - Gera Neufeld
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Neta Ilan
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Israel Vlodavsky
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion, Haifa, Israel.
| |
Collapse
|
37
|
Kaur R, Deb PK, Diwan V, Saini B. Heparanase Inhibitors in Cancer Progression: Recent Advances. Curr Pharm Des 2021; 27:43-68. [PMID: 33185156 DOI: 10.2174/1381612826666201113105250] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 08/25/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND An endo-β-glucuronidase enzyme, Heparanase (HPSE), degrades the side chains of polymeric heparan sulfate (HS), a glycosaminoglycan formed by alternate repetitive units of D-glucosamine and D-glucuronic acid/L-iduronic acid. HS is a major component of the extracellular matrix and basement membranes and has been implicated in processes of the tissue's integrity and functional state. The degradation of HS by HPSE enzyme leads to conditions like inflammation, angiogenesis, and metastasis. An elevated HPSE expression with a poor prognosis and its multiple roles in tumor growth and metastasis has attracted significant interest for its inhibition as a potential anti-neoplastic target. METHODS We reviewed the literature from journal publication websites and electronic databases such as Bentham, Science Direct, PubMed, Scopus, USFDA, etc., about HPSE, its structure, functions, and role in cancer. RESULTS The present review is focused on Heparanase inhibitors (HPIns) that have been isolated from natural resources or chemically synthesized as new therapeutics for metastatic tumors and chronic inflammatory diseases in recent years. The recent developments made in the HPSE structure and function are also discussed, which can lead to the future design of HPIns with more potency and specificity for the target. CONCLUSION HPIns can be a better target to be explored against various cancers.
Collapse
Affiliation(s)
- Rajwinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Pran Kishore Deb
- Faculty of Pharmacy, Philadelphia University, Philadelphia, Jordan
| | - Vishal Diwan
- Faculty of Medicine, The University of Queensland, Queensland, Australia
| | - Balraj Saini
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
38
|
Yang C, Zhang S, Chang X, Huang Y, Cui D, Liu Z. MicroRNA-219a-2-3p modulates the proliferation of thyroid cancer cells via the HPSE/cyclin D1 pathway. Exp Ther Med 2021; 21:659. [PMID: 33968189 DOI: 10.3892/etm.2021.10091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 03/25/2021] [Indexed: 12/18/2022] Open
Abstract
Heparanase (HPSE) is an endo-β-D-glucuronidase overexpressed in different types of human cancer, and a predicted target of microRNA (miRNA/miR)-219a-2-3p in thyroid cancer. The present study aimed to investigate the potential role of HPSE and miR-219a-2-3p in thyroid cancer, and the molecular mechanism of miR-219a-2-3p regulating the proliferation of thyroid cancer cells via HPSE was confirmed. Immunohistochemistry analysis was performed to detect HPSE expression in thyroid cancer sections. In addition, reverse transcription-quantitative PCR analysis was performed to detect mRNA and miR-219a-2-3p expression levels in thyroid cancer samples and cell lines. miR-219-2-3p mimic or HPSE plasmid were transfected into B-CPAP and TPC-1 thyroid cancer cells. Furthermore, western blot analysis was performed to detect the protein expression levels of HPSE and cyclin D1. Cell cycle analysis was performed using propidium iodide staining and flow cytometry, and EdU incorporation was performed to detect cell proliferation. The results demonstrated that high HPSE expression was significantly associated with tumor size, extracapsular invasion and lymph node metastasis. Notably, a statistically negative correlation was observed between HPSE mRNA expression and miR-219a-2-3p expression in thyroid cancer tumors, as well as in thyroid cancer cell lines. When exogenously expressed in B-CPAP and TPC-1 cells, miR-219a-2-3p induced cell cycle arrest at the G0/G1 phase and decreased the percentage of proliferating cells. Furthermore, HPSE and cyclin D1 protein expression decreased following transfection with miR-219a-2-3p. Notably, when HPSE was ectopically expressed in miR-219a-2-3p transfected cells, cyclin D1 expression and the number of proliferative cells increased. Taken together, these results suggest that HPSE contributes to the proliferation of thyroid cancer cells. In addition, miR-219a-2-3p was confirmed to target HPSE and inhibit cell proliferation, which was associated with cyclin D1 suppression-mediated cell cycle arrest.
Collapse
Affiliation(s)
- Chuanjia Yang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Siyang Zhang
- Science and Experiment Center, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Xiaoying Chang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yonglian Huang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Dongxu Cui
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Zhen Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
39
|
Syndecan-1 (CD138), Carcinomas and EMT. Int J Mol Sci 2021; 22:ijms22084227. [PMID: 33921767 PMCID: PMC8072910 DOI: 10.3390/ijms22084227] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/14/2021] [Indexed: 12/16/2022] Open
Abstract
Cell surface proteoglycans are known to be important regulators of many aspects of cell behavior. The principal family of transmembrane proteoglycans is the syndecans, of which there are four in mammals. Syndecan-1 is mostly restricted to epithelia, and bears heparan sulfate chains that are capable of interacting with a large array of polypeptides, including extracellular matrix components and potent mediators of proliferation, adhesion and migration. For this reason, it has been studied extensively with respect to carcinomas and tumor progression. Frequently, but not always, syndecan-1 levels decrease as tumor grade, stage and invasiveness and dedifferentiation increase. This parallels experiments that show depletion of syndecan-1 can be accompanied by loss of cadherin-mediated adhesion. However, in some tumors, levels of syndecan-1 increase, but the characterization of its distribution is relevant. There can be loss of membrane staining, but acquisition of cytoplasmic and/or nuclear staining that is abnormal. Moreover, the appearance of syndecan-1 in the tumor stroma, either associated with its cellular component or the collagenous matrix, is nearly always a sign of poor prognosis. Given its relevance to myeloma progression, syndecan-1-directed antibody—toxin conjugates are being tested in clinical and preclinical trials, and may have future relevance to some carcinomas.
Collapse
|
40
|
Piperigkou Z, Kyriakopoulou K, Koutsakis C, Mastronikolis S, Karamanos NK. Key Matrix Remodeling Enzymes: Functions and Targeting in Cancer. Cancers (Basel) 2021; 13:1441. [PMID: 33809973 PMCID: PMC8005147 DOI: 10.3390/cancers13061441] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/05/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022] Open
Abstract
Tissue functionality and integrity demand continuous changes in distribution of major components in the extracellular matrices (ECMs) under normal conditions aiming tissue homeostasis. Major matrix degrading proteolytic enzymes are matrix metalloproteinases (MMPs), plasminogen activators, atypical proteases such as intracellular cathepsins and glycolytic enzymes including heparanase and hyaluronidases. Matrix proteases evoke epithelial-to-mesenchymal transition (EMT) and regulate ECM turnover under normal procedures as well as cancer cell phenotype, motility, invasion, autophagy, angiogenesis and exosome formation through vital signaling cascades. ECM remodeling is also achieved by glycolytic enzymes that are essential for cancer cell survival, proliferation and tumor progression. In this article, the types of major matrix remodeling enzymes, their effects in cancer initiation, propagation and progression as well as their pharmacological targeting and ongoing clinical trials are presented and critically discussed.
Collapse
Affiliation(s)
- Zoi Piperigkou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), 265 04 Patras, Greece
| | - Konstantina Kyriakopoulou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
| | - Christos Koutsakis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
| | | | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), 265 04 Patras, Greece
| |
Collapse
|
41
|
Endothelial Glycocalyx as a Regulator of Fibrotic Processes. Int J Mol Sci 2021; 22:ijms22062996. [PMID: 33804258 PMCID: PMC7999025 DOI: 10.3390/ijms22062996] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/02/2021] [Accepted: 03/11/2021] [Indexed: 12/31/2022] Open
Abstract
The endothelial glycocalyx, the gel layer covering the endothelium, is composed of glycosaminoglycans, proteoglycans, and adsorbed plasma proteins. This structure modulates vessels’ mechanotransduction, vascular permeability, and leukocyte adhesion. Thus, it regulates several physiological and pathological events. In the present review, we described the mechanisms that disturb glycocalyx stability such as reactive oxygen species, matrix metalloproteinases, and heparanase. We then focused our attention on the role of glycocalyx degradation in the induction of profibrotic events and on the possible pharmacological strategies to preserve this delicate structure.
Collapse
|
42
|
Jiang S, Fu R, Shi J, Wu H, Mai J, Hua X, Chen H, Liu J, Lu M, Li N. CircRNA-Mediated Regulation of Angiogenesis: A New Chapter in Cancer Biology. Front Oncol 2021; 11:553706. [PMID: 33777729 PMCID: PMC7988083 DOI: 10.3389/fonc.2021.553706] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 01/07/2021] [Indexed: 12/15/2022] Open
Abstract
Angiogenesis is necessary for carcinoma progression and is regulated by a variety of pro- and anti-angiogenesis factors. CircRNAs are RNA molecules that do not have a 5'-cap or a 3'-polyA tail and are involved in a variety of biological functions. While circRNA-mediated regulation of tumor angiogenesis has received much attention, the detailed biological regulatory mechanism remains unclear. In this review, we investigated circRNAs in tumor angiogenesis from multiple perspectives, including its upstream and downstream factors. We believe that circRNAs have natural advantages and great potential for the diagnosis and treatment of tumors, which deserves further exploration.
Collapse
Affiliation(s)
- Shaotao Jiang
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Rongdang Fu
- Department of Hepatic Surgery, The First People's Hospital of Foshan, Affiliated Foshan Hospital of Sun Yat-sen University, Foshan, China
| | - Jiewei Shi
- Department of General Surgery, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Huijie Wu
- Department of Obstetrics, The First People's Hospital of Foshan, Affiliated Foshan Hospital of Sun Yat-sen University, Foshan, China
| | - Jialuo Mai
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xuefeng Hua
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Huan Chen
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jie Liu
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Minqiang Lu
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ning Li
- Department of HBP SURGERY II, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
43
|
Karamanos NK, Theocharis AD, Piperigkou Z, Manou D, Passi A, Skandalis SS, Vynios DH, Orian-Rousseau V, Ricard-Blum S, Schmelzer CEH, Duca L, Durbeej M, Afratis NA, Troeberg L, Franchi M, Masola V, Onisto M. A guide to the composition and functions of the extracellular matrix. FEBS J 2021; 288:6850-6912. [PMID: 33605520 DOI: 10.1111/febs.15776] [Citation(s) in RCA: 362] [Impact Index Per Article: 120.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
Extracellular matrix (ECM) is a dynamic 3-dimensional network of macromolecules that provides structural support for the cells and tissues. Accumulated knowledge clearly demonstrated over the last decade that ECM plays key regulatory roles since it orchestrates cell signaling, functions, properties and morphology. Extracellularly secreted as well as cell-bound factors are among the major members of the ECM family. Proteins/glycoproteins, such as collagens, elastin, laminins and tenascins, proteoglycans and glycosaminoglycans, hyaluronan, and their cell receptors such as CD44 and integrins, responsible for cell adhesion, comprise a well-organized functional network with significant roles in health and disease. On the other hand, enzymes such as matrix metalloproteinases and specific glycosidases including heparanase and hyaluronidases contribute to matrix remodeling and affect human health. Several cell processes and functions, among them cell proliferation and survival, migration, differentiation, autophagy, angiogenesis, and immunity regulation are affected by certain matrix components. Structural alterations have been also well associated with disease progression. This guide on the composition and functions of the ECM gives a broad overview of the matrisome, the major ECM macromolecules, and their interaction networks within the ECM and with the cell surface, summarizes their main structural features and their roles in tissue organization and cell functions, and emphasizes the importance of specific ECM constituents in disease development and progression as well as the advances in molecular targeting of ECM to design new therapeutic strategies.
Collapse
Affiliation(s)
- Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Dimitra Manou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Demitrios H Vynios
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Véronique Orian-Rousseau
- Karlsruhe Institute of Technology, Institute of Biological and Chemical Systems- Functional Molecular Systems, Eggenstein-Leopoldshafen, Germany
| | - Sylvie Ricard-Blum
- University of Lyon, UMR 5246, ICBMS, Université Lyon 1, CNRS, Villeurbanne Cedex, France
| | - Christian E H Schmelzer
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Germany.,Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2: Matrix Aging and Vascular Remodelling, Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Madeleine Durbeej
- Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, Sweden
| | - Nikolaos A Afratis
- Department Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Linda Troeberg
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Norwich, UK
| | - Marco Franchi
- Department for Life Quality Study, University of Bologna, Rimini, Italy
| | | | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, Italy
| |
Collapse
|
44
|
Li QW, Zhang GL, Hao CX, Ma YF, Sun X, Zhang Y, Cao KX, Li BX, Yang GW, Wang XM. SANT, a novel Chinese herbal monomer combination, decreasing tumor growth and angiogenesis via modulating autophagy in heparanase overexpressed triple-negative breast cancer. JOURNAL OF ETHNOPHARMACOLOGY 2021; 266:113430. [PMID: 33011366 DOI: 10.1016/j.jep.2020.113430] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 09/15/2020] [Accepted: 09/26/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Astragalus mongholicus, Solanum nigrum Linn, Lotus plumule, Ligusticum are widely used traditional herbal medicines for cancer treatment in China. They were typical drugs selected from Gubenyiliu II and series of formula (GYII), which were developed on the foundation of YIQIHUOXUEJIEDU theory. In the present study, four active ingredients (Astragaloside IV, α-solanine, neferine, and 2,3,5,6-tetramethylpyrazine) derived from medicines above were applied in combination as SANT. AIM OF THE STUDY Triple-negative breast cancer (TNBC) is a serious threat to women's health worldwide. Heparanase (HPSE) is often up-regulated in breast cancer with the properties of facilitating tumorigenesis and influencing the autophagy process in cancer cells. This study aimed at evaluating the anti-tumor potential of SANT in treating HPSE related TNBC both in-vitro and in-vivo. MATERIALS AND METHODS In this study, we explored the correlation between HPSE expression and survival of breast cancer patients in databases. We performed MTS, trans-well and wound scratch assays to assess the impact of SANT on cell proliferation and migration. Confocal microscopy observation and western blots were applied to verify the autophagy flux induced by SANT. Mice models were employed to evaluate the efficacy and safety of SANT in-vivo by tumor weights and volumes or serum index, respectively. To analyze the underlying mechanisms of SANT, we conducted human autophagy PCR array and angiogenesis proteome profiler on tumor tissues. RESULTS Patients with elevated HPSE expression were associated with a poor outcome in both RFS (P = 1.7e-12) and OS (P = 0.00016). SANT administration significantly inhibited cancer cells' proliferation and migration, enhanced autophagy flux, and slightly reduced the active form of HPSE in-vitro. SANT also suppressed tumor growth and angiogenesis in-vivo. Human autophagy PCR array results indicated that SANT increased the ATG16L1, ATG9B, ATG4D gene expressions while decreased TMEM74 and TNF gene expressions.Angiogenesis proteome profiler results showed SANT reduced protein level of HB-EGF, thrombospondin-2, amphiregulin, leptin, IGFBP-9, EGF, coagulation factor III, and MMP-9 (pro and active form) in tumor, raised the protein expression of serpin E1 and platelet factor 4. CONCLUSIONS These findings indicated that herbal compounds SANT may be a promising candidate in anti-cancer drug discovery. It also provides novel strategies for using natural compounds to achieve optimized effect.
Collapse
MESH Headings
- Angiogenesis Inhibitors/administration & dosage
- Angiogenesis Inhibitors/pharmacology
- Animals
- Antineoplastic Agents, Phytogenic/administration & dosage
- Antineoplastic Agents, Phytogenic/pharmacology
- Autophagy/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Drugs, Chinese Herbal/administration & dosage
- Drugs, Chinese Herbal/pharmacology
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Glucuronidase/genetics
- Humans
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/pathology
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/genetics
- Triple Negative Breast Neoplasms/pathology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Qi-Wei Li
- School of Graduates, Beijing University of Chinese Medicine, Beijing 100029, China; Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Gan-Lin Zhang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China.
| | - Cai-Xia Hao
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Yun-Fei Ma
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Xu Sun
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China; Department of Integrated Traditional Chinese and Western Medicine, The Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou Henan 450008, China
| | - Yi Zhang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Ke-Xin Cao
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Bing-Xue Li
- School of Graduates, Beijing University of Chinese Medicine, Beijing 100029, China; Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Guo-Wang Yang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Xiao-Min Wang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China.
| |
Collapse
|
45
|
Groult H, Carregal-Romero S, Castejón D, Azkargorta M, Miguel-Coello AB, Pulagam KR, Gómez-Vallejo V, Cousin R, Muñoz-Caffarel M, Lawrie CH, Llop J, Piot JM, Elortza F, Maugard T, Ruiz-Cabello J, Fruitier-Arnaudin I. Heparin length in the coating of extremely small iron oxide nanoparticles regulates in vivo theranostic applications. NANOSCALE 2021; 13:842-861. [PMID: 33351869 DOI: 10.1039/d0nr06378a] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The positive contrast of extremely small iron oxide nanoparticles (ESIONP) in magnetic resonance imaging (MRI) rejuvenates this class of metal nanoparticles (NP).Yet, the current synthesis often lacks the possibility of adjusting the core size (while it is a key element for ESIONP-based MRI contrast behaviour), and also involved multiple complex steps before obtaining a ready-to-use probe for medical applications. In this study, we faced these challenges by applying heparin oligosaccharides (HO) of different lengths as coatings for the preparation of HEP-ESIONP with a one-pot microwave method. We demonstrated that the HO length could control the core size during the synthesis to achieve optimal positive MRI contrast, and that HEP-ESIONP were endowed directly with anticoagulant properties and/or a specific antitumor activity, according to the HO used. Relevantly, positron emission tomography (PET)-based in vivo biodistribution study conducted with 68Ga core-doped HEP-ESIONP analogues revealed significant changes in the probe behaviours, the shortening of HO promoting a shift from hepatic to renal clearance. The different conformations of HO coatings and a thorough in vitro characterisation of the probes' protein coronas provided insight into this crucial impact of HO length on opsonization-mediated immune response and elimination. Overall, we were able to identify a precise HO length to get an ESIONP probe showing prolonged vascular lifetime and moderate accumulation in a tumor xenograft, balanced with a low uptake by non-specific organs and favourable urinary clearance. This probe met all prerequisites for advanced theranostic medical applications with a dual MRI/PET hot spot capability and potential antitumor activity.
Collapse
Affiliation(s)
- Hugo Groult
- BCBS team (Biotechnologies et Chimie des Bioressources pour la Santé), LIENSs Laboratory (Littoral environment et Sociétés), UMR CNRS 7266, University of La Rochelle, La Rochelle, France.
| | - Susana Carregal-Romero
- CIC biomaGUNE and Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Gipuzkoa, Spain. and CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - David Castejón
- Unidad de RMN - CAI Bioimagen Complutense, Universidad Complutense de Madrid, Spain
| | - Mikel Azkargorta
- Proteomics Platform CIC bioGUNE, Bizkaia Science and Technology, Derio, Spain
| | - Ana-Beatriz Miguel-Coello
- CIC biomaGUNE and Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Gipuzkoa, Spain.
| | - Krishna Reddy Pulagam
- CIC biomaGUNE and Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Gipuzkoa, Spain.
| | - Vanessa Gómez-Vallejo
- CIC biomaGUNE and Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Gipuzkoa, Spain.
| | - Rémi Cousin
- BCBS team (Biotechnologies et Chimie des Bioressources pour la Santé), LIENSs Laboratory (Littoral environment et Sociétés), UMR CNRS 7266, University of La Rochelle, La Rochelle, France.
| | - María Muñoz-Caffarel
- Molecular Oncology Group, Biodonostia Health Research Institute, San Sebastian, Spain and Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Charles H Lawrie
- Molecular Oncology Group, Biodonostia Health Research Institute, San Sebastian, Spain and Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Jordi Llop
- CIC biomaGUNE and Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Gipuzkoa, Spain. and CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Jean-Marie Piot
- BCBS team (Biotechnologies et Chimie des Bioressources pour la Santé), LIENSs Laboratory (Littoral environment et Sociétés), UMR CNRS 7266, University of La Rochelle, La Rochelle, France.
| | - Felix Elortza
- Proteomics Platform CIC bioGUNE, Bizkaia Science and Technology, Derio, Spain
| | - Thierry Maugard
- BCBS team (Biotechnologies et Chimie des Bioressources pour la Santé), LIENSs Laboratory (Littoral environment et Sociétés), UMR CNRS 7266, University of La Rochelle, La Rochelle, France.
| | - Jesús Ruiz-Cabello
- CIC biomaGUNE and Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián, Gipuzkoa, Spain. and CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain and Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain and Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Madrid, Spain
| | - Ingrid Fruitier-Arnaudin
- BCBS team (Biotechnologies et Chimie des Bioressources pour la Santé), LIENSs Laboratory (Littoral environment et Sociétés), UMR CNRS 7266, University of La Rochelle, La Rochelle, France.
| |
Collapse
|
46
|
Heparan sulfate binds the extracellular Annexin A1 and blocks its effects on pancreatic cancer cells. Biochem Pharmacol 2020; 182:114252. [DOI: 10.1016/j.bcp.2020.114252] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022]
|
47
|
Abstract
Heparanase is the only mammalian enzyme that cleaves heparan sulphate, an important component of the extracellular matrix. This leads to the remodelling of the extracellular matrix, whilst liberating growth factors and cytokines bound to heparan sulphate. This in turn promotes both physiological and pathological processes such as angiogenesis, immune cell migration, inflammation, wound healing and metastasis. Furthermore, heparanase exhibits non-enzymatic actions in cell signalling and in regulating gene expression. Cancer is underpinned by key characteristic features that promote malignant growth and disease progression, collectively termed the 'hallmarks of cancer'. Essentially, all cancers examined to date have been reported to overexpress heparanase, leading to enhanced tumour growth and metastasis with concomitant poor patient survival. With its multiple roles within the tumour microenvironment, heparanase has been demonstrated to regulate each of these hallmark features, in turn highlighting the need for heparanase-targeted therapies. However, recent discoveries which demonstrated that heparanase can also regulate vital anti-tumour mechanisms have cast doubt on this approach. This review will explore the myriad ways by which heparanase functions as a key regulator of the hallmarks of cancer and will highlight its role as a major component within the tumour microenvironment. The dual role of heparanase within the tumour microenvironment, however, emphasises the need for further investigation into defining its precise mechanism of action in different cancer settings.
Collapse
Affiliation(s)
- Krishnath M Jayatilleke
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Plenty Road & Kingsbury Drive, Melbourne, VIC, 3086, Australia
| | - Mark D Hulett
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Plenty Road & Kingsbury Drive, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
48
|
Cao H, Yang S, Yu X, Xi M. Correlation between heparanase gene polymorphism and susceptibility to endometrial cancer. Mol Genet Genomic Med 2020; 8:e1257. [PMID: 32869952 PMCID: PMC7549562 DOI: 10.1002/mgg3.1257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 10/30/2019] [Accepted: 03/01/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Endometrial cancer is one of the three most common malignancies in the female genital tract. Previous studies have demonstrated the association between heparanase (HPSE, OMIM 604,724) single-nucleotide polymorphism (SNP) and cancer risk in several cancers. However, its role in endometrial cancer remains unclear. The present study investigated the effects of HPSE SNPs on the susceptibility and clinicopathological parameters in patients with endometrial cancer. METHODS HPSE SNPs of rs4693608 (G > A) and rs4364254 (C > T) were analyzed using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) assay in 270 endometrial cancer patients and 320 healthy controls. RESULTS The investigation indicated that the HPSE SNP rs4693608 with GG showed a protective effect from EC in both codominant (adjusted OR = 0.41, 95%CI = 0.21-0.81, p = .026) and recessive models (adjusted OR = 0.43, 95%CI = 0.22-0.82, p = .0076). No significant differences were found in the incidences of EC patients with the rs4364254 polymorphisms compared to controls. Moreover, a significantly increased distribution of A/A (rs4693608) was observed in patients with grade ≥ 2 (p = .03) and in patients with positive cervical invasion (p = .042) while patients with T/C (rs4364254) had lower tumor grade. CONCLUSION Our study suggested that HPSE SNP of rs4693608 correlated strongly with susceptibility to EC, and HPSE SNPs might be a potential biomarker for prognosis of endometrial cancer.
Collapse
Affiliation(s)
- Hanyu Cao
- Department of Gynecology and ObstetricsWest China Second University HospitalSichuan UniversityChengduChina
| | - Shuo Yang
- Key Laboratory of Obstetrics, Gynecology, Pediatric Disease, and Birth DefectsMinistry of EducationWest China Second University HospitalChengduChina
| | - Xiuzhang Yu
- Department of Gynecology and ObstetricsWest China Second University HospitalSichuan UniversityChengduChina
- Key Laboratory of Birth Defects and Related Diseases of Women and ChildrenMinistry of EducationChengduChina
| | - Mingrong Xi
- Department of Gynecology and ObstetricsWest China Second University HospitalSichuan UniversityChengduChina
| |
Collapse
|
49
|
Receptor tyrosine kinases and heparan sulfate proteoglycans: Interplay providing anticancer targeting strategies and new therapeutic opportunities. Biochem Pharmacol 2020; 178:114084. [DOI: 10.1016/j.bcp.2020.114084] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/04/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022]
|
50
|
Li S, Fu H, Wang Y, Wang L, Jia B, Bian Y. Curcumin inhibits CT26 cells metastasis by decreasing heparanase expression. J Leukoc Biol 2020; 108:1727-1733. [PMID: 32640496 DOI: 10.1002/jlb.1ma0620-357r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/29/2020] [Accepted: 06/16/2020] [Indexed: 12/22/2022] Open
Abstract
This study tested the hypothesis that heparanase (HPSE) is related to tumor metastasis and curcumin (CCM) inhibits tumor metastasis by down-regulating HPSE expression. MTT, Transwell assays, and RT-PCR were used to study the effects of CCM on the migration and invasion of CT26 cells and the expression of HPSE. CT26 cells were transfected with lentivirus to establish HPSE-overexpressing cells (OE) and corresponding negative control cells (NC). Signal pathways involved in down-regulating the expression of HPSE and inhibiting the migration and invasion of CT26 cells by CCM were screened by the liquid crystal chip. HPSE promoted CT26 cells migration and invasion, and CCM inhibited the proliferation and metastasis of CT26 cells. The results of RT-PCR indicated that CCM down-regulated HPSE expression. Liquid phase microarray showed that CCM inhibited the phosphorylation of P38 and STAT5 in CT26 cells and NC cells. In contrast, the inhibitory function of CCM was markedly enhanced when HPSE was overexpressed (P < 0.05). In short, HPSE is closely related to metastasis of colon cancer cells. CCM inhibits colon cancer cell migration and invasion by inhibiting HPSE expression, which may be related to P38 MAPK and JAK/STAT5 signal pathways.
Collapse
Affiliation(s)
- Shanshan Li
- Tianjin Key Laboratory of Early Human Development and Reproduction Regulation, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Hui Fu
- College of Integrated Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yiyang Wang
- College of Integrated Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Li Wang
- Pharmacy Department, Tianjin Second People's Hospital, Tianjin, China
| | - Beitian Jia
- College of Integrated Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhong Bian
- College of Integrated Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|