1
|
Fujii Y, Asadi Z, Mehla K. Cathepsins: Emerging targets in the tumor ecosystem to overcome cancers. Semin Cancer Biol 2025; 112:150-166. [PMID: 40228591 DOI: 10.1016/j.semcancer.2025.04.001] [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: 08/09/2024] [Revised: 03/13/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
Cathepsins, a group of lysosomal peptidases, have traditionally been recognized as tumor facilitators. Recent research, however, highlights their critical role in orchestrating cancer and the tumor microenvironment (TME). Primality, cathepsins degrade extracellular matrix, enabling cancer cells to invade and metastasize, while also promoting vascular endothelial infiltration and subsequent angiogenesis. Additionally, cathepsins boost fibroblast growth, thereby supporting tumor progression. More importantly, cathepsins are pivotal in modulating immune cells within the TME by regulating their recruitment, antigen processing and presentation, differentiation, and cell death, primarily contributing to immune suppression. Given their overexpression in tumors and elevated levels in the circulation of cancer patients, it is crucial to consider the systemic effects of cathepsins. Although the comprehensive role of cathepsins in cancer patients' bodies remains underexplored, they likely influence systemic immunity and inflammation, cellular metabolism, muscle wasting, and distant metastasis through their unique proteolytic functions. Notably, cathepsins also confer resistance to chemoradiotherapy by rewriting the cellular profile within the TME. In this context, promising results are emerging from studies combining cathepsin inhibitors with conventional therapies to suppress tumor development effectively. This review aims to decipher the cathepsin-driven networks within cancer cells and the TME, detailing their contribution to chemoradioresistance by reshaping both micro- and macroenvironments. Furthermore, we explore current and future perspectives on therapies targeting cathepsins' interactions, offering insights into innovative treatment strategies.
Collapse
Affiliation(s)
- Yuki Fujii
- Department of Oncology Science, University of Oklahoma Health Sciences, Oklahoma City, OK 73014, USA
| | - Zahra Asadi
- Department of Oncology Science, University of Oklahoma Health Sciences, Oklahoma City, OK 73014, USA; Department of Pathology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA
| | - Kamiya Mehla
- Department of Oncology Science, University of Oklahoma Health Sciences, Oklahoma City, OK 73014, USA; Department of Microbiology and Immunology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA; Department of Pathology, University of Oklahoma Health Sciences, Oklahoma City, OK 73104, USA.
| |
Collapse
|
2
|
Giri A, Hong IS, Kwon TK, Kang JS, Jeong JH, Kweon S, Yook S. Exploring therapeutic and diagnostic potential of cysteine cathepsin as targets for cancer therapy with nanomedicine. Int J Biol Macromol 2025; 315:144324. [PMID: 40398760 DOI: 10.1016/j.ijbiomac.2025.144324] [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: 03/30/2025] [Revised: 05/05/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025]
Abstract
Cysteine cathepsins have been discovered to be substantially expressed in multiple types of cancer. They play a key role in the progression and growth of these cancers, rendering them appealing targets for nanoscale delivery and noninvasive diagnostic imaging. This review explores cathepsins from the papain-like enzyme family (C1) within the cysteine peptidase clan (CA), emphasizing the role of cathepsin-responsive nanoparticles in tumor growth. Furthermore, it also explores how nanotechnology can harness cathepsin activity to enable targeted drug delivery, improve tumor imaging, and reduce systemic toxicity. By examining the molecular mechanisms governing cathepsin function and evaluating different nanocarrier systems, this work aims to enhance our understanding of targeted cancer treatment. Despite significant advances, challenges remain in translating these nanomedicine platforms into clinical use, including improving delivery efficiency, biocompatibility, long-term safety, and addressing issues such as interspecies protease variability and scalable nanomanufacturing. Future advancement, integrating advanced biomaterials, patient-derived organoid models, bispecific immune-protease targeting, CRISPR-based cathepsin editing, and artificial intelligence-driven pharmacokinetic modeling and analysis will be critical to fully realizing the clinical potential of cathepsin targeted nanomedicines. These innovations hold promises for advancing precision oncology by overcoming current limitations and improving patient outcomes.
Collapse
Affiliation(s)
- Anil Giri
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - In-Sun Hong
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Republic of Korea; Center for Forensic Pharmaceutical Science, Keimyung University, Daegu, 42601, Republic of Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, Gyeonggi 16419, Republic of Korea
| | - Seho Kweon
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Simmyung Yook
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
3
|
Eweje F, Ibrahim V, Shajii A, Walsh ML, Ahmad K, Alrefai A, Miyasato D, Davis JR, Ham H, Li K, Roehrl M, Haller CA, Liu DR, Chen J, Chaikof EL. Self-assembling protein nanoparticles for cytosolic delivery of nucleic acids and proteins. Nat Biotechnol 2025:10.1038/s41587-025-02664-2. [PMID: 40374955 DOI: 10.1038/s41587-025-02664-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 04/01/2025] [Indexed: 05/18/2025]
Abstract
Intracellular delivery of biomacromolecules is hampered by low efficiency and cytotoxicity. Here we report the development of elastin-based nanoparticles for therapeutic delivery (ENTER), a recombinant elastin-like polypeptide (ELP)-based delivery system for effective cytosolic delivery of biomacromolecules in vitro and in vivo. Through iterative design, we developed fourth-generation ELPs fused to cationic endosomal escape peptides (EEPs) that self-assemble into pH-responsive micellar nanoparticles and enable cytosolic entry of cargo following endocytic uptake. In silico screening of α-helical peptide libraries led to the discovery of an EEP (EEP13) with 48% improved protein delivery efficiency versus a benchmark peptide. Our lead ELP-EEP13 showed similar or superior performance compared to lipid-based transfection reagents in the delivery of mRNA-encoded, DNA-encoded and protein-form Cre recombinase and CRISPR gene editors as well as short interfering RNAs to multiple cell lines and primary cell types. Intranasal administration of ELP-EEP13 combined with Cre protein achieved efficient editing of lung epithelial cells in reporter mice.
Collapse
Affiliation(s)
- Feyisayo Eweje
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard/MIT MD-PhD Program, Boston, MA, USA
- Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Vanessa Ibrahim
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Aram Shajii
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Michelle L Walsh
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard/MIT MD-PhD Program, Boston, MA, USA
| | - Kiran Ahmad
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Assma Alrefai
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Dominie Miyasato
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jessie R Davis
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hyunok Ham
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Kaicheng Li
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Michael Roehrl
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Carolyn A Haller
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - David R Liu
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jiaxuan Chen
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| | - Elliot L Chaikof
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| |
Collapse
|
4
|
Katagiri D, Nagasaka S, Takahashi K, Wang S, Pozzi A, Zent R, Shimizu A, Zhang MZ, Göthert JR, van Kuppevelt TH, Harris RC, Takahashi T. Endothelial eNOS deficiency causes podocyte injury through NFAT2 and heparanase in diabetic mice. Sci Rep 2024; 14:29179. [PMID: 39587144 PMCID: PMC11589149 DOI: 10.1038/s41598-024-79501-0] [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: 02/02/2024] [Accepted: 11/11/2024] [Indexed: 11/27/2024] Open
Abstract
The pivotal role of endothelial nitric oxide synthase (eNOS) in diabetic nephropathy (DN) has been demonstrated using global eNOS knockout (eNOSGKO) mice. However, the precise role of endothelially expressed eNOS and how its deficiency advances DN are still unclear. Here, we targeted endothelial eNOS expression (E-eNOSKO) after the onset of diabetes using the floxed eNOS and endSCL-CreERT alleles. Diabetes was induced by low-dose streptozotocin injections. To evaluate the role of nuclear factor of activated T cells-2 (NFAT2) in podocyte injury in this condition, podocyte-specific NFAT2KO mice were also generated on eNOSGKO mice. The mechanisms of podocyte injury were investigated using cultured podocytes. Compared with diabetic wild-type mice, diabetic E-eNOSKO mice showed more advanced DN accompanied by NFAT2 expression in podocytes. NO donor suppressed NFAT2 expression and activation in high-glucose cultured podocytes as well as in diabetic E-eNOSKO mice. Furthermore, podocyte-specific deletion of NFAT2 attenuated DN in diabetic eNOSGKO mice accompanied by decreased heparanase (HPSE) expression in podocytes. Consistent with this finding, HPSE was upregulated by NFAT2 transfection and suppressed by NFAT2 siRNA or NO donor treatment in cultured podocytes. HPSE transfection reduced podocyte attachment to extracellular matrix concurrent with syndecan-4 (SDC4) shedding, and this effect was attenuated by co-transfection of SDC4. Finally, HPSE inhibitor treatment attenuated podocyte injury in diabetic E-eNOSKO mice with increased SDC4 expression in podocytes. Collectively, our data suggest that endothelial eNOS deficiency causes podocyte HPSE expression in diabetic mice through NFAT2, and HPSE promotes podocyte detachment in part through SDC4 shedding, advancing DN.
Collapse
Affiliation(s)
- Daisuke Katagiri
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223, MCN, Nashville, TN, 37232, USA
| | - Shinya Nagasaka
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223, MCN, Nashville, TN, 37232, USA
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Keiko Takahashi
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223, MCN, Nashville, TN, 37232, USA
| | - Suwan Wang
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223, MCN, Nashville, TN, 37232, USA
| | - Ambra Pozzi
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223, MCN, Nashville, TN, 37232, USA
- Department of Veterans Affairs, Nashville, TN, USA
| | - Roy Zent
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223, MCN, Nashville, TN, 37232, USA
- Department of Veterans Affairs, Nashville, TN, USA
| | - Akira Shimizu
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Ming-Zhi Zhang
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223, MCN, Nashville, TN, 37232, USA
| | - Joachim R Göthert
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Toin H van Kuppevelt
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Raymond C Harris
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223, MCN, Nashville, TN, 37232, USA
- Department of Veterans Affairs, Nashville, TN, USA
| | - Takamune Takahashi
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223, MCN, Nashville, TN, 37232, USA.
| |
Collapse
|
5
|
Załęcka J, Zielińska Z, Ołdak Ł, Sakowicz A, Mańka G, Kiecka M, Spaczyński R, Piekarski P, Banaszewska B, Jakimiuk A, Issat T, Młodawski J, Szubert M, Sieroszewski P, Raba G, Szczupak K, Kluz T, Kluza M, Pierzyński P, Ciebiera M, Wojtyła C, Lipa M, Warzecha D, Wielgoś M, Cendrowski K, Gorodkiewicz E, Laudański P. The SPRi determination of cathepsin L and S in plasma and peritoneal fluid of women with endometriosis. Adv Med Sci 2024; 69:224-230. [PMID: 38642611 DOI: 10.1016/j.advms.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/26/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
PURPOSE Endometriosis is a common disease with a complex pathomechanism and atypical symptoms, often leading to delayed diagnosis. Currently, the sole method for confirming the presence of the disease is through laparoscopy and histopathological examination of collected tissue. However, this invasive procedure carries potential risk and complications, necessitating the exploration of non-surgical diagnostic methods for endometriosis. This study aims to analyze peritoneal fluid and plasma samples for the expression of cathepsin L and cathepsin S to identify potential biomarkers for non-invasive diagnostic approaches to endometriosis. MATERIAL AND METHODS In this cross-sectional study, plasma and peritoneal fluid samples were obtained during laparoscopy from 63 patients diagnosed with chronic pelvic pain or infertility. The study group consisted of women with confirmed endometriosis. The concentrations of cathepsins L and S were determined using an SPRi biosensor. RESULTS The study did not reveal significant differences in the concentrations of cathepsin L and cathepsin S between the control group and the study group, both in peritoneal fluid and plasma. CONCLUSIONS Based on the results of this study, it appears that cathepsins L and S are not suitable candidates as biomarkers for endometriosis.
Collapse
Affiliation(s)
- Julia Załęcka
- Chair and Department of Obstetrics, Gynecology and Gynecological Oncology, Medical University of Warsaw, Poland
| | - Zuzanna Zielińska
- Bioanalysis Laboratory, Doctoral School of Exact and Natural Science, Faculty of Chemistry, University of Bialystok, Ciolkowskiego 1K, Bialystok, Poland
| | - Łukasz Ołdak
- Bioanalysis Laboratory, Faculty of Chemistry, University of Bialystok, Ciolkowskiego 1K, Bialystok, Poland
| | - Agata Sakowicz
- Department of Medical Biotechnology, Medical University of Lodz, Lodz, Poland
| | | | | | - Robert Spaczyński
- Center for Gynecology, Obstetrics and Infertility Treatment Pastelova, Poznan, Poland; Collegium Medicum, Institute of Health Sciences, University of Zielona Gora, Poland
| | - Piotr Piekarski
- Gynecological Obstetric Clinical Hospital of Poznan University of Medical Sciences, Minimally Invasive Gynecological Surgery, Poznan, Poland
| | - Beata Banaszewska
- Chair and Department of Laboratory Diagnostics Poznan University of Medical Sciences, Poland
| | - Artur Jakimiuk
- Department of Reproductive Health, Institute of Mother and Child in Warsaw, Warsaw, Poland; Department of Gynecology, Oncological Gynecology and Reproduction, National Medical Institute of the Ministry of the Interior and Administration, Warsaw, Poland
| | - Tadeusz Issat
- Department of Obstetrics and Gynecology, Institute of Mother and Child in Warsaw, Warsaw, Poland
| | - Jakub Młodawski
- Collegium Medicum Jan Kochanowski University in Kielce, Poland; Clinic of Obstetrics and Gynecology, Provincial Combined Hospital in Kielce, Kielce, Poland
| | - Maria Szubert
- Department of Gynecology and Obstetrics, Medical University of Lodz, Lodz, Poland; Department of Surgical Gynecology and Oncology, Medical University of Lodz, Lodz, Poland
| | - Piotr Sieroszewski
- Department of Gynecology and Obstetrics, Medical University of Lodz, Lodz, Poland; Department of Fetal Medicine and Gynecology, Medical University of Lodz, Lodz, Poland
| | - Grzegorz Raba
- Clinic of Obstetrics and Gynecology in Przemysl, Przemysl, Poland; Department of Obstetrics and Gynecology, University of Rzeszow, Rzeszow, Poland
| | - Kamil Szczupak
- Clinic of Obstetrics and Gynecology in Przemysl, Przemysl, Poland; Department of Obstetrics and Gynecology, University of Rzeszow, Rzeszow, Poland
| | - Tomasz Kluz
- Department of Gynecology, Gynecology Oncology and Obstetrics, Institute of Medical Sciences, Medical College of Rzeszow University, Rzeszow, Poland
| | - Marek Kluza
- Department of Gynecology, Gynecology Oncology and Obstetrics, Institute of Medical Sciences, Medical College of Rzeszow University, Rzeszow, Poland
| | | | - Michał Ciebiera
- Second Department of Obstetrics and Gynecology, Centre of Postrgraduate Medical Education, Warsaw, Poland; Warsaw Institute of Women's Health, Warsaw, Poland
| | - Cezary Wojtyła
- OVIklinika Infertility Center, Warsaw, Poland; Women's Health Research Institute, Calisia University, Kalisz, Poland
| | - Michał Lipa
- Department of Obstetrics and Perinatology, National Medical Institute of the Ministry of the Interior and Administration, Warsaw, Poland
| | - Damian Warzecha
- OVIklinika Infertility Center, Warsaw, Poland; City South Hospital Warsaw, Warsaw, Poland; Faculty of Medicine, University of Warsaw, Warsaw, Poland
| | - Mirosław Wielgoś
- Department of Obstetrics and Perinatology, National Medical Institute of the Ministry of the Interior and Administration, Warsaw, Poland; Premium Medical Clinic, Warsaw, Poland; Medical Faculty, Lazarski University, Warsaw, Poland
| | - Krzysztof Cendrowski
- Chair and Department of Obstetrics, Gynecology and Gynecological Oncology, Medical University of Warsaw, Poland
| | - Ewa Gorodkiewicz
- Bioanalysis Laboratory, Faculty of Chemistry, University of Bialystok, Ciolkowskiego 1K, Bialystok, Poland
| | - Piotr Laudański
- Chair and Department of Obstetrics, Gynecology and Gynecological Oncology, Medical University of Warsaw, Poland; OVIklinika Infertility Center, Warsaw, Poland; Women's Health Research Institute, Calisia University, Kalisz, Poland.
| |
Collapse
|
6
|
Hamo-Giladi DB, Fokra A, Sabo E, Kabala A, Minkov I, Hamoud S, Hadad S, Abassi Z, Khamaysi I. Involvement of heparanase in the pathogenesis of acute pancreatitis: Implication of novel therapeutic approaches. J Cell Mol Med 2024; 28:e18512. [PMID: 39248454 PMCID: PMC11382361 DOI: 10.1111/jcmm.18512] [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: 11/02/2023] [Revised: 06/07/2024] [Accepted: 06/18/2024] [Indexed: 09/10/2024] Open
Abstract
Acute pancreatitis (AP) is a common gastrointestinal disease with high morbidity and mortality rate. Unfortunately, neither the etiology nor the pathophysiology of AP are fully understood and causal treatment options are not available. Recently we demonstrated that heparanase (Hpa) is adversely involved in the pathogenesis of AP and inhibition of this enzyme ameliorates the manifestation of the disease. Moreover, a pioneer study demonstrated that Aspirin has partial inhibitory effect on Hpa. Another compound, which possesses a mild pancreato-protective effect against AP, is Trehalose, a common disaccharide. We hypothesized that combination of Aspirin, Trehalose, PG545 (Pixatimod) and SST0001 (Roneparstat), specific inhibitors of Hpa, may exert pancreato-protective effect better than each drug alone. Thus, the current study examines the pancreato-protective effects of Aspirin, Trehalose, PG545 and SST0001 in experimental model of AP induced by cerulein in wild-type (WT) and Hpa over-expressing (Hpa-Tg) mice. Cerulein-induced AP in WT mice was associated with significant rises in the serum levels of lipase (X4) and amylase (X3) with enhancement of pancreatic edema index, inflammatory response, and autophagy. Responses to cerulein were all more profound in Hpa-Tg mice versus WT mice, evident by X7 and X5 folds increase in lipase and amylase levels, respectively. Treatment with Aspirin or Trehalose alone and even more so in combination with PG545 or SST0001 were highly effective, restoring the serum level of lipase back to the basal level. Importantly, a novel newly synthesized compound termed Aspirlose effectively ameliorated the pathogenesis of AP as a single agent. Collectively, the results strongly indicate that targeting Hpa by using anti-Hpa drug combinations constitute a novel therapy for this common orphan disease.
Collapse
Affiliation(s)
- Dalit B Hamo-Giladi
- Department of Physiology, The Ruth & Bruce Rappaport Faculty of Medicine, Haifa, Israel
| | - Ahmad Fokra
- Department of Physiology, The Ruth & Bruce Rappaport Faculty of Medicine, Haifa, Israel
| | - Edmond Sabo
- Department of Pathology, Carmel Hospital, Haifa, Israel
| | - Aviva Kabala
- Department of Physiology, The Ruth & Bruce Rappaport Faculty of Medicine, Haifa, Israel
| | - Irena Minkov
- Department of Pathology, Rambam Health Care Center, Haifa, Israel
| | - Shadi Hamoud
- Department of Internal Medicine E, Rambam Health Care Center, Haifa, Israel
| | - Salim Hadad
- Department of Pharmacy, Rambam Health Care Center, Haifa, Israel
| | - Zaid Abassi
- Department of Physiology, The Ruth & Bruce Rappaport Faculty of Medicine, Haifa, Israel
- Department of Laboratory Medicine, Rambam Health Care Center, Haifa, Israel
| | - Iyad Khamaysi
- Department of Gastroenterology, Rambam Health Care Center, Haifa, Israel
| |
Collapse
|
7
|
Shi J, Onuki Y, Kawanami F, Miyagawa N, Iwasaki F, Tsuda H, Takahashi K, Oku T, Suzuki M, Higashi K, Adachi H, Nishimura Y, Nakajima M, Irimura T, Higashi N. The Uptake of Heparanase into Mast Cells Is Regulated by Its Enzymatic Activity to Degrade Heparan Sulfate. Int J Mol Sci 2024; 25:6281. [PMID: 38892469 PMCID: PMC11173065 DOI: 10.3390/ijms25116281] [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: 05/08/2024] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024] Open
Abstract
Mast cells take up extracellular latent heparanase and store it in secretory granules. The present study examined whether the enzymatic activity of heparanase regulates its uptake efficiency. Recombinant mouse heparanase mimicking both the latent and mature forms (L-Hpse and M-Hpse, respectively) was internalized into mastocytoma MST cells, peritoneal cell-derived mast cells, and bone marrow-derived mast cells. The internalized amount of L-Hpse was significantly higher than that of M-Hpse. In MST cells, L-Hpse was continuously internalized for up to 8 h, while the uptake of M-Hpse was saturated after 2 h of incubation. L-Hpse and M-Hpse are similarly bound to the MST cell surface. The expression level of cell surface heparan sulfate was reduced in MST cells incubated with M-Hpse. The internalized amount of M-Hpse into mast cells was significantly increased in the presence of heparastatin (SF4), a small molecule heparanase inhibitor that does not affect the binding of heparanase to immobilized heparin. Enzymatically quiescent M-Hpse was prepared with a point mutation at Glu335. The internalized amount of mutated M-Hpse was significantly higher than that of wild-type M-Hpse but similar to that of wild-type and mutated L-Hpse. These results suggest that the enzymatic activity of heparanase negatively regulates the mast cell-mediated uptake of heparanase, possibly via the downregulation of cell surface heparan sulfate expression.
Collapse
Affiliation(s)
- Jia Shi
- Department of Biochemistry, Hoshi University School of Pharmacy, 2-4-41, Ebara, Shinagawa-ku 142-8501, Tokyo, Japan; (J.S.); (Y.O.); (H.T.); (K.T.)
| | - Yoshiki Onuki
- Department of Biochemistry, Hoshi University School of Pharmacy, 2-4-41, Ebara, Shinagawa-ku 142-8501, Tokyo, Japan; (J.S.); (Y.O.); (H.T.); (K.T.)
| | - Fumiya Kawanami
- Department of Biochemistry, Hoshi University School of Pharmacy, 2-4-41, Ebara, Shinagawa-ku 142-8501, Tokyo, Japan; (J.S.); (Y.O.); (H.T.); (K.T.)
| | - Naoko Miyagawa
- Department of Biochemistry, Hoshi University School of Pharmacy, 2-4-41, Ebara, Shinagawa-ku 142-8501, Tokyo, Japan; (J.S.); (Y.O.); (H.T.); (K.T.)
| | - Fumika Iwasaki
- Department of Biochemistry, Hoshi University School of Pharmacy, 2-4-41, Ebara, Shinagawa-ku 142-8501, Tokyo, Japan; (J.S.); (Y.O.); (H.T.); (K.T.)
| | - Haruna Tsuda
- Department of Biochemistry, Hoshi University School of Pharmacy, 2-4-41, Ebara, Shinagawa-ku 142-8501, Tokyo, Japan; (J.S.); (Y.O.); (H.T.); (K.T.)
| | - Katsuhiko Takahashi
- Department of Biochemistry, Hoshi University School of Pharmacy, 2-4-41, Ebara, Shinagawa-ku 142-8501, Tokyo, Japan; (J.S.); (Y.O.); (H.T.); (K.T.)
| | - Teruaki Oku
- Department of Microbiology, Hoshi University School of Pharmacy, 2-4-41, Ebara, Shinagawa-ku 142-8501, Tokyo, Japan;
| | - Masato Suzuki
- Department of Clinical and Analytical Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641, Yamazaki, Noda 278-8510, Chiba, Japan (K.H.)
| | - Kyohei Higashi
- Department of Clinical and Analytical Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641, Yamazaki, Noda 278-8510, Chiba, Japan (K.H.)
| | - Hayamitsu Adachi
- Institute of Microbial Chemistry (BIKAKEN), 18-24, Miyamoto, Numazu 410-0301, Shizuoka, Japan;
| | - Yoshio Nishimura
- Institute of Microbial Chemistry (BIKAKEN), 3-14-23, Kamiosaki, Shinagawa-ku 141-0021, Tokyo, Japan;
| | - Motowo Nakajima
- SBI Pharmaceuticals Co., Ltd., 1-6-1, Roppongi, Minato-ku 106-6019, Tokyo, Japan;
| | - Tatsuro Irimura
- Division of Glycobiologics, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku 113-8421, Tokyo, Japan;
| | - Nobuaki Higashi
- Department of Biochemistry, Hoshi University School of Pharmacy, 2-4-41, Ebara, Shinagawa-ku 142-8501, Tokyo, Japan; (J.S.); (Y.O.); (H.T.); (K.T.)
| |
Collapse
|
8
|
Radisky ES. Extracellular proteolysis in cancer: Proteases, substrates, and mechanisms in tumor progression and metastasis. J Biol Chem 2024; 300:107347. [PMID: 38718867 PMCID: PMC11170211 DOI: 10.1016/j.jbc.2024.107347] [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: 02/09/2024] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 06/02/2024] Open
Abstract
A vast ensemble of extracellular proteins influences the development and progression of cancer, shaped and reshaped by a complex network of extracellular proteases. These proteases, belonging to the distinct classes of metalloproteases, serine proteases, cysteine proteases, and aspartic proteases, play a critical role in cancer. They often become dysregulated in cancer, with increases in pathological protease activity frequently driven by the loss of normal latency controls, diminished regulation by endogenous protease inhibitors, and changes in localization. Dysregulated proteases accelerate tumor progression and metastasis by degrading protein barriers within the extracellular matrix (ECM), stimulating tumor growth, reactivating dormant tumor cells, facilitating tumor cell escape from immune surveillance, and shifting stromal cells toward cancer-promoting behaviors through the precise proteolysis of specific substrates to alter their functions. These crucial substrates include ECM proteins and proteoglycans, soluble proteins secreted by tumor and stromal cells, and extracellular domains of cell surface proteins, including membrane receptors and adhesion proteins. The complexity of the extracellular protease web presents a significant challenge to untangle. Nevertheless, technological strides in proteomics, chemical biology, and the development of new probes and reagents are enabling progress and advancing our understanding of the pivotal importance of extracellular proteolysis in cancer.
Collapse
Affiliation(s)
- Evette S Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida, USA.
| |
Collapse
|
9
|
Rabinowitz ZM, Somers J, Wang Z, Cui L. Chemical toolbox to interrogate Heparanase-1 activity. Curr Opin Chem Biol 2024; 80:102452. [PMID: 38555836 DOI: 10.1016/j.cbpa.2024.102452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 04/02/2024]
Abstract
The development of a robust chemical toolbox to interrogate the activity of heparanase-1 (HPSE-1), an endo-β-d-glucuronidase and the only known enzyme that cleaves heparan sulfate (HS), has become critically important. The primary function of HPSE-1, cleaving HS side chains from heparan sulfate proteoglycans (HSPGs), regulates the integrity of the extracellular matrix (ECM) and the bioavailability of active, heparan sulfate-binding partners such as enzymes, growth factors, chemokines, and cytokines. HPSE-1 enzymatic activity is strictly regulated and has been found to play fundamental roles in pathophysiological processes. HPSE-1 is significantly overexpressed under various conditions including cancer, metastasis, angiogenesis, and inflammation, making HPSE-1 a promising therapeutic and diagnostic target. Chemical tools that can detect and image HPSE-1 activity in vitro and/or in vivo can help drive the discovery of novel and efficacious anti-HPSE-1 drugs, investigate the basic biology of HPSE-1, and help serve as a diagnostic tool in clinical applications. Here, we will give an overview of the common chemical tools to detect HPSE-1 activity and highlight the novel heparanase probes recently developed in our lab.
Collapse
Affiliation(s)
- Zachary M Rabinowitz
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Johnathan Somers
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Zhishen Wang
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Lina Cui
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
10
|
Rabinowitz ZM, Wang Z, Liu J, Zhang Y, Ybargollin AJ, Saketkhou M, Cui L. A Fluorogenic Green Merocyanine-Based Probe to Detect Heparanase-1 Activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.25.581963. [PMID: 38464176 PMCID: PMC10925095 DOI: 10.1101/2024.02.25.581963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Heparanase-1 (HPSE-1), an endo-β-D-glucuronidase, is an extracellular matrix (ECM) remodeling enzyme that degrades heparan sulfate (HS) chains of heparan sulfate proteoglycans (HSPGs). HPSE-1 functions to remodel the ECM and thereby disseminate cells, liberate HS-bound bioactive molecules, and release biologically active HS fragments. Being the only known enzyme for the cleavage of HS, HPSE-1 regulates a number of fundamental cellular processes including cell migration, cytokine regulation, angiogenesis, and wound healing. Overexpression of HPSE-1 has been discovered in most cancers, inflammatory diseases, viral infections, among others. As an emerging therapeutic target, the biological role of HPSE-1 remains to be explored but is hampered by a lack of research tools. To expand the chemical tool-kit of fluorogenic probes to interrogate HPSE-1 activity, we design and synthesized a fluorogenic green disaccharide-based HPSE-1 probe using our design strategy of tuning the electronic effect of the aryl aglycon. The novel probe exhibits a highly sensitive 278-fold fluorescence turn-on response in the presence of recombinant human HPSE-1, while emitting green light at 560 nm, enabling the fluorescence imaging of HPSE-1 activity in cells.
Collapse
|
11
|
Imai Y, Suzuki R, Matsuda D, Tanaka-Yamamoto N, Ohki Y, Tabata R, Kato S, Sugisaki M, Fujimoto N, Fukunaga T, Kato S, Takahashi T, Kakinuma H. Discovery of a novel tetrahydroimidazo[1,2-a]pyridine-5-carboxylic acid derivative as a potent and selective heparanase-1 inhibitor utilizing an improved synthetic approach. Bioorg Med Chem Lett 2024; 97:129543. [PMID: 37939863 DOI: 10.1016/j.bmcl.2023.129543] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/25/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
Heparanase-1 (HPSE1) is an endo-β-d-glucuronidase that catalyzes degradation of heparan sulfate proteoglycans. Inhibition of HPSE1 appears to be a useful therapeutic target against cancer and proteinuric kidney diseases. We previously reported tetrahydroimidazo[1,2-a]pyridine 2 as a potent HPSE1 inhibitor after optimization of the synthetic reaction. However, synthesis of 2 involves a total of 19 steps, including a cyclization process that accompanies a strong odor due to the use of Lawesson's reagent and an epimerization reaction; furthermore, 2 exhibited insufficient selectivity for HPSE1 over exo-β-d-glucuronidase (GUSβ) and glucocerebrosidase (GBA), which also needed to be addressed. First, the cyclization reaction was optimized to synthesize tetrahydroimidazo[1,2-a]pyridine without using Lawesson's reagent or epimerization, with reference to previous reports. Next, 16 and 17 containing a bulkier substituent at position 6 than the 6-methoxyl group in 2 were designed and synthesized using the improved cyclization conditions, so that the synthetic route of 16 and 17 was shortened by five steps as compared with that of 2. The inhibitory activities of 16 and 17 against GUSβ and GBA were reduced as compared with those of 2, that is, the compounds showed improved selectivity for HPSE1 over GUSβ and GBA. In addition, 16 showed enhanced inhibitory activity against HPSE1 as compared with that of 2. Compound 16 appears promising as an HPSE1 inhibitor with therapeutic potential due to its highly potent inhibitory activity against HPSE1 with high selectivity for HPSE1.
Collapse
Affiliation(s)
- Yudai Imai
- Medicinal Chemistry Laboratories, Taisho Pharmaceutical Co, Ltd, 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Ryo Suzuki
- Medicinal Chemistry Laboratories, Taisho Pharmaceutical Co, Ltd, 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Daisuke Matsuda
- Medicinal Chemistry Laboratories, Taisho Pharmaceutical Co, Ltd, 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Nozomi Tanaka-Yamamoto
- Medicinal Chemistry Laboratories, Taisho Pharmaceutical Co, Ltd, 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Yuta Ohki
- Medicinal Chemistry Laboratories, Taisho Pharmaceutical Co, Ltd, 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Ryotaro Tabata
- Pharmacology Laboratories, Taisho Pharmaceutical Co Ltd, 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Sota Kato
- Pharmacology Laboratories, Taisho Pharmaceutical Co Ltd, 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Mami Sugisaki
- Pharmacology Laboratories, Taisho Pharmaceutical Co Ltd, 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Natsuko Fujimoto
- Discovery Technologies Laboratories, Taisho Pharmaceutical Co Ltd, 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Takuya Fukunaga
- Discovery Technologies Laboratories, Taisho Pharmaceutical Co Ltd, 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Sayaka Kato
- Discovery Technologies Laboratories, Taisho Pharmaceutical Co Ltd, 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Teisuke Takahashi
- Pharmacology Laboratories, Taisho Pharmaceutical Co Ltd, 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Hiroyuki Kakinuma
- Medicinal Chemistry Laboratories, Taisho Pharmaceutical Co, Ltd, 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan.
| |
Collapse
|
12
|
Imai Y, Suzuki R, Wakasugi D, Matsuda D, Tanaka-Yamamoto N, Ohki Y, Mima M, Endo M, Tabata R, Matsuzawa H, Hasegawa Y, Kato S, Sugisaki M, Miyagawa H, Fujimoto N, Fukunaga T, Kato S, Takahashi T, Kakinuma H. Structure-based lead optimization to improve potency and selectivity of a novel tetrahydroimidazo[1,2-a]pyridine-5-carboxylic acid series of heparanase-1 inhibitor. Bioorg Med Chem 2023; 93:117460. [PMID: 37660465 DOI: 10.1016/j.bmc.2023.117460] [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: 07/04/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/05/2023]
Abstract
Heparanase-1 (HPSE1) is an endo-β-d-glucuronidase that is the only mammalian enzyme known to cleave heparan sulfate (HS) of heparan sulfate proteoglycans (HSPG), a key component of the glycocalyx layer of the vascular endothelium matrix. Inhibition of HPSE1 has therapeutic potential for cancer and proteinuric kidney diseases. We previously reported that 2 showed a moderate potency as an HPSE1 inhibitor and an issue of selectivity against exo-β-d-glucuronidase (GUSβ) and glucocerebrosidase (GBA) remained. A structure-based lead optimization of 2 using X-ray co-crystal structure analysis and fragment molecular orbital calculation resulted in 4e, which showed a more than 7-fold increase in HPSE1 inhibitory activity. The subsequent introduction of a methyl group into the 6-hydroxy group of 4e resulted in 18 with reduced inhibitory activities against GUSβ and GBA while maintaining the inhibitory activity against HPSE1. The inhibitory activities of 18 against serum HPSE1 in mice were significant and lasted for 4 h at doses of 3, 30, and 100 mg/kg. Compound 18 could be a novel lead compound for HPSE1 inhibitors with improved inhibitory activity against HPSE1 and increased HPSE1 selectivity over GUSβ and GBA.
Collapse
Affiliation(s)
- Yudai Imai
- Medicinal Chemistry Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Ryo Suzuki
- Medicinal Chemistry Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Daisuke Wakasugi
- Medicinal Chemistry Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Daisuke Matsuda
- Medicinal Chemistry Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Nozomi Tanaka-Yamamoto
- Medicinal Chemistry Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Yuta Ohki
- Medicinal Chemistry Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Masashi Mima
- Discovery Technologies Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Mayumi Endo
- Discovery Technologies Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Ryotaro Tabata
- Pharmacology Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Hitomi Matsuzawa
- Drug Safety and Pharmacokinetics Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Yoshitaka Hasegawa
- Drug Safety and Pharmacokinetics Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Sota Kato
- Pharmacology Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Mami Sugisaki
- Pharmacology Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Hiroh Miyagawa
- Discovery Technologies Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Natsuko Fujimoto
- Discovery Technologies Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Takuya Fukunaga
- Discovery Technologies Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Sayaka Kato
- Discovery Technologies Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Teisuke Takahashi
- Pharmacology Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan
| | - Hiroyuki Kakinuma
- Medicinal Chemistry Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama 331-9530, Japan.
| |
Collapse
|
13
|
Potokar M, Zorec R, Jorgačevski J. Astrocytes Are a Key Target for Neurotropic Viral Infection. Cells 2023; 12:2307. [PMID: 37759529 PMCID: PMC10528686 DOI: 10.3390/cells12182307] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 08/28/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Astrocytes are increasingly recognized as important viral host cells in the central nervous system. These cells can produce relatively high quantities of new virions. In part, this can be attributed to the characteristics of astrocyte metabolism and its abundant and dynamic cytoskeleton network. Astrocytes are anatomically localized adjacent to interfaces between blood capillaries and brain parenchyma and between blood capillaries and brain ventricles. Moreover, astrocytes exhibit a larger membrane interface with the extracellular space than neurons. These properties, together with the expression of various and numerous viral entry receptors, a relatively high rate of endocytosis, and morphological plasticity of intracellular organelles, render astrocytes important target cells in neurotropic infections. In this review, we describe factors that mediate the high susceptibility of astrocytes to viral infection and replication, including the anatomic localization of astrocytes, morphology, expression of viral entry receptors, and various forms of autophagy.
Collapse
Affiliation(s)
- Maja Potokar
- Laboratory of Neuroendocrinology–Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
- Celica Biomedical, Tehnološki Park 24, 1000 Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Neuroendocrinology–Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
- Celica Biomedical, Tehnološki Park 24, 1000 Ljubljana, Slovenia
| | - Jernej Jorgačevski
- Laboratory of Neuroendocrinology–Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
- Celica Biomedical, Tehnološki Park 24, 1000 Ljubljana, Slovenia
| |
Collapse
|
14
|
Manganelli V, Misasi R, Riitano G, Capozzi A, Mattei V, Caglar TR, Ialongo D, Madia VN, Messore A, Costi R, Di Santo R, Sorice M, Garofalo T. Role of a Novel Heparanase Inhibitor on the Balance between Apoptosis and Autophagy in U87 Human Glioblastoma Cells. Cells 2023; 12:1891. [PMID: 37508554 PMCID: PMC10378526 DOI: 10.3390/cells12141891] [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: 06/21/2023] [Revised: 07/07/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Heparanase (HPSE) is an endo-β-glucuronidase that cleaves heparan sulfate side chains, leading to the disassembly of the extracellular matrix, facilitating cell invasion and metastasis dissemination. In this research, we investigated the role of a new HPSE inhibitor, RDS 3337, in the regulation of the autophagic process and the balance between apoptosis and autophagy in U87 glioblastoma cells. METHODS After treatment with RDS 3337, cell lysates were analyzed for autophagy and apoptosis-related proteins by Western blot. RESULTS We observed, firstly, that LC3II expression increased in U87 cells incubated with RDS 3337, together with a significant increase of p62/SQSTM1 levels, indicating that RDS 3337 could act through the inhibition of autophagic-lysosomal flux of LC3-II, thereby leading to accumulation of lipidated LC3-II form. Conversely, the suppression of autophagic flux could activate apoptosis mechanisms, as revealed by the activation of caspase 3, the increased level of cleaved Parp1, and DNA fragmentation. CONCLUSIONS These findings support the notion that HPSE promotes autophagy, providing evidence that RDS 3337 blocks autophagic flux. It indicates a role for HPSE inhibitors in the balance between apoptosis and autophagy in U87 human glioblastoma cells, suggesting a potential role for this new class of compounds in the control of tumor growth progression.
Collapse
Affiliation(s)
- Valeria Manganelli
- Department of Experimental Medicine, "Sapienza" University, 00161 Rome, Italy
| | - Roberta Misasi
- Department of Experimental Medicine, "Sapienza" University, 00161 Rome, Italy
| | - Gloria Riitano
- Department of Experimental Medicine, "Sapienza" University, 00161 Rome, Italy
| | - Antonella Capozzi
- Department of Experimental Medicine, "Sapienza" University, 00161 Rome, Italy
| | - Vincenzo Mattei
- Biomedicine and Advanced Technologies Rieti Center, Sabina Universitas, 02100 Rieti, Italy
| | - Tuba Rana Caglar
- Department of Experimental Medicine, "Sapienza" University, 00161 Rome, Italy
| | - Davide Ialongo
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, "Sapienza" University of Rome, 00185 Rome, Italy
| | - Valentina Noemi Madia
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, "Sapienza" University of Rome, 00185 Rome, Italy
| | - Antonella Messore
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, "Sapienza" University of Rome, 00185 Rome, Italy
| | - Roberta Costi
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, "Sapienza" University of Rome, 00185 Rome, Italy
| | - Roberto Di Santo
- Dipartimento di Chimica e Tecnologie del Farmaco, Istituto Pasteur-Fondazione Cenci Bolognetti, "Sapienza" University of Rome, 00185 Rome, Italy
| | - Maurizio Sorice
- Department of Experimental Medicine, "Sapienza" University, 00161 Rome, Italy
| | - Tina Garofalo
- Department of Experimental Medicine, "Sapienza" University, 00161 Rome, Italy
| |
Collapse
|
15
|
Vlodavsky I, Kayal Y, Hilwi M, Soboh S, Sanderson RD, Ilan N. Heparanase-A single protein with multiple enzymatic and nonenzymatic functions. PROTEOGLYCAN RESEARCH 2023; 1:e6. [PMID: 37547889 PMCID: PMC10398610 DOI: 10.1002/pgr2.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 08/08/2023]
Abstract
Heparanase (Hpa1) is expressed by tumor cells and cells of the tumor microenvironment and functions extracellularly to remodel the extracellular matrix (ECM) and regulate the bioavailability of ECM-bound factors, augmenting, among other effects, gene transcription, autophagy, exosome formation, and heparan sulfate (HS) turnover. Much of the impact of heparanase on tumor progression is related to its function in mediating tumor-host crosstalk, priming the tumor microenvironment to better support tumor growth, metastasis, and chemoresistance. The enzyme appears to fulfill some normal functions associated, for example, with vesicular traffic, lysosomal-based secretion, autophagy, HS turnover, and gene transcription. It activates cells of the innate immune system, promotes the formation of exosomes and autophagosomes, and stimulates signal transduction pathways via enzymatic and nonenzymatic activities. These effects dynamically impact multiple regulatory pathways that together drive tumor growth, dissemination, and drug resistance as well as inflammatory responses. The emerging premise is that heparanase expressed by tumor cells, immune cells, endothelial cells, and other cells of the tumor microenvironment is a key regulator of the aggressive phenotype of cancer, an important contributor to the poor outcome of cancer patients and a valid target for therapy. So far, however, antiheparanase-based therapy has not been implemented in the clinic. Unlike heparanase, heparanase-2 (Hpa2), a close homolog of heparanase (Hpa1), does not undergo proteolytic processing and hence lacks intrinsic HS-degrading activity, the hallmark of heparanase. Hpa2 retains the capacity to bind heparin/HS and exhibits an even higher affinity towards HS than heparanase, thus competing for HS binding and inhibiting heparanase enzymatic activity. It appears that Hpa2 functions as a natural inhibitor of Hpa1 regulates the expression of selected genes that maintain tissue hemostasis and normal function, and plays a protective role against cancer and inflammation, together emphasizing the significance of maintaining a proper balance between Hpa1 and Hpa2.
Collapse
Affiliation(s)
- Israel Vlodavsky
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| | - Yasmin Kayal
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| | - Maram Hilwi
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| | - Soaad Soboh
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| | - Ralph D. Sanderson
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Neta Ilan
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| |
Collapse
|
16
|
The Interplay of Glycosaminoglycans and Cysteine Cathepsins in Mucopolysaccharidosis. Biomedicines 2023; 11:biomedicines11030810. [PMID: 36979788 PMCID: PMC10045161 DOI: 10.3390/biomedicines11030810] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/27/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Mucopolysaccharidosis (MPS) consists of a group of inherited lysosomal storage disorders that are caused by a defect of certain enzymes that participate in the metabolism of glycosaminoglycans (GAGs). The abnormal accumulation of GAGs leads to progressive dysfunctions in various tissues and organs during childhood, contributing to premature death. As the current therapies are limited and inefficient, exploring the molecular mechanisms of the pathology is thus required to address the unmet needs of MPS patients to improve their quality of life. Lysosomal cysteine cathepsins are a family of proteases that play key roles in numerous physiological processes. Dysregulation of cysteine cathepsins expression and activity can be frequently observed in many human diseases, including MPS. This review summarizes the basic knowledge on MPS disorders and their current management and focuses on GAGs and cysteine cathepsins expression in MPS, as well their interplay, which may lead to the development of MPS-associated disorders.
Collapse
|
17
|
Lee CS, Zhai Y, Shang R, Wong T, Mattison AJ, Cen HH, Johnson JD, Vlodavsky I, Hussein B, Rodrigues B. Flow-Induced Secretion of Endothelial Heparanase Regulates Cardiac Lipoprotein Lipase and Changes Following Diabetes. J Am Heart Assoc 2022; 11:e027958. [PMID: 36416172 PMCID: PMC9851453 DOI: 10.1161/jaha.122.027958] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Background Lipoprotein lipase (LPL)-derived fatty acid is a major source of energy for cardiac contraction. Synthesized in cardiomyocytes, LPL requires translocation to the vascular lumen for hydrolysis of lipoprotein triglyceride, an action mediated by endothelial cell (EC) release of heparanase. We determined whether flow-mediated biophysical forces can cause ECs to secrete heparanase and thus regulate cardiac metabolism. Methods and Results Isolated hearts were retrogradely perfused. Confluent rat aortic ECs were exposed to laminar flow using an orbital shaker. Cathepsin L activity was determined using gelatin-zymography. Diabetes was induced in rats with streptozotocin. Despite the abundance of enzymatically active heparanase in the heart, it was the enzymatically inactive, latent heparanase that was exceptionally responsive to flow-induced release. EC exposed to orbital rotation exhibited a similar pattern of heparanase secretion, an effect that was reproduced by activation of the mechanosensor, Piezo1. The laminar flow-mediated release of heparanase from EC required activation of both the purinergic receptor and protein kinase D, a kinase that assists in vesicular transport of proteins. Heparanase influenced cardiac metabolism by increasing cardiomyocyte LPL displacement along with subsequent replenishment. The flow-induced heparanase secretion was augmented following diabetes and could explain the increased heparin-releasable pool of LPL at the coronary lumen in these diabetic hearts. Conclusions ECs sense fluid shear-stress and communicate this information to subjacent cardiomyocytes with the help of heparanase. This flow-induced mechanosensing and its dynamic control of cardiac metabolism to generate ATP, using LPL-derived fatty acid, is exquisitely adapted to respond to disease conditions, like diabetes.
Collapse
Affiliation(s)
- Chae Syng Lee
- Faculty of Pharmaceutical SciencesUBCVancouverBritish ColumbiaCanada
| | - Yajie Zhai
- Faculty of Pharmaceutical SciencesUBCVancouverBritish ColumbiaCanada
| | - Rui Shang
- Faculty of Pharmaceutical SciencesUBCVancouverBritish ColumbiaCanada
| | - Trevor Wong
- Faculty of Pharmaceutical SciencesUBCVancouverBritish ColumbiaCanada
| | - Aurora J. Mattison
- Department of Cellular and Physiological Sciences & Department of SurgeryDiabetes Focus Team, Life Sciences Institute, UBCVancouverBritish ColumbiaCanada
| | - Haoning Howard Cen
- Department of Cellular and Physiological Sciences & Department of SurgeryDiabetes Focus Team, Life Sciences Institute, UBCVancouverBritish ColumbiaCanada
| | - James D. Johnson
- Department of Cellular and Physiological Sciences & Department of SurgeryDiabetes Focus Team, Life Sciences Institute, UBCVancouverBritish ColumbiaCanada
| | - Israel Vlodavsky
- Cancer and Vascular Biology Research CenterRappaport Faculty of Medicine, TechnionHaifaIsrael
| | - Bahira Hussein
- Faculty of Pharmaceutical SciencesUBCVancouverBritish ColumbiaCanada
| | - Brian Rodrigues
- Faculty of Pharmaceutical SciencesUBCVancouverBritish ColumbiaCanada
| |
Collapse
|
18
|
Heparanase: A Novel Therapeutic Target for the Treatment of Atherosclerosis. Cells 2022; 11:cells11203198. [PMID: 36291066 PMCID: PMC9599978 DOI: 10.3390/cells11203198] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/07/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death and disability worldwide, and its management places a huge burden on healthcare systems through hospitalisation and treatment. Atherosclerosis is a chronic inflammatory disease of the arterial wall resulting in the formation of lipid-rich, fibrotic plaques under the subendothelium and is a key contributor to the development of CVD. As such, a detailed understanding of the mechanisms involved in the development of atherosclerosis is urgently required for more effective disease treatment and prevention strategies. Heparanase is the only mammalian enzyme known to cleave heparan sulfate of heparan sulfate proteoglycans, which is a key component of the extracellular matrix and basement membrane. By cleaving heparan sulfate, heparanase contributes to the regulation of numerous physiological and pathological processes such as wound healing, inflammation, tumour angiogenesis, and cell migration. Recent evidence suggests a multifactorial role for heparanase in atherosclerosis by promoting underlying inflammatory processes giving rise to plaque formation, as well as regulating lesion stability. This review provides an up-to-date overview of the role of heparanase in physiological and pathological processes with a focus on the emerging role of the enzyme in atherosclerosis.
Collapse
|
19
|
Takahashi I. Importance of Heparan Sulfate Proteoglycans in Pancreatic Islets and β-Cells. Int J Mol Sci 2022; 23:12082. [PMID: 36292936 PMCID: PMC9603760 DOI: 10.3390/ijms232012082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 09/20/2022] [Indexed: 12/15/2022] Open
Abstract
β-cells in the islets of Langerhans of the pancreas secrete insulin in response to the glucose concentration in the blood. When these pancreatic β-cells are damaged, diabetes develops through glucose intolerance caused by insufficient insulin secretion. High molecular weight polysaccharides, such as heparin and heparan sulfate (HS) proteoglycans, and HS-degrading enzymes, such as heparinase, participate in the protection, maintenance, and enhancement of the functions of pancreatic islets and β-cells, and the demand for studies on glycobiology within the field of diabetes research has increased. This review introduces the roles of complex glycoconjugates containing high molecular weight polysaccharides and their degrading enzymes in pancreatic islets and β-cells, including those obtained in studies conducted by us earlier. In addition, from the perspective of glycobiology, this study proposes the possibility of application to diabetes medicine.
Collapse
Affiliation(s)
- Iwao Takahashi
- Division of Molecular and Cellular Pharmacology, Department of Pathophysiology and Pharmacology, School of Pharmacy, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Morioka 028-3694, Iwate, Japan
| |
Collapse
|
20
|
Denamur S, Chazeirat T, Maszota-Zieleniak M, Vivès RR, Saidi A, Zhang F, Linhardt RJ, Labarthe F, Samsonov SA, Lalmanach G, Lecaille F. Binding of heparan sulfate to human cystatin C modulates inhibition of cathepsin L: Putative consequences in mucopolysaccharidosis. Carbohydr Polym 2022; 293:119734. [DOI: 10.1016/j.carbpol.2022.119734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/30/2022] [Accepted: 06/11/2022] [Indexed: 11/02/2022]
|
21
|
The Key Role of Lysosomal Protease Cathepsins in Viral Infections. Int J Mol Sci 2022; 23:ijms23169089. [PMID: 36012353 PMCID: PMC9409221 DOI: 10.3390/ijms23169089] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Cathepsins encompass a family of lysosomal proteases that mediate protein degradation and turnover. Although mainly localized in the endolysosomal compartment, cathepsins are also found in the cytoplasm, nucleus, and extracellular space, where they are involved in cell signaling, extracellular matrix assembly/disassembly, and protein processing and trafficking through the plasma and nuclear membrane and between intracellular organelles. Ubiquitously expressed in the body, cathepsins play regulatory roles in a wide range of physiological processes including coagulation, hormone secretion, immune responses, and others. A dysregulation of cathepsin expression and/or activity has been associated with many human diseases, including cancer, diabetes, obesity, cardiovascular and inflammatory diseases, kidney dysfunctions, and neurodegenerative disorders, as well as infectious diseases. In viral infections, cathepsins may promote (1) activation of the viral attachment glycoproteins and entry of the virus into target cells; (2) antigen processing and presentation, enabling the virus to replicate in infected cells; (3) up-regulation and processing of heparanase that facilitates the release of viral progeny and the spread of infection; and (4) activation of cell death that may either favor viral clearance or assist viral propagation. In this review, we report the most relevant findings on the molecular mechanisms underlying cathepsin involvement in viral infection physiopathology, and we discuss the potential of cathepsin inhibitors for therapeutical applications in viral infectious diseases.
Collapse
|
22
|
Kong L, Ji H, Gan X, Cao S, Li Z, Jin Y. Knockdown of CD44 inhibits proliferation, migration and invasion of osteosarcoma cells accompanied by downregulation of cathepsin S. J Orthop Surg Res 2022; 17:154. [PMID: 35264209 PMCID: PMC8905747 DOI: 10.1186/s13018-022-03048-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/02/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Osteosarcoma (OS) is a malignant bone tumour of mesenchymal origin. These tumours are characterised by rich vascularisation, therefore promoting rapid proliferation and facilitating metastasis. CD44 has been reported to be involved in OS, but its role and molecular mechanisms in the pathogenesis of the disease are not fully determined. METHODS In this study, we investigated the antitumor effect of CD44 on the development of OS and further explored the molecular mechanisms. The expression of CD44, cathepsin S and MMP-9 was detected by Western blot (WB) and reverse transcription-polymerase chain reaction (RT-qPCR) in different cell lines (MG63, U2OS OS and hFOB 1.19). To elucidate the role of CD44 in OS, MG63 and U2OS cells were treated with small interference RNA (siRNA) to knock down CD44, and the knockdown efficiency was validated with GFP and RT-qPCR. Furthermore, cell proliferation was assayed using Cell Counting Kit‑8 (CCK-8) and colony formation assays, and cell migration and invasion were assayed by transwell and wound-healing assays. RESULTS We found that CD44 expression in the MG63 and U2OS OS cell lines was markedly increased compared to that of the human osteoblast hFOB 1.19 cell line. Knockdown of CD44 inhibited proliferation, migration and invasion of MG63 and U2OS cells. Cathepsin S expression in the MG63 and U2OS OS cell lines was increased compared to that of the human osteoblast hFOB 1.19 cell line. When CD44 was knocked down, its expression level went down. CONCLUSION Taken together, our data reinforced the evidence that CD44 knockdown inhibited cell proliferation, migration and invasion of OS cells accompanied by altered expression of cathepsin S. These findings offer new clues for OS development and progression, suggesting CD44 as a potential therapeutic target for OS.
Collapse
Affiliation(s)
- Lingwei Kong
- Department of Orthopaedics, The Affiliated Hospital of Chengde Medical College, No. 1 Nanyingzi Street, Chengde, 067000, Hebei, China
| | - Hairu Ji
- Pathology Teaching and Research Section, Chengde Medical College, Chengde, 067000, Hebei, China
| | - Xintian Gan
- Department of Orthopaedics, The Affiliated Hospital of Chengde Medical College, No. 1 Nanyingzi Street, Chengde, 067000, Hebei, China
| | - Sheng Cao
- Department of Orthopaedics, The Affiliated Hospital of Chengde Medical College, No. 1 Nanyingzi Street, Chengde, 067000, Hebei, China
| | - Zhehong Li
- Department of Orthopaedics, The Affiliated Hospital of Chengde Medical College, No. 1 Nanyingzi Street, Chengde, 067000, Hebei, China
| | - Yu Jin
- Department of Orthopaedics, The Affiliated Hospital of Chengde Medical College, No. 1 Nanyingzi Street, Chengde, 067000, Hebei, China.
| |
Collapse
|
23
|
Gureeva TA, Timoshenko OS, Kugaevskaya EV, Solovyova NI. [Cysteine cathepsins: structure, physiological functions and their role in carcinogenesis]. BIOMEDITSINSKAIA KHIMIIA 2021; 67:453-464. [PMID: 34964439 DOI: 10.18097/pbmc20216706453] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Cysteine cathepsins (Cts) also known as thiol proteinases belong to the superfamily of cysteine proteinases (EC 3.4.22). Cts are known as lysosomal proteases responsible for the intracellular proteins degradation. All Cts are synthesized as zymogens, activation of which occurs autocatalytically. Their activity is regulated by endogenous inhibitors. Cts can be secreted into the extracellular environment, which is of particular importance in tumor progression. Extracellular Cts not only hydrolyze extracellular matrix (ECM) proteins, but also contribute to ECM remodeling, processing and/or release of cell adhesion molecules, growth factors, cytokines and chemokines. In cancer, the expression and activity of Cts sharply increase both in cell lysosomes and in the intercellular space, which correlates with neoplastic transformation, invasion, metastasis and leads to further tumor progression. It has been shown that Cts expression depends on the cells type, therefore, their role in the tumor development differs depending on their cellular origin. The mechanism of Cts action in cancer is not limited only by their proteolytic action. The Cts influence on signal transduction pathways associated with cancer development, including the pathway involving growth factors, which is mediated through receptors tyrosine kinases (RTK) and various signaling mitogen-activated protein kinases (MAPK), has been proven. In addition, Cts are able to promote the epithelial-mesenchymal transition (EMT) by activating signal transduction pathways such as Wnt, Notch, and the pathway involving TGF-β. So, Ctc perform specific both destructive and regulatory functions, carrying out proteolysis, both inside and outside the cell.
Collapse
Affiliation(s)
- T A Gureeva
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | | | | |
Collapse
|
24
|
Mayfosh AJ, Nguyen TK, Hulett MD. The Heparanase Regulatory Network in Health and Disease. Int J Mol Sci 2021; 22:11096. [PMID: 34681753 PMCID: PMC8541136 DOI: 10.3390/ijms222011096] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/07/2021] [Accepted: 10/11/2021] [Indexed: 12/24/2022] Open
Abstract
The extracellular matrix (ECM) is a structural framework that has many important physiological functions which include maintaining tissue structure and integrity, serving as a barrier to invading pathogens, and acting as a reservoir for bioactive molecules. This cellular scaffold is made up of various types of macromolecules including heparan sulfate proteoglycans (HSPGs). HSPGs comprise a protein core linked to the complex glycosaminoglycan heparan sulfate (HS), the remodeling of which is important for many physiological processes such as wound healing as well as pathological processes including cancer metastasis. Turnover of HS is tightly regulated by a single enzyme capable of cleaving HS side chains: heparanase. Heparanase upregulation has been identified in many inflammatory diseases including atherosclerosis, fibrosis, and cancer, where it has been shown to play multiple roles in processes such as epithelial-mesenchymal transition, angiogenesis, and cancer metastasis. Heparanase expression and activity are tightly regulated. Understanding the regulation of heparanase and its downstream targets is attractive for the development of treatments for these diseases. This review provides a comprehensive overview of the regulators of heparanase as well as the enzyme's downstream gene and protein targets, and implications for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Alyce J. Mayfosh
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3083, Australia; (A.J.M.); (T.K.N.)
| | - Tien K. Nguyen
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3083, Australia; (A.J.M.); (T.K.N.)
| | - Mark D. Hulett
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3083, Australia; (A.J.M.); (T.K.N.)
| |
Collapse
|
25
|
Cousin R, Groult H, Manseur C, Ferru-Clément R, Gani M, Havret R, Toucheteau C, Prunier G, Colin B, Morel F, Piot JM, Lanneluc I, Baranger K, Maugard T, Fruitier-Arnaudin I. A Marine λ-Oligocarrageenan Inhibits Migratory and Invasive Ability of MDA-MB-231 Human Breast Cancer Cells through Actions on Heparanase Metabolism and MMP-14/MMP-2 Axis. Mar Drugs 2021; 19:md19100546. [PMID: 34677445 PMCID: PMC8539239 DOI: 10.3390/md19100546] [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: 07/21/2021] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 11/16/2022] Open
Abstract
Sugar-based molecules such as heparins or natural heparan sulfate polysaccharides have been developed and widely studied for controlling heparanase (HPSE) enzymatic activity, a key player in extracellular matrix remodelling during cancer pathogenesis. However, non-enzymatic functions of HPSE have also been described in tumour mechanisms. Given their versatile properties, we hypothesized that sugar-based inhibitors may interfere with enzymatic but also non-enzymatic HPSE activities. In this work, we assessed the effects of an original marine λ-carrageenan derived oligosaccharide (λ-CO) we previously described, along with those of its native counterpart and heparins, on cell viability, proliferation, migration, and invasion of MDA-MB-231 breast cancer cells but also of sh-MDA-MB-231 cells, in which the expression of HPSE was selectively downregulated. We observed no cytotoxic and no anti-proliferative effects of our compounds but surprisingly λ-CO was the most efficient to reduce cell migration and invasion compared with heparins, and in a HPSE-dependent manner. We provided evidence that λ-CO tightly controlled a HPSE/MMP-14/MMP-2 axis, leading to reduced MMP-2 activity. Altogether, this study highlights λ-CO as a potent HPSE “modulator” capable of reducing not only the enzymatic activity of HPSE but also the functions controlled by the HPSE levels.
Collapse
Affiliation(s)
- Rémi Cousin
- BCBS Group (Biotechnologies et Chimie des Bioressources pour la Santé), Laboratoire Littoral Environnement et Sociétés, La Rochelle University, UMR CNRS 7266, 17000 La Rochelle, France; (R.C.); (H.G.); (C.M.); (R.F.-C.); (M.G.); (R.H.); (C.T.); (G.P.); (B.C.); (J.-M.P.); (I.L.); (T.M.)
| | - Hugo Groult
- BCBS Group (Biotechnologies et Chimie des Bioressources pour la Santé), Laboratoire Littoral Environnement et Sociétés, La Rochelle University, UMR CNRS 7266, 17000 La Rochelle, France; (R.C.); (H.G.); (C.M.); (R.F.-C.); (M.G.); (R.H.); (C.T.); (G.P.); (B.C.); (J.-M.P.); (I.L.); (T.M.)
| | - Chanez Manseur
- BCBS Group (Biotechnologies et Chimie des Bioressources pour la Santé), Laboratoire Littoral Environnement et Sociétés, La Rochelle University, UMR CNRS 7266, 17000 La Rochelle, France; (R.C.); (H.G.); (C.M.); (R.F.-C.); (M.G.); (R.H.); (C.T.); (G.P.); (B.C.); (J.-M.P.); (I.L.); (T.M.)
| | - Romain Ferru-Clément
- BCBS Group (Biotechnologies et Chimie des Bioressources pour la Santé), Laboratoire Littoral Environnement et Sociétés, La Rochelle University, UMR CNRS 7266, 17000 La Rochelle, France; (R.C.); (H.G.); (C.M.); (R.F.-C.); (M.G.); (R.H.); (C.T.); (G.P.); (B.C.); (J.-M.P.); (I.L.); (T.M.)
| | - Mario Gani
- BCBS Group (Biotechnologies et Chimie des Bioressources pour la Santé), Laboratoire Littoral Environnement et Sociétés, La Rochelle University, UMR CNRS 7266, 17000 La Rochelle, France; (R.C.); (H.G.); (C.M.); (R.F.-C.); (M.G.); (R.H.); (C.T.); (G.P.); (B.C.); (J.-M.P.); (I.L.); (T.M.)
| | - Rachel Havret
- BCBS Group (Biotechnologies et Chimie des Bioressources pour la Santé), Laboratoire Littoral Environnement et Sociétés, La Rochelle University, UMR CNRS 7266, 17000 La Rochelle, France; (R.C.); (H.G.); (C.M.); (R.F.-C.); (M.G.); (R.H.); (C.T.); (G.P.); (B.C.); (J.-M.P.); (I.L.); (T.M.)
| | - Claire Toucheteau
- BCBS Group (Biotechnologies et Chimie des Bioressources pour la Santé), Laboratoire Littoral Environnement et Sociétés, La Rochelle University, UMR CNRS 7266, 17000 La Rochelle, France; (R.C.); (H.G.); (C.M.); (R.F.-C.); (M.G.); (R.H.); (C.T.); (G.P.); (B.C.); (J.-M.P.); (I.L.); (T.M.)
| | - Grégoire Prunier
- BCBS Group (Biotechnologies et Chimie des Bioressources pour la Santé), Laboratoire Littoral Environnement et Sociétés, La Rochelle University, UMR CNRS 7266, 17000 La Rochelle, France; (R.C.); (H.G.); (C.M.); (R.F.-C.); (M.G.); (R.H.); (C.T.); (G.P.); (B.C.); (J.-M.P.); (I.L.); (T.M.)
| | - Béatrice Colin
- BCBS Group (Biotechnologies et Chimie des Bioressources pour la Santé), Laboratoire Littoral Environnement et Sociétés, La Rochelle University, UMR CNRS 7266, 17000 La Rochelle, France; (R.C.); (H.G.); (C.M.); (R.F.-C.); (M.G.); (R.H.); (C.T.); (G.P.); (B.C.); (J.-M.P.); (I.L.); (T.M.)
| | - Franck Morel
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, Poitiers University, LITEC EA 4331, 86073 Poitiers, France;
| | - Jean-Marie Piot
- BCBS Group (Biotechnologies et Chimie des Bioressources pour la Santé), Laboratoire Littoral Environnement et Sociétés, La Rochelle University, UMR CNRS 7266, 17000 La Rochelle, France; (R.C.); (H.G.); (C.M.); (R.F.-C.); (M.G.); (R.H.); (C.T.); (G.P.); (B.C.); (J.-M.P.); (I.L.); (T.M.)
| | - Isabelle Lanneluc
- BCBS Group (Biotechnologies et Chimie des Bioressources pour la Santé), Laboratoire Littoral Environnement et Sociétés, La Rochelle University, UMR CNRS 7266, 17000 La Rochelle, France; (R.C.); (H.G.); (C.M.); (R.F.-C.); (M.G.); (R.H.); (C.T.); (G.P.); (B.C.); (J.-M.P.); (I.L.); (T.M.)
| | - Kévin Baranger
- Aix-Marseille University, CNRS, INP, Inst Neurophysiopathol, 13385 Marseille, France;
| | - Thierry Maugard
- BCBS Group (Biotechnologies et Chimie des Bioressources pour la Santé), Laboratoire Littoral Environnement et Sociétés, La Rochelle University, UMR CNRS 7266, 17000 La Rochelle, France; (R.C.); (H.G.); (C.M.); (R.F.-C.); (M.G.); (R.H.); (C.T.); (G.P.); (B.C.); (J.-M.P.); (I.L.); (T.M.)
| | - Ingrid Fruitier-Arnaudin
- BCBS Group (Biotechnologies et Chimie des Bioressources pour la Santé), Laboratoire Littoral Environnement et Sociétés, La Rochelle University, UMR CNRS 7266, 17000 La Rochelle, France; (R.C.); (H.G.); (C.M.); (R.F.-C.); (M.G.); (R.H.); (C.T.); (G.P.); (B.C.); (J.-M.P.); (I.L.); (T.M.)
- Correspondence: ; Tel.: +33-546-458-562
| |
Collapse
|
26
|
Perišić Nanut M, Pečar Fonović U, Jakoš T, Kos J. The Role of Cysteine Peptidases in Hematopoietic Stem Cell Differentiation and Modulation of Immune System Function. Front Immunol 2021; 12:680279. [PMID: 34335582 PMCID: PMC8322073 DOI: 10.3389/fimmu.2021.680279] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 07/01/2021] [Indexed: 01/21/2023] Open
Abstract
Cysteine cathepsins are primarily involved in the degradation and recycling of proteins in endo-lysosomal compartments but are also gaining recognition as pivotal proteolytic contributors to various immune functions. Through their extracellular proteolytic activities within the hematopoietic stem cell niche, they are involved in progenitor cell mobilization and differentiation. Cysteine cathepsins, such as cathepsins L and S contribute to antigen-induced adaptive immunity through major histocompatibility complex class II antigen presentation whereas cathepsin X regulates T-cell migration. By regulating toll-like receptor signaling and cytokine secretion cysteine cathepsins activate innate immune cells and affect their functional differentiation. Cathepsins C and H are expressed in cytotoxic T lymphocytes and natural killer cells and are involved in processing of pro-granzymes into proteolytically active forms. Cytoplasmic activities of cathepsins B and L contribute to the maintenance of homeostasis of the adaptive immune response by regulating cell death of T and B lymphocytes. The expression pattern, localization, and activity of cysteine cathepsins is tightly connected to their function in immune cells. Furthermore, cysteine cathepsins together with their endogenous inhibitors, serve as mediators in the interplay between cancer and immune cells that results in immune cell anergy. The aim of the present article is to review the mechanisms of dysregulation of cysteine cathepsins and their inhibitors in relation to immune dysfunction to address new possibilities for regulation of their function.
Collapse
Affiliation(s)
| | | | - Tanja Jakoš
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Janko Kos
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia.,Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
27
|
Eustes AS, Campbell RA, Middleton EA, Tolley ND, Manne BK, Montenont E, Rowley JW, Krauel K, Blair A, Guo L, Kosaka Y, Medeiros-de-Moraes IM, Lacerda M, Hottz ED, Neto HCF, Zimmerman GA, Weyrich AS, Petrey A, Rondina MT. Heparanase expression and activity are increased in platelets during clinical sepsis. J Thromb Haemost 2021; 19:1319-1330. [PMID: 33587773 PMCID: PMC8218538 DOI: 10.1111/jth.15266] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/22/2021] [Accepted: 02/08/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Heparanase (HPSE) is the only known mammalian enzyme that can degrade heparan sulfate. Heparan sulfate proteoglycans are essential components of the glycocalyx, and maintain physiological barriers between the blood and endothelial cells. HPSE increases during sepsis, which contributes to injurious glyocalyx degradation, loss of endothelial barrier function, and mortality. OBJECTIVES As platelets are one of the most abundant cellular sources of HPSE, we sought to determine whether HPSE expression and activity increases in human platelets during clinical sepsis. We also examined associations between platelet HPSE expression and clinical outcomes. PATIENTS/METHODS Expression and activity of HPSE was determined in platelets isolated from septic patients (n = 59) and, for comparison, sex-matched healthy donors (n = 46) using complementary transcriptomic, proteomic, and functional enzymatic assays. Septic patients were followed for the primary outcome of mortality, and clinical data were captured prospectively for septic patients. RESULTS The mRNA expression of HPSE was significantly increased in platelets isolated from septic patients. Ribosomal footprint profiling, followed by [S35] methionine labeling assays, demonstrated that HPSE mRNA translation and HPSE protein synthesis were significantly upregulated in platelets during sepsis. While both the pro- and active forms of HPSE protein increased in platelets during sepsis, only the active form of HPSE protein significantly correlated with sepsis-associated mortality. Consistent with transcriptomic and proteomic upregulation, HPSE enzymatic activity was also increased in platelets during sepsis. CONCLUSIONS During clinical sepsis HPSE, translation, and enzymatic activity are increased in platelets. Increased expression of the active form of HPSE protein is associated with sepsis-associated mortality.
Collapse
Affiliation(s)
- Alicia S Eustes
- Department of Internal Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
- Hospitals and Clinics Pathology, Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Robert A Campbell
- Department of Internal Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
| | - Elizabeth A Middleton
- Department of Internal Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
| | - Neal D Tolley
- Department of Internal Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
| | - Bhanu K Manne
- Department of Internal Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
| | - Emilie Montenont
- Department of Internal Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
| | - Jesse W Rowley
- Department of Internal Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
| | - Krystin Krauel
- Department of Internal Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Freiburg, Germany
| | - Antoinette Blair
- Department of Internal Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
| | - Li Guo
- Department of Internal Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
| | - Yasuhiro Kosaka
- Department of Internal Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
| | - Isabel M Medeiros-de-Moraes
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz, Rio de Janeiro and Manaus, Brazil
| | - Marcus Lacerda
- Fundacao de Medicina Tropical - Dr. Heitor Vieira Dourado (FMT-HVD) and Fiocruz Manaus, Manaus, Brazil
| | - Eugenio D Hottz
- Department of Internal Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz, Rio de Janeiro and Manaus, Brazil
- Immunothrombosis Laboratory, Department of Biochemistry, Federal University of Juiz de Fora, Juiz de Fora, Brazil
- Instituto Nacional de Infectologia Evandro Chagas, Rio de Janeiro, Brazil
| | - Hugo Castro Faria Neto
- Fundacao de Medicina Tropical - Dr. Heitor Vieira Dourado (FMT-HVD) and Fiocruz Manaus, Manaus, Brazil
| | - Guy A Zimmerman
- Department of Internal Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
| | - Andrew S Weyrich
- Department of Internal Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Aaron Petrey
- Department of Internal Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Matthew T Rondina
- Department of Internal Medicine and Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
- Department of Internal Medicine and GRECC, George E. Wahlen VAMC, Salt Lake City, Utah, USA
| |
Collapse
|
28
|
Melo CM, Nader HB, Justo GZ, Pinhal MAS. Heparanase modulation by Wingless/INT (Wnt). Mol Biol Rep 2021; 48:3117-3125. [PMID: 33891270 DOI: 10.1007/s11033-021-06348-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/08/2021] [Indexed: 11/28/2022]
Abstract
Heparanase is an endo-beta-glucuronidase, the only enzyme in mammals capable of cleaving heparan sulfate/heparin chains from proteoglycans. The oligosaccharides generated by heparanase present extensive biological functions since such oligosaccharides interact with adhesion molecules, growth factors, angiogenic factors and cytokines, modulating cell proliferation, migration, inflammation, and carcinogenesis. However, the regulation of heparanase activity is not fully understood. It is known that heparanase is synthesized as an inactive 65 kDa isoform and that post-translation processing forms an active 50 kDa enzyme. In the present study, we are interested in investigating whether heparanase is regulated by its own substrate as observed with many other enzymes. Wild-type Chinese hamster (Cricetulus griséus) ovary cells (CHO-K1) were treated with different doses of heparin. Heparanase expression was analyzed by Real-time PCR and flow cytometry. Also, heparanase activity was measured. The heparanase activity assay was performed using a coated plate with biotinylated heparan sulfate. In the present assay, a competitive heparin inhibition scenario was set aside. Exogenous heparin trigged a cell signaling pathway that increased heparanase mRNA and protein levels. The Wnt/beta-catenin pathway, judged by TCF-driven luciferase activity, seems to be involved to enhance heparanase profile during treatment with exogenous heparin. Lithium chloride treatment, an activator of the Wnt/beta-catenin pathway, confirmed such mechanism of transduction in vivo using zebrafish embryos and in vitro using CHO-K1 cells. Taken together the results suggest that heparin modulates heparanase expression by Wnt/beta-catenin.
Collapse
Affiliation(s)
- Carina Mucciolo Melo
- Department of Biochemistry, Universidade Federal de São Paulo, Rua Três de Maio, 100, 4a. andar, Biologia Molecular, São Paulo, SP, 04044-020, Brazil.,Department of Biochemistry, Faculdade de Medicina do ABC, Avenida Príncipe de Gales, 821, Bioquímica, Santo André, SP, 09060-650, Brazil
| | - Helena Bonciani Nader
- Department of Biochemistry, Universidade Federal de São Paulo, Rua Três de Maio, 100, 4a. andar, Biologia Molecular, São Paulo, SP, 04044-020, Brazil
| | - Giselle Zenker Justo
- Department of Biochemistry, Universidade Federal de São Paulo, Rua Três de Maio, 100, 4a. andar, Biologia Molecular, São Paulo, SP, 04044-020, Brazil.,Department of Biochemistry, Universidade Federal de São Paulo, Rua Prof. Artur Riedel, no. 275 - Jd. Eldorado, Diadema, SP, CEP: 09972-270, Brazil
| | - Maria Aparecida Silva Pinhal
- Department of Biochemistry, Universidade Federal de São Paulo, Rua Três de Maio, 100, 4a. andar, Biologia Molecular, São Paulo, SP, 04044-020, Brazil. .,Department of Biochemistry, Faculdade de Medicina do ABC, Avenida Príncipe de Gales, 821, Bioquímica, Santo André, SP, 09060-650, Brazil.
| |
Collapse
|
29
|
Schleyer KA, Fetrow B, Zannes Fatland P, Liu J, Chaaban M, Ma B, Cui L. Dual-Mechanism Quenched Fluorogenic Probe Provides Selective and Rapid Detection of Cathepsin L Activity*. ChemMedChem 2021; 16:1082-1087. [PMID: 33295147 PMCID: PMC8202353 DOI: 10.1002/cmdc.202000823] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Indexed: 12/18/2022]
Abstract
Cathepsin L (CTL) is a cysteine protease demonstrating upregulated activity in many disease states. Overlapping substrate specificity makes selective detection of CTL activity difficult to parse from that of its close homologue CTV and the ubiquitous CTB. Current probes of CTL activity have limited applications due to either poor contrast or extra assay steps required to achieve selectivity. We have developed a fluorogenic probe, CTLAP, that displays good selectivity for CTL over CTB and CTV while exhibiting low background fluorescence attributed to dual quenching mechanisms. CTLAP achieves optimum CTL selectivity in the first 10 min of incubation, thus suggesting that it is amenable for rapid detection of CTL, even in the presence of competing cathepsins.
Collapse
Affiliation(s)
- Kelton A Schleyer
- Department of Medicinal Chemistry, UF Health Science Center, UF Health Cancer Center, University of Florida, 1345 Center Dr., Gainesville, FL 32610, USA
- Department of Chemistry and Chemical Biology, UNM Comprehensive Cancer Center, University of New Mexico, 300 Terrace St. NE, Albuquerque, NM 87131, USA
| | - Ben Fetrow
- Department of Chemistry and Chemical Biology, UNM Comprehensive Cancer Center, University of New Mexico, 300 Terrace St. NE, Albuquerque, NM 87131, USA
| | - Peter Zannes Fatland
- Department of Chemistry and Chemical Biology, UNM Comprehensive Cancer Center, University of New Mexico, 300 Terrace St. NE, Albuquerque, NM 87131, USA
| | - Jun Liu
- Department of Medicinal Chemistry, UF Health Science Center, UF Health Cancer Center, University of Florida, 1345 Center Dr., Gainesville, FL 32610, USA
- Department of Chemistry and Chemical Biology, UNM Comprehensive Cancer Center, University of New Mexico, 300 Terrace St. NE, Albuquerque, NM 87131, USA
| | - Maya Chaaban
- Department of Chemistry and Biochemistry, Florida State University, 95 Chieftan Way 118 DLC, Tallahassee, FL 32306, USA
| | - Biwu Ma
- Department of Chemistry and Biochemistry, Florida State University, 95 Chieftan Way 118 DLC, Tallahassee, FL 32306, USA
| | - Lina Cui
- Department of Medicinal Chemistry, UF Health Science Center, UF Health Cancer Center, University of Florida, 1345 Center Dr., Gainesville, FL 32610, USA
- Department of Chemistry and Chemical Biology, UNM Comprehensive Cancer Center, University of New Mexico, 300 Terrace St. NE, Albuquerque, NM 87131, USA
| |
Collapse
|
30
|
Koganti R, Memon A, Shukla D. Emerging Roles of Heparan Sulfate Proteoglycans in Viral Pathogenesis. Semin Thromb Hemost 2021; 47:283-294. [PMID: 33851373 DOI: 10.1055/s-0041-1725068] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Heparan sulfate is a glycosaminoglycan present in nearly all mammalian tissues. Heparan sulfate moieties are attached to the cell surface via heparan sulfate proteoglycans (HSPGs) which are composed of a protein core bound to multiple heparan sulfate chains. HSPGs contribute to the structural integrity of the extracellular matrix and participate in cell signaling by releasing bound cytokines and chemokines once cleaved by an enzyme, heparanase. HSPGs are often exploited by viruses during infection, particularly during attachment and egress. Loss or inhibition of HSPGs initially during infection can yield significant decreases in viral entry and infectivity. In this review, we provide an overview of HSPGs in the lifecycle of multiple viruses, including herpesviruses, human immunodeficiency virus, dengue virus, human papillomavirus, and coronaviruses.
Collapse
Affiliation(s)
- Raghuram Koganti
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Abdullah Memon
- College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois.,Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
31
|
Vlodavsky I, Barash U, Nguyen HM, Yang SM, Ilan N. Biology of the Heparanase-Heparan Sulfate Axis and Its Role in Disease Pathogenesis. Semin Thromb Hemost 2021; 47:240-253. [PMID: 33794549 DOI: 10.1055/s-0041-1725066] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cell surface proteoglycans are important constituents of the glycocalyx and participate in cell-cell and cell-extracellular matrix (ECM) interactions, enzyme activation and inhibition, and multiple signaling routes, thereby regulating cell proliferation, survival, adhesion, migration, and differentiation. Heparanase, the sole mammalian heparan sulfate degrading endoglycosidase, acts as an "activator" of HS proteoglycans, thus regulating tissue hemostasis. Heparanase is a multifaceted enzyme that together with heparan sulfate, primarily syndecan-1, drives signal transduction, immune cell activation, exosome formation, autophagy, and gene transcription via enzymatic and nonenzymatic activities. An important feature is the ability of heparanase to stimulate syndecan-1 shedding, thereby impacting cell behavior both locally and distally from its cell of origin. Heparanase releases a myriad of HS-bound growth factors, cytokines, and chemokines that are sequestered by heparan sulfate in the glycocalyx and ECM. Collectively, the heparan sulfate-heparanase axis plays pivotal roles in creating a permissive environment for cell proliferation, differentiation, and function, often resulting in the pathogenesis of diseases such as cancer, inflammation, endotheliitis, kidney dysfunction, tissue fibrosis, and viral infection.
Collapse
Affiliation(s)
- Israel Vlodavsky
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Uri Barash
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Hien M Nguyen
- Department of Chemistry, Wayne State University, Detroit, Michigan
| | - Shi-Ming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Neta Ilan
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
32
|
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: 11.0] [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
|
33
|
Li B, Mu L, Li Y, Xia K, Yang Y, Aman S, Ahmad B, Li S, Wu H. TIMELESS inhibits breast cancer cell invasion and metastasis by down-regulating the expression of MMP9. Cancer Cell Int 2021; 21:38. [PMID: 33430865 PMCID: PMC7798230 DOI: 10.1186/s12935-021-01752-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 01/02/2021] [Indexed: 12/20/2022] Open
Abstract
Breast cancer is the first killer leading to female death, and tumor metastasis is one of the important factors leading to the death of patients, but the specific mechanism of breast cancer metastasis is not very clear at present. Our study showed that overexpression of TIMELESS could significantly inhibit the invasion and metastasis of breast cancer cells ZR-75-30 and the assembly of F-actin protein. On the contrary, knockdown of TIMELESS promoted the invasion and metastasis of breast cancer cells. Further study revealed that TIMELESS overexpression decreased the mRNA and protein levels of MMP9. Furthermore, TIMELESS could interact with p65, leading to repress the association of p65 and its acetyltransferase CBP and down-regulating the acetylation level of p65, which inhibited the activation of NF-κB signal pathway. In conclusion, our research showed that TIMELESS may repress the invasion and metastasis of breast cancer cells via inhibiting the acetylation of p65, inhibiting the activation of NF-κB, thus down-regulating the expression of MMP9, and then inhibiting the invasion and metastasis of breast cancer cells.
Collapse
Affiliation(s)
- Bowen Li
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, 2 Ling Gong Road, Dalian, 116024, Liaoning, China
| | - Liying Mu
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, 2 Ling Gong Road, Dalian, 116024, Liaoning, China
| | - Yanan Li
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, 2 Ling Gong Road, Dalian, 116024, Liaoning, China
| | - Kangkai Xia
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, 2 Ling Gong Road, Dalian, 116024, Liaoning, China
| | - Yuxi Yang
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, 2 Ling Gong Road, Dalian, 116024, Liaoning, China
| | - Sattout Aman
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, 2 Ling Gong Road, Dalian, 116024, Liaoning, China
| | - Bashir Ahmad
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, 2 Ling Gong Road, Dalian, 116024, Liaoning, China
| | - Shujing Li
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, 2 Ling Gong Road, Dalian, 116024, Liaoning, China.
| | - Huijian Wu
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Liaoning Province, Dalian University of Technology, 2 Ling Gong Road, Dalian, 116024, Liaoning, China.
| |
Collapse
|
34
|
Ulčakar L, Novinec M. Inhibition of Human Cathepsins B and L by Caffeic Acid and Its Derivatives. Biomolecules 2020; 11:E31. [PMID: 33383850 PMCID: PMC7824550 DOI: 10.3390/biom11010031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/18/2020] [Accepted: 12/25/2020] [Indexed: 12/15/2022] Open
Abstract
Caffeic acid (CA) and its derivatives caffeic acid phenethyl ester (CAPE) and chlorogenic acid (CGA) are phenolic compounds of plant origin with a wide range of biological activities. Here, we identify and characterize their inhibitory properties against human cathepsins B and L, potent, ubiquitously expressed cysteine peptidases involved in protein turnover and homeostasis, as well as pathological conditions, such as cancer. We show that CAPE and CGA inhibit both peptidases, while CA shows a preference for cathepsin B, resulting in the strongest inhibition among these combinations. All compounds are linear (complete) inhibitors acting via mixed or catalytic mechanisms. Cathepsin B is more strongly inhibited at pH 7.4 than at 5.5, and CA inhibits its endopeptidase activity preferentially over its peptidyl-dipeptidase activity. Altogether, the results identify the CA scaffold as a promising candidate for the development of cathepsin B inhibitors, specifically targeting its endopeptidase activity associated with pathological proteolysis of extracellular substrates.
Collapse
Affiliation(s)
| | - Marko Novinec
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, 1000 Ljubljana, Slovenia;
| |
Collapse
|
35
|
Koganti R, Suryawanshi R, Shukla D. Heparanase, cell signaling, and viral infections. Cell Mol Life Sci 2020; 77:5059-5077. [PMID: 32462405 PMCID: PMC7252873 DOI: 10.1007/s00018-020-03559-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/17/2020] [Accepted: 05/22/2020] [Indexed: 12/13/2022]
Abstract
Heparanase (HPSE) is a multifunctional protein endowed with many non-enzymatic functions and a unique enzymatic activity as an endo-β-D-glucuronidase. The latter allows it to serve as a key modulator of extracellular matrix (ECM) via a well-regulated cleavage of heparan sulfate side chains of proteoglycans at cell surfaces. The cleavage and associated changes at the ECM cause release of multiple signaling molecules with important cellular and pathological functions. New and emerging data suggest that both enzymatic as well as non-enzymatic functions of HPSE are important for health and illnesses including viral infections and virally induced cancers. This review summarizes recent findings on the roles of HPSE in activation, inhibition, or bioavailability of key signaling molecules such as AKT, VEGF, MAPK-ERK, and EGFR, which are known regulators of common viral infections in immune and non-immune cell types. Altogether, our review provides a unique overview of HPSE in cell-survival signaling pathways and how they relate to viral infections.
Collapse
Affiliation(s)
- Raghuram Koganti
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 W. Taylor St, Chicago, IL, 60612, USA
| | - Rahul Suryawanshi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 W. Taylor St, Chicago, IL, 60612, USA
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, 1855 W. Taylor St, Chicago, IL, 60612, USA.
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
36
|
Amin R, Tripathi K, Sanderson RD. Nuclear Heparanase Regulates Chromatin Remodeling, Gene Expression and PTEN Tumor Suppressor Function. Cells 2020; 9:cells9092038. [PMID: 32899927 PMCID: PMC7564302 DOI: 10.3390/cells9092038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 02/08/2023] Open
Abstract
Heparanase (HPSE) is an endoglycosidase that cleaves heparan sulfate and has been shown in various cancers to promote metastasis, angiogenesis, osteolysis, and chemoresistance. Although heparanase is thought to act predominantly extracellularly or within the cytoplasm, it is also present in the nucleus, where it may function in regulating gene transcription. Using myeloma cell lines, we report here that heparanase enhances chromatin accessibility and confirm a previous report that it also upregulates the acetylation of histones. Employing the Multiple Myeloma Research Foundation CoMMpass database, we demonstrate that patients expressing high levels of heparanase display elevated expression of proteins involved in chromatin remodeling and several oncogenic factors compared to patients expressing low levels of heparanase. These signatures were consistent with the known function of heparanase in driving tumor progression. Chromatin opening and downstream target genes were abrogated by inhibition of heparanase. Enhanced levels of heparanase in myeloma cells led to a dramatic increase in phosphorylation of PTEN, an event known to stabilize PTEN, leading to its inactivity and loss of tumor suppressor function. Collectively, this study demonstrates that heparanase promotes chromatin opening and transcriptional activity, some of which likely is through its impact on diminishing PTEN tumor suppressor activity.
Collapse
|
37
|
Iriyama S, Yasuda M, Nishikawa S, Takai E, Hosoi J, Amano S. Decrease of laminin-511 in the basement membrane due to photoaging reduces epidermal stem/progenitor cells. Sci Rep 2020; 10:12592. [PMID: 32724130 PMCID: PMC7387558 DOI: 10.1038/s41598-020-69558-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 07/14/2020] [Indexed: 02/08/2023] Open
Abstract
Daily sunlight exposure damages the epidermal basement membrane (BM) and disrupts epidermal homeostasis. Inter-follicular epidermal stem cells (IFE-SCs) regulate epidermal proliferation and differentiation, which supports epidermal homeostasis. Here, we examine how photoaging affects the function of IFE-SCs and we identify key components in their cellular environment (niche). We found that sun-exposed skin showed a decrease of MCSP-positive and β1-integrin-positive cells concomitantly with a decrease of laminin-511 at the dermal-epidermal junction (DEJ), as compared with sun-protected skin. Higher levels of laminin-511 were associated with not only increased efficiency of colony formation, but also higher expression levels of MCSP as well as other stem cell markers such as Lrig1, ITGB1, CD44, CD46, DLL1, and K15 in keratinocytes from skin of 12- to 62-year-old subjects. UVB exposure to cultured human skin impaired laminin-511 integrity at the dermal-epidermal junction and reduced MCSP-positive basal epidermal cells as well as K15-positive cells. Combined treatment with matrix metalloproteinase and heparanase inhibitors protected the integrity of laminin-511 and inhibited the reduction of MCSP-positive cells and K15-positive cells. These results suggest that photoaging may reduce the levels of MCSP-positive and K15-positive epidermal stem/progenitor cells in the epidermis via loss of laminin-511 at the dermal-epidermal junction.
Collapse
Affiliation(s)
- Shunsuke Iriyama
- Shiseido Global Innovation Center, 1-2-11 Takashima, Nishi-ku, Yokohama, 220-0011, Japan.
| | - Masahito Yasuda
- Department of Dermatology, Gunma University Graduate School of Medicine, 3-39-22 Showa-Machi, Maebashi, Gunma, 371-8511, Japan
| | - Saori Nishikawa
- Shiseido Global Innovation Center, 1-2-11 Takashima, Nishi-ku, Yokohama, 220-0011, Japan
| | - Eisuke Takai
- Shiseido Global Innovation Center, 1-2-11 Takashima, Nishi-ku, Yokohama, 220-0011, Japan
| | - Junichi Hosoi
- Shiseido Global Innovation Center, 1-2-11 Takashima, Nishi-ku, Yokohama, 220-0011, Japan
| | - Satoshi Amano
- Shiseido Global Innovation Center, 1-2-11 Takashima, Nishi-ku, Yokohama, 220-0011, Japan
| |
Collapse
|
38
|
Khanna M, Parish CR. Heparanase: Historical Aspects and Future Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:71-96. [PMID: 32274707 DOI: 10.1007/978-3-030-34521-1_3] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Heparanase is an endo-β-glucuronidase that cleaves at a limited number of internal sites the glycosaminoglycan heparan sulfate (HS). Heparanase enzymatic activity was first reported in 1975 and by 1983 evidence was beginning to emerge that the enzyme was a facilitator of tumor metastasis by cleaving HS chains present in blood vessel basement membranes and, thereby, aiding the passage of tumor cells through blood vessel walls. Due to a range of technical difficulties, it took another 16 years before heparanase was cloned and characterized in 1999 and a further 14 years before the crystal structure of the enzyme was solved. Despite these substantial deficiencies, there was steady progress in our understanding of heparanase long before the enzyme was fully characterized. For example, it was found as early as 1984 that activated T cells upregulate heparanase expression, like metastatic tumor cells, and the enzyme aids the entry of T cells and other leukocytes into inflammatory sites. Furthermore, it was discovered in 1989 that heparanase releases pre-existing growth factors and cytokines associated with HS in the extracellular matrix (ECM), the liberated growth factors/cytokines enhancing angiogenesis and wound healing. There were also the first hints that heparanase may have functions other than enzymatic activity, in 1995 it being reported that under certain conditions the enzyme could act as a cell adhesion molecule. Also, in the same year PI-88 (Muparfostat), the first heparanase inhibitor to reach and successfully complete a Phase III clinical trial was patented.Nevertheless, the cloning of heparanase (also known as heparanase-1) in 1999 gave the field an enormous boost and some surprises. The biggest surprise was that there is only one heparanase encoding gene in the mammalian genome, despite earlier research, based on substrate specificity, suggesting that there are at least three different heparanases. This surprising conclusion has remained unchanged for the last 20 years. It also became evident that heparanase is a family 79 glycoside hydrolase that is initially produced as a pro-enzyme that needs to be processed by proteases to form an enzymatically active heterodimer. A related molecule, heparanase-2, was also discovered that is enzymatically inactive but, remarkably, recently has been shown to inhibit heparanase-1 activity as well as acting as a tumor suppressor that counteracts many of the pro-tumor properties of heparanase-1.The early claim that heparanase plays a key role in tumor metastasis, angiogenesis and inflammation has been confirmed by many studies over the last 20 years. In fact, heparanase expression is enhanced in all major cancer types, namely carcinomas, sarcomas, and hematological malignancies, and correlates with increased metastasis and poor prognosis. Also, there is mounting evidence that heparanase plays a central role in the induction of inflammation-associated cancers. The enzymatic activity of heparanase has also emerged in unexpected situations, such as in the spread of HS-binding viruses and in Type-1 diabetes where the destruction of intracellular HS in pancreatic insulin-producing beta cells precipitates diabetes. But the most extraordinary recent discoveries have been with the realization that heparanase can exert a range of biological activities that are independent of its enzymatic function, most notably activation of several signaling pathways and being a transcription factor that controls methylation of histone tails. Collectively, these data indicate that heparanase is a truly multifunctional protein that has the additional property of cleaving HS chains and releasing from ECM and cell surfaces hundreds of HS-binding proteins with a plethora of functional consequences. Clearly, there are many unique features of this intriguing molecule that still remain to be explored and are highlighted in this Chapter.
Collapse
Affiliation(s)
- Mayank Khanna
- Department of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia.,Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Christopher R Parish
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia.
| |
Collapse
|
39
|
Heparanase: Cloning, Function and Regulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:189-229. [PMID: 32274711 DOI: 10.1007/978-3-030-34521-1_7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In 2019, we mark the 20th anniversary of the cloning of the human heparanase gene. Heparanase remains the only known enzyme to cleave heparan sulfate, which is an abundant component of the extracellular matrix. Thus, elucidating the mechanisms underlying heparanase expression and activity is critical to understanding its role in healthy and pathological settings. This chapter provides a historical account of the race to clone the human heparanase gene, describes the intracellular and extracellular function of the enzyme, and explores the various mechanisms regulating heparanase expression and activity at the gene, transcript, and protein level.
Collapse
|
40
|
El-Nadi M, Hassan H, Saleh ME, Nassar E, Ismail YM, Amer M, Greve B, Götte M, El-Shinawi M, Ibrahim SA. Induction of heparanase via IL-10 correlates with a high infiltration of CD163+ M2-type tumor-associated macrophages in inflammatory breast carcinomas. Matrix Biol Plus 2020; 6-7:100030. [PMID: 33543027 PMCID: PMC7852308 DOI: 10.1016/j.mbplus.2020.100030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 02/22/2020] [Accepted: 02/23/2020] [Indexed: 12/29/2022] Open
Abstract
Inflammatory breast cancer (IBC) is the most aggressive and lethal form of breast cancer, characterized by a high infiltration of tumor-associated macrophages and poor prognosis. To identify new biomarkers and to elucidate the molecular mechanisms underlying IBC pathogenesis, we investigated the expression pattern of heparanase (HPSE) and its activator cathepsin L (CTSL). First, we quantitated the HPSE and CTSL mRNA levels in a cohort of breast cancer patients after curative surgery (20 IBC and 20-non-IBC). We discovered that both HPSE and CTSL mRNA levels were significantly induced in IBC tissue vis-à-vis non-IBC patients (p <0 .05 and p <0 .001, respectively). According to the molecular subtypes, HPSE mRNA levels were significantly higher in carcinoma tissues of triple negative (TN)-IBC as compared to TN-non-IBC (p <0 .05). Mechanistically, we discovered that pharmacological inhibition of HPSE activity resulted in a significant reduction of invasiveness in the IBC SUM149 cell line. Moreover, siRNA-mediated HPSE knockdown significantly downregulated the expression of the metastasis-related gene MMP2 and the cancer stem cell marker CD44. We also found that IBC tumors revealed robust heparanase immune-reactivity and CD163+ M2-type tumor-associated macrophages, with a positive correlation of both markers. Moreover, the secretome of axillary tributaries blood IBC CD14+ monocytes and the cytokine IL-10 significantly upregulated HPSE mRNA and protein expression in SUM149 cells. Intriguingly, massively elevated IL-10 mRNA expression with a trend of positive correlation with HPSE mRNA expression was detected in carcinoma tissue of IBC. Our findings highlight a possible role played by CD14+ monocytes and CD163+ M2-type tumor-associated macrophages in regulating HPSE expression possibly via IL-10. Overall, we suggest that heparanase, cathepsin L and CD14+ monocytes-derived IL-10 may play an important role in the pathogenesis of IBC and their targeting could have therapeutic implications.
Collapse
Key Words
- CD163+ M2-type tumor-associated macrophages
- CTSL, cathepsin L
- Cathepsin L
- ECM, extracellular matrix
- ER, estrogen receptor
- FFPE, Formalin-Fixed Paraffin-Embedded
- HER-2, human epidermal growth factor receptor-2
- HPSE, heparanase
- HSPGs, heparan sulfate proteoglycans
- Heparanase
- IBC, inflammatory breast cancer;
- IL-10
- IRB, Institutional Review Board
- Inflammatory breast cancer
- Invasion
- MMP2, matrix metalloproteinase2
- MTT, 3-(4,5-dimethyl-2-yl)-2,5-diphenyltetrazolium bromide
- OGT 2115, 2-[4-[[3-(4-Bromophenyl)-1-oxo-2-propenyl]amino]-3-fluorophenyl]-5-benzoxazoleacetic acid
- PR, progesterone receptor
- TAMs, tumor-associated macrophages
- TN, triple negative
- TNF-α, tumor necrosis factor-α
- Triple negative subtype
- qPCR, quantitative real-time PCR
- rh IL-10, recombinant human interleukin-10
Collapse
Affiliation(s)
- Mennatullah El-Nadi
- Department of Zoology, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Hebatallah Hassan
- Department of Zoology, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Moshira Ezzat Saleh
- Department of Zoology, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Eyyad Nassar
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Yahia Mahmoud Ismail
- Medical Oncology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| | - Mahmoud Amer
- Department of Zoology, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Burkhard Greve
- Department of Radiotherapy-Radiooncology, University Hospital Münster, Münster, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Mohamed El-Shinawi
- Department of General Surgery, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| | | |
Collapse
|
41
|
Heparanase-The Message Comes in Different Flavors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:253-283. [DOI: 10.1007/978-3-030-34521-1_9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
42
|
Puerta-Guardo H, Glasner DR, Espinosa DA, Biering SB, Patana M, Ratnasiri K, Wang C, Beatty PR, Harris E. Flavivirus NS1 Triggers Tissue-Specific Vascular Endothelial Dysfunction Reflecting Disease Tropism. Cell Rep 2020; 26:1598-1613.e8. [PMID: 30726741 PMCID: PMC6934102 DOI: 10.1016/j.celrep.2019.01.036] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 10/27/2018] [Accepted: 01/09/2019] [Indexed: 01/22/2023] Open
Abstract
Flaviviruses cause systemic or neurotropic-encephalitic pathology in humans. The flavivirus nonstructural protein 1 (NS1) is a secreted glycoprotein involved in viral replication, immune evasion, and vascular leakage during dengue virus infection. However, the contribution of secreted NS1 from related flaviviruses to viral pathogenesis remains unknown. Here, we demonstrate that NS1 from dengue, Zika, West Nile, Japanese encephalitis, and yellow fever viruses selectively binds to and alters permeability of human endothelial cells from lung, dermis, umbilical vein, brain, and liver in vitro and causes tissue-specific vascular leakage in mice, reflecting the pathophysiology of each flavivirus. Mechanistically, each flavivirus NS1 leads to differential disruption of endothelial glycocalyx components, resulting in endothelial hyperpermeability. Our findings reveal the capacity of a secreted viral protein to modulate endothelial barrier function in a tissue-specific manner both in vitro and in vivo, potentially influencing virus dissemination and pathogenesis and providing targets for antiviral therapies and vaccine development. Puerta-Guardo et al. discover that five flavivirus NS1 proteins trigger hyperpermeability and vascular dysfunction in human endothelial cells and mice in a manner reflecting disease tropism. This tissue-specific tropism is partially determined by the capacity of NS1 to bind endothelial cells and is characterized by disruption of endothelial glycocalyx components.
Collapse
Affiliation(s)
- Henry Puerta-Guardo
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Dustin R Glasner
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Diego A Espinosa
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Scott B Biering
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Mark Patana
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Kalani Ratnasiri
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Chunling Wang
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - P Robert Beatty
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
43
|
Nasser NJ, Fox J, Agbarya A. Potential Mechanisms of Cancer-Related Hypercoagulability. Cancers (Basel) 2020; 12:cancers12030566. [PMID: 32121387 PMCID: PMC7139427 DOI: 10.3390/cancers12030566] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 11/16/2022] Open
Abstract
The association between cancer and thrombosis has been known for over a century and a half. However, the mechanisms that underlie this correlation are not fully characterized. Hypercoagulability in cancer patients can be classified into two main categories: Type I and Type II. Type I occurs when the balance of endogenous heparin production and degradation is disturbed, with increased degradation of endogenous heparin by tumor-secreted heparanase. Type II hypercoagulability includes all the other etiologies, with factors related to the patient, the tumor, and/or the treatment. Patients with poor performance status are at higher risk of venous thromboembolism (VTE). Tumors can result in VTE through direct pressure on blood vessels, resulting in stasis. Several medications for cancer are correlated with a high risk of thrombosis. These include hormonal therapy (e.g., tamoxifen), chemotherapy (e.g., cisplatin, thalidomide and asparaginase), molecular targeted therapy (e.g., lenvatinib, osimertinib), and anti-angiogenesis monoclonal antibodies (e.g., bevacizumab and ramucirumab).
Collapse
Affiliation(s)
- Nicola J. Nasser
- Department of Radiation Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, NY 10467, USA;
- Correspondence:
| | - Jana Fox
- Department of Radiation Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, NY 10467, USA;
| | - Abed Agbarya
- Institute of Oncology, Bnai Zion Medical Center, Haifa 31048, Israel;
| |
Collapse
|
44
|
Shang R, Lal N, Puri K, Hussein B, Rodrigues B. Involvement of Heparanase in Endothelial Cell-Cardiomyocyte Crosstalk. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:721-745. [PMID: 32274734 DOI: 10.1007/978-3-030-34521-1_30] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Traditionally, the management of diabetes has focused mainly on controlling high blood glucose levels. Unfortunately, despite valiant efforts to normalize this blood glucose, poor medication management predisposes these patients to heart failure. Following diabetes, how the heart utilizes different sources of fuel for energy is key to the development of heart failure. The diabetic heart switches from using both glucose and fats, to predominately using fats as an energy resource for maintaining its activities. This transformation to using fats as an exclusive source of energy is helpful in the initial stages of the disease and is tightly controlled. However, over the progression of diabetes, there is a loss of this controlled supply and use of fats, which ultimately has terrible consequences since the uncontrolled use of fats produces toxic by-products which weaken heart function and cause heart disease. Heparanase is a key player that directs how much fats are provided to the heart and does so in association with several partners like LPL and VEGFs. Together, they regulate the amount of fats supplied, and their subsequent breakdown to provide energy. Following diabetes, there is a disruption in this network resulting in fat oversupply and cell death. Understanding how the heparanase-LPL-VEGFs "ensemble" cooperates, and its dysfunction in the diabetic heart would be useful in restoring metabolic equilibrium and limiting diabetes-related cardiac damage.
Collapse
Affiliation(s)
- Rui Shang
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Nathaniel Lal
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Karanjit Puri
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Bahira Hussein
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Brian Rodrigues
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
45
|
Abstract
From 1999-2003, Oxford GlycoSciences (OGS) ran a successful drug discovery oncology programme to discover small molecule inhibitors of the Heparanase I enzyme (HPSE1). HPSE1 at the time was widely regarded as being the sole mammalian enzyme capable of cleaving Heparan Sulfate (HS). A second family protein member however called Heparanase 2 (HPSE2) including splice forms was subsequently discovered by PCR analysis based on EST sequences. HPSE2 was found to be expressed mainly in smooth muscle containing tissues, particularly bladder and brain. HPSE2 is poorly expressed in haematopoietic cells and placenta which contrasts with the HPSE1 distribution pattern. HPSE2 binds more strongly to HS than HPSE1 and is believed to out compete for substrate binding and so in effect act as a tumor suppressor. So far, all attempts to show specific HPSE2 endoglycosidase activity against HS have failed suggesting that the enzyme may act as a pseudoenzyme that has evolved to retain only certain non-catalytic heparanase like functions. A breakthrough in the elucidation of functional roles for HPSE2 came about in 2010 with the linkage of HPSE2 gene deletions and mutations to the development of Ochoa/Urofacial Syndrome. Future work into the mechanistic analysis of HPSE2's role in signalling, tumor suppression and bladder/nerve functioning are needed to fully explore the role of this family of proteins.
Collapse
|
46
|
Simeonovic CJ, Popp SK, Brown DJ, Li FJ, Lafferty ARA, Freeman C, Parish CR. Heparanase and Type 1 Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:607-630. [PMID: 32274728 DOI: 10.1007/978-3-030-34521-1_24] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Type 1 diabetes (T1D) results from autoimmune destruction of insulin-producing beta cells in pancreatic islets. The degradation of the glycosaminoglycan heparan sulfate (HS) by the endo-β-D-glycosidase heparanase plays a critical role in multiple stages of the disease process. Heparanase aids (i) migration of inflammatory leukocytes from the vasculature to the islets, (ii) intra-islet invasion by insulitis leukocytes, and (iii) selective destruction of beta cells. These disease stages are marked by the solubilization of HS in the subendothelial basement membrane (BM), HS breakdown in the peri-islet BM, and the degradation of HS inside beta cells, respectively. Significantly, healthy islet beta cells are enriched in highly sulfated HS which is essential for their viability, protection from damage by reactive oxygen species (ROS), beta cell function and differentiation. Consequently, mouse and human beta cells but not glucagon-producing alpha cells (which contain less-sulfated HS) are exquisitely vulnerable to heparanase-mediated damage. In vitro, the death of HS-depleted mouse and human beta cells can be prevented by HS replacement using highly sulfated HS mimetics or analogues. T1D progression in NOD mice and recent-onset T1D in humans correlate with increased expression of heparanase by circulating leukocytes of myeloid origin and heparanase-expressing insulitis leukocytes. Treatment of NOD mice with the heparanase inhibitor and HS replacer, PI-88, significantly reduced T1D incidence by 50%, impaired the development of insulitis and preserved beta cell HS. These outcomes identified heparanase as a novel destructive tool in T1D, distinct from the conventional cytotoxic and apoptosis-inducing mechanisms of autoreactive T cells. In contrast to exogenous catalytically active heparanase, endogenous heparanase may function in HS homeostasis, gene expression and insulin secretion in normal beta cells and immune gene expression in leukocytes. In established diabetes, the interplay between hyperglycemia, local inflammatory cells (e.g. macrophages) and heparanase contributes to secondary micro- and macro-vascular disease. We have identified dual activity heparanase inhibitors/HS replacers as a novel class of therapeutic for preventing T1D progression and potentially for mitigating secondary vascular disease that develops with long-term T1D.
Collapse
Affiliation(s)
- Charmaine J Simeonovic
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia.
| | - Sarah K Popp
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Debra J Brown
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Fei-Ju Li
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Antony R A Lafferty
- Department of Paediatrics, The Canberra Hospital, Woden, ACT, Australia.,The ANU Medical School, The Australian National University, Canberra, ACT, Australia
| | - Craig Freeman
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Christopher R Parish
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
47
|
Role of Heparanase in Macrophage Activation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:445-460. [PMID: 32274721 DOI: 10.1007/978-3-030-34521-1_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Macrophages represent one of the most diverse immunocyte populations, constantly shifting between various phenotypes/functional states. In addition to execution of vital functions in normal physiological conditions, macrophages represent a key contributing factor in the pathogenesis of some of the most challenging diseases, such as chronic inflammatory disorders, diabetes and its complications, and cancer. Macrophage polarization studies focus primarily on cytokine-mediated mechanisms. However, to explore the full spectrum of macrophage action, additional, non-cytokine pathways responsible for altering macrophage phenotype have to be taken into consideration as well. Heparanase, the only known mammalian endoglycosidase that cleaves heparan sulfate glycosaminoglycans, has been shown to contribute to the altered macrophage phenotypes in vitro and in numerous animal models of inflammatory conditions, occurring either in the presence of microbial products or in the setting of non-infectious "aseptic" inflammation. Here we discuss the involvement of heparanase in shaping macrophage responses and provide information that may help to establish the rationale for heparanase-targeting interventions aimed at preventing abnormal macrophage activation in various disorders.
Collapse
|
48
|
van der Vlag J, Buijsers B. Heparanase in Kidney Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:647-667. [PMID: 32274730 DOI: 10.1007/978-3-030-34521-1_26] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The primary filtration of blood occurs in the glomerulus in the kidney. Destruction of any of the layers of the glomerular filtration barrier might result in proteinuric disease. The glomerular endothelial cells and especially its covering layer, the glycocalyx, play a pivotal role in development of albuminuria. One of the main sulfated glycosaminoglycans in the glomerular endothelial glycocalyx is heparan sulfate. The endoglycosidase heparanase degrades heparan sulfate, thereby affecting glomerular barrier function, immune reactivity and inflammation. Increased expression of glomerular heparanase correlates with loss of glomerular heparan sulfate in many glomerular diseases. Most importantly, heparanase knockout in mice prevented the development of albuminuria after induction of experimental diabetic nephropathy and experimental glomerulonephritis. Therefore, heparanase could serve as a pharmacological target for glomerular diseases. Several factors that regulate heparanase expression and activity have been identified and compounds aiming to inhibit heparanase activity are currently explored.
Collapse
Affiliation(s)
- Johan van der Vlag
- Department of Nephrology (480), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands.
| | - Baranca Buijsers
- Department of Nephrology (480), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| |
Collapse
|
49
|
Vlodavsky I, Sanderson RD, Ilan N. Forty Years of Basic and Translational Heparanase Research. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:3-59. [PMID: 32274705 PMCID: PMC7142273 DOI: 10.1007/978-3-030-34521-1_1] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review summarizes key developments in the heparanase field obtained 20 years prior to cloning of the HPSE gene and nearly 20 years after its cloning. Of the numerous publications and review articles focusing on heparanase, we have selected those that best reflect the progression in the field as well as those we regard important accomplishments with preference to studies performed by scientists and groups that contributed to this book. Apart from a general 'introduction' and 'concluding remarks', the abstracts of these studies are presented essentially as published along the years. We apologize for not being objective and not being able to include some of the most relevant abstracts and references, due to space limitation. Heparanase research can be divided into two eras. The first, initiated around 1975, dealt with identifying the enzyme, establishing the relevant assay systems and investigating its biological activities and significance in cancer and other pathologies. Studies performed during the first area are briefly introduced in a layman style followed by the relevant abstracts presented chronologically, essentially as appears in PubMed. The second era started in 1999 when the heparanase gene was independently cloned by 4 research groups [1-4]. As expected, cloning of the heparanase gene boosted heparanase research by virtue of the readily available recombinant enzyme, molecular probes, and anti-heparanase antibodies. Studies performed during the second area are briefly introduced followed by selected abstracts of key findings, arranged according to specific topics.
Collapse
Affiliation(s)
- Israel Vlodavsky
- Technion Integrated Cancer Center (TICC) Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Haifa Israel
| | - Ralph D. Sanderson
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Neta Ilan
- Technion Integrated Cancer Center (TICC) Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Haifa Israel
| |
Collapse
|
50
|
Wu L, Davies GJ. An Overview of the Structure, Mechanism and Specificity of Human Heparanase. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:139-167. [PMID: 32274709 DOI: 10.1007/978-3-030-34521-1_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The retaining endo-β-D-glucuronidase Heparanase (HPSE) is the primary mammalian enzyme responsible for breakdown of the glycosaminoglycan heparan sulfate (HS). HPSE activity is essential for regulation and turnover of HS in the extracellular matrix, and its activity affects diverse processes such as inflammation, angiogenesis and cell migration. Aberrant heparanase activity is strongly linked to cancer metastasis, due to structural breakdown of extracellular HS networks and concomitant release of sequestered HS-binding growth factors. A full appreciation of HPSE activity in health and disease requires a structural understanding of the enzyme, and how it engages with its HS substrates. This chapter summarizes key findings from the recent crystal structures of human HPSE and its proenzyme. We present details regarding the 3-dimensional protein structure of HPSE and the molecular basis for its interaction with HS substrates of varying sulfation states. We also examine HPSE in a wider context against related β-D-glucuronidases from other species, highlighting the structural features that control exo/endo - glycosidase selectivity in this family of enzymes.
Collapse
Affiliation(s)
- Liang Wu
- York Structural Biology Laboratory, Department of Chemistry, The University of York, York, UK.
| | - Gideon J Davies
- York Structural Biology Laboratory, Department of Chemistry, The University of York, York, UK
| |
Collapse
|