1
|
Tu Y, Li Y, Qu G, Ning Y, Li B, Li G, Wu M, Li S, Huang Y. A Review of Basic Fibroblast Growth Factor Delivery Strategies and Applications in Regenerative Medicine. J Biomed Mater Res A 2025; 113:e37834. [PMID: 39740125 DOI: 10.1002/jbm.a.37834] [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: 08/29/2024] [Revised: 10/24/2024] [Accepted: 10/26/2024] [Indexed: 01/02/2025]
Abstract
Basic fibroblast growth factor (bFGF) is a significant member of the fibroblast growth factor (FGF) family. The bFGF has a three-dimensional structure comprising 12 reverse parallel β-folds. This structure facilitates tissue wound repair, angiogenesis, bone formation, cartilage repair, and nerve regeneration. Consequently, it has garnered significant attention from scholars both domestically and internationally. However, the instability and degradation properties of bFGF in vivo have limited its clinical application. Significant interest has arisen in the development of novel bFGF delivery systems that can address the shortcomings of bFGF and enhance its bioavailability by controlling the release amount, timing, and location. This article offers a comprehensive overview of the research and recent advances in various bFGF delivery systems, including hydrogels, liposomes, microspheres, and nanoparticles. Subsequently, the applications of bFGF pharmaceutical preparations in various fields are described. Finally, the current clinical applications of bFGF drug formulations and those in clinical trials are discussed, along with their clinical translation and future trends.
Collapse
Affiliation(s)
- Yuhan Tu
- Department of Pharmacy, Yueqing Third People's Hospital, Wenzhou, China
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Yang Li
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Gaoer Qu
- Department of Pharmacy, Yueqing Third People's Hospital, Wenzhou, China
| | - Yangyang Ning
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Bin Li
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Guoben Li
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Min Wu
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Shijun Li
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Yangge Huang
- Department of Pharmacy, Yueqing Third People's Hospital, Wenzhou, China
| |
Collapse
|
2
|
Shi J, Guan Y, Song H, Zhu L, Li J, Li Q, Hou N, Han F, Wang M, Zhang K, Shan M, Sun X, Qiu H. Exploring heparin's protective mechanism against AGEs induced endothelial injury. iScience 2024; 27:111084. [PMID: 39493878 PMCID: PMC11530820 DOI: 10.1016/j.isci.2024.111084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/12/2024] [Accepted: 09/27/2024] [Indexed: 11/05/2024] Open
Abstract
Advanced glycation end products (AGEs) in diabetes can cause endothelial damage. Heparin, widely known as a recognized anticoagulant, is also a multifunctional therapeutic drug. This study investigated whether heparin could ameliorate AGEs-induced endothelial injury. Remarkably, heparin effectively attenuated this cellular damage and assumed a reparative role. Furthermore, heparin inhibited the AGEs-RAGE-NFκB axis, thereby mitigating endothelial inflammatory injury. Comprehensive proteome and knockdown experiments suggested that heparin may exert a positive influence on cell growth and further alleviate pathological damage by upregulating the expression of LYAR (cell growth-regulating nucleolar protein). Diabetic mouse model was also used to further verify the changes of endothelial tissue in diabetic state and heparin intervention. In summary, these findings demonstrate that heparin has the potential to ameliorate AGEs-induced endothelial injury, opening new avenues for exploring the expanded therapeutic roles of heparin and its potential application in the management of diabetes and its associated complications.
Collapse
Affiliation(s)
- Junfeng Shi
- Department of Endocrinology and Metabolism, Affiliated Hospital of Shandong Second Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Yudong Guan
- School of Stomatology, Henan University, Kaifeng, Henan, China
| | - Hongwei Song
- Department of Endocrinology and Metabolism, Affiliated Hospital of Shandong Second Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Liang Zhu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Shandong Second Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Jingjing Li
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Qinying Li
- Department of Endocrinology and Metabolism, Affiliated Hospital of Shandong Second Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Shandong Second Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Meng Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Shandong Second Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Ming Shan
- Medical Research Center, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, P.R. China
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Shandong Second Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Hongyan Qiu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Shandong Second Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| |
Collapse
|
3
|
Ferreira FHDC, Farrell NP, Costa LAS. Spermine and spermidine SI-PPCs: Molecular dynamics reveals enhanced biomolecular interactions. Int J Biol Macromol 2024; 278:134654. [PMID: 39128748 DOI: 10.1016/j.ijbiomac.2024.134654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
In this paper the effects on the interaction of highly positively charged substitution-inert platinum polynuclear complexes (SI-PPCs) with negatively charged DNA and heparin are examined and compared by theoretical chemistry methods. Electrostatic and hydrogen bonding interactions contribute to the overall effects on the biomolecule. Root Mean Square (RMS) deviation, Solvent Accessible Surface, RMS fluctuation, and interaction analysis all confirm similar effects on both biomolecules, dictated predominantly by the total positive charge and total number of hydrogen bonds formed. Especially, changes in structural parameters suggesting condensation and reduction of available surface area will reduce or prevent normal protein recognition and may thus potentially inhibit biological mechanisms related to apoptosis (DNA) or reduced vascularization viability (HEP). Thermodynamic analyses supported these findings with favourable interaction energies. The comparison of DNA and heparin confirms the general intersectionality between the two biomolecules and confirms the intrinsic dual-nature function of this chemotype. The distinction between the two-limiting mode of actions (HS or DNA-centred) could reflect an intriguing balance between extracellular (GAG) and intracellular (DNA) binding and affinities. The results underline the need to fully understand GAG-small molecule interactions and their contribution to drug pharmacology and related therapeutic modalities. This report contributes to that understanding.
Collapse
Affiliation(s)
- Frederico Henrique do C Ferreira
- NEQC - Núcleo de Estudos em Química Computacional, Departamento de Química, ICE, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais 36036-900, Brazil
| | - Nicholas P Farrell
- Department of Chemistry, Virginia Commonwealth University, Richmond, VA 23284-2006, USA
| | - Luiz Antônio S Costa
- NEQC - Núcleo de Estudos em Química Computacional, Departamento de Química, ICE, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais 36036-900, Brazil.
| |
Collapse
|
4
|
Kocak FZ, Yar M, Rehman IU. In vitro degradation, swelling, and bioactivity performances of in situ forming injectable chitosan-matrixed hydrogels for bone regeneration and drug delivery. Biotechnol Bioeng 2024; 121:2767-2779. [PMID: 38837342 DOI: 10.1002/bit.28755] [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: 05/26/2023] [Revised: 12/25/2023] [Accepted: 05/12/2024] [Indexed: 06/07/2024]
Abstract
Injectable, tissue mimetic, bioactive, and biodegradable hydrogels offer less invasive regeneration and repair of tissues. The monitoring swelling and in vitro degradation capacities of hydrogels are highly important for drug delivery and tissue regeneration processes. Bioactivity of bone tissue engineered constructs in terms of mineralized apatite formation capacity is also pivotal. We have previously reported in situ forming chitosan-based injectable hydrogels integrated with hydroxyapatite and heparin for bone regeneration, promoting angiogenesis. These hydrogels were functionalized by glycerol and pH to improve their mechano-structural properties. In the present study, functionalized hybrid hydrogels were investigated for their swelling, in vitro degradation, and bioactivity performances. Hydrogels have degraded gradually in phosphate-buffered saline (PBS) with and without lysozyme enzyme. The percentage weight loss of hydrogels and their morphological and chemical properties, and pH of media were analyzed. The swelling ratio of hydrogels (55%-68%(wt), 6 h of equilibrium) indicated a high degree of cross-linking, can be suitable for controlled drug release. Hydrogels have gradually degraded reaching to 60%-70% (wt%) in 42 days in the presence and absence of lysozyme, respectively. Simulated body fluid (SBF)-treated hydrogels containing hydroxyapatite-induced needle-like carbonated-apatite mineralization was further enhanced by heparin content significantly.
Collapse
Affiliation(s)
- Fatma Zehra Kocak
- Engineering-Architecture Faculty, Metallurgy and Materials Engineering, Nevsehir Haci Bektas Veli University, Nevsehir, Turkey
- Engineering Department, Lancaster University, Lancaster, UK
| | - Muhammad Yar
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| | - Ihtesham U Rehman
- School of Medicine and Dentistry, University of Central Lancashire, Lancashire, UK
| |
Collapse
|
5
|
Demeter F, Peleskei Z, Kútvölgyi K, Rusznyák Á, Fenyvesi F, Kajtár R, Sipos É, Lekli I, Molnár P, Szöllősi AG, Lisztes E, Tóth BI, Borbás A, Herczeg M. Synthesis and Biological Profiling of Seven Heparin and Heparan Sulphate Analogue Trisaccharides. Biomolecules 2024; 14:1052. [PMID: 39334821 PMCID: PMC11429564 DOI: 10.3390/biom14091052] [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: 07/11/2024] [Revised: 08/07/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
Researchers are paying increasing attention to the strongly negatively charged heteropolysaccharides in cells, in the extracellular matrix or in the cell wall. Examples of such molecules are glycosaminoglycans (e.g., heparin, heparan sulphate). It is well known from the literature that heparin and its derivatives have anti-inflammatory, angiogenic, metastatic and growth factor inhibitory activity. Herein, we present the efficient synthesis of six non-glycosaminoglycan (Glc-GlcA-Glc-sequenced) and one heparin-related (GlcN-GlcA-Glc-sequenced) trisaccharides with various functional group patterns. The anti-inflammatory, antioxidant and cell growth-inhibitory/cytotoxic effects of the synthesized compounds were tested. Among the investigated molecules, we have found some derivatives with a promising anti-inflammatory and antioxidant effect.
Collapse
Affiliation(s)
- Fruzsina Demeter
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Zsófia Peleskei
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Katalin Kútvölgyi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Ágnes Rusznyák
- Department of Molecular and Nanopharmaceutics, Faculty of Pharmacy, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary
- Institute of Healthcare Industry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Ferenc Fenyvesi
- Department of Molecular and Nanopharmaceutics, Faculty of Pharmacy, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary
| | - Richárd Kajtár
- Department of Pharmacodynamics, Faculty of Pharmacy, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary
| | - Éva Sipos
- Department of Pharmacodynamics, Faculty of Pharmacy, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary
| | - István Lekli
- Department of Pharmacodynamics, Faculty of Pharmacy, University of Debrecen, Nagyerdei Körút 98, H-4032 Debrecen, Hungary
| | - Petra Molnár
- Department of Immunology, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Attila Gábor Szöllősi
- Department of Immunology, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Erika Lisztes
- Department of Physiology, University of Debrecen, P.O. Box 22, H-4012 Debrecen, Hungary
| | - Balázs István Tóth
- Department of Physiology, University of Debrecen, P.O. Box 22, H-4012 Debrecen, Hungary
- Department of Physiology, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Anikó Borbás
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
- HUN-REN-DE Molecular Recognition and Interaction Research Group, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Mihály Herczeg
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
- HUN-REN-DE Molecular Recognition and Interaction Research Group, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| |
Collapse
|
6
|
O’Hare N, Millican K, Ebong EE. Unraveling neurovascular mysteries: the role of endothelial glycocalyx dysfunction in Alzheimer's disease pathogenesis. Front Physiol 2024; 15:1394725. [PMID: 39027900 PMCID: PMC11254711 DOI: 10.3389/fphys.2024.1394725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/27/2024] [Indexed: 07/20/2024] Open
Abstract
While cardiovascular disease, cancer, and human immunodeficiency virus (HIV) mortality rates have decreased over the past 20 years, Alzheimer's Disease (AD) deaths have risen by 145% since 2010. Despite significant research efforts, effective AD treatments remain elusive due to a poorly defined etiology and difficulty in targeting events that occur too downstream of disease onset. In hopes of elucidating alternative treatment pathways, now, AD is commonly being more broadly defined not only as a neurological disorder but also as a progression of a variety of cerebrovascular pathologies highlighted by the breakdown of the blood-brain barrier. The endothelial glycocalyx (GCX), which is an essential regulator of vascular physiology, plays a crucial role in the function of the neurovascular system, acting as an essential vascular mechanotransducer to facilitate ultimate blood-brain homeostasis. Shedding of the cerebrovascular GCX could be an early indication of neurovascular dysfunction and may subsequently progress neurodegenerative diseases like AD. Recent advances in in vitro modeling, gene/protein silencing, and imaging techniques offer new avenues of scrutinizing the GCX's effects on AD-related neurovascular pathology. Initial studies indicate GCX degradation in AD and other neurodegenerative diseases and have begun to demonstrate a possible link to GCX loss and cerebrovascular dysfunction. This review will scrutinize the GCX's contribution to known vascular etiologies of AD and propose future work aimed at continuing to uncover the relationship between GCX dysfunction and eventual AD-associated neurological deterioration.
Collapse
Affiliation(s)
- Nicholas O’Hare
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
| | - Karina Millican
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Eno E. Ebong
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
- Department of Bioengineering, Northeastern University, Boston, MA, United States
- Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
7
|
Kim BS, Kim JU, Lee J, Ryu KM, Kim SH, Hwang NS. Decellularized brain extracellular matrix based NGF-releasing cryogel for brain tissue engineering in traumatic brain injury. J Control Release 2024; 368:140-156. [PMID: 38373473 DOI: 10.1016/j.jconrel.2024.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 02/21/2024]
Abstract
Traumatic brain injuries(TBI) pose significant challenges to human health, specifically neurological disorders and related motor activities. After TBI, the injured neuronal tissue is known for hardly regenerated and recovered to their normal neuron physiology and tissue compositions. For this reason, tissue engineering strategies that promote neuronal regeneration have gained increasing attention. This study explored the development of a novel neural tissue regeneration cryogel by combining brain-derived decellularized extracellular matrix (ECM) with heparin sulfate crosslinking that can perform nerve growth factor (NGF) release ability. Morphological and mechanical characterizations of the cryogels were performed to assess their suitability as a neural regeneration platform. After that, the heparin concnentration dependent effects of varying NGF concentrations on cryogel were investigated for their controlled release and impact on neuronal cell differentiation. The results revealed a direct correlation between the concentration of released NGF and the heparin sulfate ratio in cryogel, indicating that the cryogel can be tailored to carry higher loads of NGF with heparin concentration in cryogel that induced higher neuronal cell differentiation ratio. Furthermore, the study evaluated the NGF loaded cryogels on neuronal cell proliferation and brain tissue regeneration in vivo. The in vivo results suggested that the NGF loaded brain ECM derived cryogel significantly affects the regeneration of brain tissue. Overall, this research contributes to the development of advanced neural tissue engineering strategies and provides valuable insights into the design of regenerative cryogels that can be customized for specific therapeutic applications.
Collapse
Affiliation(s)
- Beom-Seok Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Jeong-Uk Kim
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaewoo Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyung Min Ryu
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Su-Hwan Kim
- Department of Chemical Engineering (BK21 FOUR), Dong-A University, Busan 49315, Republic of Korea
| | - Nathaniel S Hwang
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea; School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea; Bio-MAX Institute, Institute of Bio-Engineering, Seoul National University, Seoul 08826, Republic of Korea; Institute of Engineering Research, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
8
|
Truchan K, Osyczka AM. Noggin promotes osteogenesis in human adipose-derived mesenchymal stem cells via FGFR2/Src/Akt and ERK signaling pathway. Sci Rep 2024; 14:6724. [PMID: 38509118 PMCID: PMC10954655 DOI: 10.1038/s41598-024-56858-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
The balance between Noggin and bone morphogenetic proteins (BMPs) is important during early development and skeletal regenerative therapies. Noggin binds BMPs in the extracellular space, thereby preventing BMP signaling. However, Noggin may affect cell response not necessarily through the modulation of BMP signaling, raising the possibility of direct Noggin signaling through yet unspecified receptors. Here we show that in osteogenic cultures of adipose-derived stem cells (ASCs), Noggin activates fibroblast growth factor receptors (FGFRs), Src/Akt and ERK kinases, and it stabilizes TAZ proteins in the presence of dexamethasone. Overall, this leads ASCs to increased expression of osteogenic markers and robust mineral deposition. Our results also indicate that Noggin can induce osteogenic genes expression in normal human bone marrow stem cells and alkaline phosphatase activity in normal human dental pulp stem cells. Besides, Noggin can specifically activate FGFR2 in osteosarcoma cells. We believe our findings open new research avenues to further explore the involvement of Noggin in cell fate modulation by FGFR2/Src/Akt/ERK signaling and potential applications of Noggin in bone regenerative therapies.
Collapse
Affiliation(s)
- Karolina Truchan
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa St. 9, 30-387, Kraków, Poland.
| | - Anna Maria Osyczka
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa St. 9, 30-387, Kraków, Poland.
| |
Collapse
|
9
|
Meher MK, Naidu G, Mishra A, Poluri KM. A review on multifaceted biomedical applications of heparin nanocomposites: Progress and prospects. Int J Biol Macromol 2024; 260:129379. [PMID: 38242410 DOI: 10.1016/j.ijbiomac.2024.129379] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/21/2024]
Abstract
Advances in polymer-based nanocomposites have revolutionized biomedical applications over the last two decades. Heparin (HP), being a highly bioactive polymer of biological origin, provides strong biotic competence to the nanocomposites, broadening the horizon of their applicability. The efficiency, biocompatibility, and biodegradability properties of nanomaterials significantly improve upon the incorporation of heparin. Further, inclusion of structural/chemical derivatives, fractionates, and mimetics of heparin enable fabrication of versatile nanocomposites. Modern nanotechnological interventions have exploited the inherent biofunctionalities of heparin by formulating various nanomaterials, including inorganic/polymeric nanoparticles, nanofibers, quantum dots, micelles, liposomes, and nanogels ensuing novel functionalities targeting diverse clinical applications involving drug delivery, wound healing, tissue engineering, biocompatible coatings, nanosensors and so on. On this note, the present review explicitly summarises the recent HP-oriented nanotechnological developments, with a special emphasis on the reported successful engagement of HP and its derivatives/mimetics in nanocomposites for extensive applications in the laboratory and health-care facility. Further, the advantages and limitations/challenges specifically associated with HP in nanocomposites, undertaken in this current review are quintessential for future innovations/discoveries pertaining to HP-based nanocomposites.
Collapse
Affiliation(s)
- Mukesh Kumar Meher
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Goutami Naidu
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur 342011, Rajasthan, India
| | - Krishna Mohan Poluri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India; Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India.
| |
Collapse
|
10
|
Dürig J, Calcagni M, Buschmann J. Transition metals in angiogenesis - A narrative review. Mater Today Bio 2023; 22:100757. [PMID: 37593220 PMCID: PMC10430620 DOI: 10.1016/j.mtbio.2023.100757] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/14/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023] Open
Abstract
The aim of this paper is to offer a narrative review of the literature regarding the influence of transition metals on angiogenesis, excluding lanthanides and actinides. To our knowledge there are not any reviews up to date offering such a summary, which inclined us to write this paper. Angiogenesis describes the process of blood vessel formation, which is an essential requirement for human growth and development. When the complex interplay between pro- and antiangiogenic mediators falls out of balance, angiogenesis can quickly become harmful. As it is so fundamental, both its inhibition and enhancement take part in various diseases, making it a target for therapeutic treatments. Current methods come with limitations, therefore, novel agents are constantly being researched, with metal agents offering promising results. Various transition metals have already been investigated in-depth, with studies indicating both pro- and antiangiogenic properties, respectively. The transition metals are being applied in various formulations, such as nanoparticles, complexes, or scaffold materials. Albeit the increasing attention this field is receiving, there remain many unanswered questions, mostly regarding the molecular mechanisms behind the observed effects. Notably, approximately half of all the transition metals have not yet been investigated regarding potential angiogenic effects. Considering the promising results which have already been established, it should be of great interest to begin investigating the remaining elements whilst also further analyzing the established effects.
Collapse
Affiliation(s)
- Johannes Dürig
- University of Zürich, Faculty of Medicine, Pestalozzistrasse 3, 8032, Zurich, Switzerland
- University Hospital of Zürich, Department of Plastic Surgery and Hand Surgery, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Maurizio Calcagni
- University Hospital of Zürich, Department of Plastic Surgery and Hand Surgery, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Johanna Buschmann
- University Hospital of Zürich, Department of Plastic Surgery and Hand Surgery, Rämistrasse 100, 8091, Zürich, Switzerland
| |
Collapse
|
11
|
Xu L, He C, Yang S, Zhu Y, Wang P, Wu S, Guo F, Wang Y. Phase-transited lysozyme nanofilm with co-immobilized copper ion and heparin as cardiovascular stent multifunctional coating. Colloids Surf B Biointerfaces 2023; 230:113530. [PMID: 37683323 DOI: 10.1016/j.colsurfb.2023.113530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023]
Abstract
Cardiovascular metal stents have shown potential in the treatment of coronary artery disease using percutaneous coronary intervention. However, thrombosis, endothelialization, and new atherosclerosis after stent implantation remain unsolved problems. Herein, a multifunctional coating material based on phase-transited lysozyme was developed to promote stent endothelialization and simultaneously reduce thrombus events by embedding moieties of heparin and co-immobilized copper ions for in-situ catalyzing nitric oxide (NO) generation. The lysozyme-based biomimetic coating is compatible with blood and enables facile loading and sustainable release of copper ions to produce NO with donors via catalytic reaction. The novel coating strategy displayed several bio-effects of anti-thrombosis; it synergistically promoted endothelial cell growth and inhibited smooth muscle cell growth. Thus, this systemic in vitro study will provide a foundation for developing multifunctional cardiovascular stents in clinical settings.
Collapse
Affiliation(s)
- Lehua Xu
- The Institute for Translational Nanomedicine, Shanghai East Hospital, the Institute for Biomedical Engineering and Nano Science, School of Medicine, Tongji University, Shanghai 200092, PR China
| | - Chenlong He
- The Institute for Translational Nanomedicine, Shanghai East Hospital, the Institute for Biomedical Engineering and Nano Science, School of Medicine, Tongji University, Shanghai 200092, PR China
| | - Shusheng Yang
- Department of Laboratory Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, PR China
| | - Yunxia Zhu
- Department of Laboratory Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, PR China
| | - Peng Wang
- The Institute for Translational Nanomedicine, Shanghai East Hospital, the Institute for Biomedical Engineering and Nano Science, School of Medicine, Tongji University, Shanghai 200092, PR China
| | - Shengming Wu
- The Institute for Translational Nanomedicine, Shanghai East Hospital, the Institute for Biomedical Engineering and Nano Science, School of Medicine, Tongji University, Shanghai 200092, PR China
| | - Fangfang Guo
- The Institute for Translational Nanomedicine, Shanghai East Hospital, the Institute for Biomedical Engineering and Nano Science, School of Medicine, Tongji University, Shanghai 200092, PR China
| | - Yilong Wang
- The Institute for Translational Nanomedicine, Shanghai East Hospital, the Institute for Biomedical Engineering and Nano Science, School of Medicine, Tongji University, Shanghai 200092, PR China.
| |
Collapse
|
12
|
Schulze C, Danielsson A, Liwo A, Huster D, Samsonov SA, Penk A. Ligand binding of interleukin-8: a comparison of glycosaminoglycans and acidic peptides. Phys Chem Chem Phys 2023; 25:24930-24947. [PMID: 37694394 DOI: 10.1039/d3cp02457a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Recognition and binding of regulatory proteins to glycosaminoglycans (GAGs) from the extracellular matrix is a process of high biological importance. The interaction between negatively charged sulfate or carboxyl groups of the GAGs and clusters of basic amino acids on the protein is crucial in this binding process and it is believed that electrostatics represent the key factor for this interaction. However, given the rather undirected nature of electrostatics, it is important to achieve a clear understanding of its role in protein-GAG interactions and how specificity and selectivity in these systems can be achieved, when the classical key-lock binding motif is not applicable. Here, we compare protein binding of a highly charged heparin (HP) hexasaccharide with four de novo designed decapeptides of varying negative net charge. The charge density of these peptides was comparable to typical GAGs of the extracellular matrix. We used the regulatory protein interleukin-8 (IL-8) because its interactions with GAGs are well described. All four peptide ligands bind to the same epitope of IL-8 but show much weaker binding affinity as revealed in 1H-15N HSQC NMR titration experiments. Complementary molecular docking and molecular dynamics simulations revealed further atomistic details of the interaction mode of GAG versus peptide ligands. Overall, similar contributions to the binding energy and hydrogen bond formation are determined for HP and the highly charged peptides, suggesting that the entropic loss of the peptides upon binding likely account for the remarkably different affinity of GAG versus peptide ligands to IL-8.
Collapse
Affiliation(s)
- Christian Schulze
- Institute for Medical Physics and Biophysics, University of Leipzig, Härtelstr. 16/18, 04107 Leipzig, Germany.
| | - Annemarie Danielsson
- Faculty of Chemistry, University of Gdańsk, Fahrenheit Union of Universities, ul. Wita Stwosza 63, 80-308 Gdańsk, Poland.
| | - Adam Liwo
- Faculty of Chemistry, University of Gdańsk, Fahrenheit Union of Universities, ul. Wita Stwosza 63, 80-308 Gdańsk, Poland.
| | - Daniel Huster
- Institute for Medical Physics and Biophysics, University of Leipzig, Härtelstr. 16/18, 04107 Leipzig, Germany.
| | - Sergey A Samsonov
- Faculty of Chemistry, University of Gdańsk, Fahrenheit Union of Universities, ul. Wita Stwosza 63, 80-308 Gdańsk, Poland.
| | - Anja Penk
- Institute for Medical Physics and Biophysics, University of Leipzig, Härtelstr. 16/18, 04107 Leipzig, Germany.
| |
Collapse
|
13
|
Katner S, Ginsburg EP, Hampton JD, Peterson EJ, Koblinski JE, Farrell NP. A Comparison of Di- and Trinuclear Platinum Complexes Interacting with Glycosaminoglycans for Targeted Chemotherapy. ACS Med Chem Lett 2023; 14:1224-1230. [PMID: 37736178 PMCID: PMC10510529 DOI: 10.1021/acsmedchemlett.3c00244] [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: 06/01/2023] [Accepted: 08/30/2023] [Indexed: 09/23/2023] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) and their associated proteins aid in tumor progression through modulation of biological events such as cell invasion, angiogenesis, metastasis, and immunological responses. Metalloshielding of the anionic heparan sulfate (HS) chains by cationic polynuclear platinum complexes (PPCs) prevents the HS from interacting with HS-associated proteins and thus diminishes the critical functions of HSPG. Studies herein exploring the PPC-HS interactions demonstrated that a series of PPCs varying in charge, nuclearity, distance between Pt centers, and hydrogen-bonding ability influence HS affinity. We report that the polyamine-linked complexes have high HS affinity and display excellent in vivo activity against breast cancer metastases and those arising in the bone and liver compared to carboplatin. Overall, the PPC-HS niche offers an attractive approach for targeting HSPG-expressing tumor cells.
Collapse
Affiliation(s)
- Samantha
J. Katner
- Department
of Biochemistry, Chemistry, and Geology, Minnesota State University, Mankato, Mankato, Minnesota 56001, United States
| | - Eric P. Ginsburg
- Department
of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - James D. Hampton
- Department
of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284, United States
- Massey
Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Erica J. Peterson
- Department
of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284, United States
- Massey
Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Jennifer E. Koblinski
- Massey
Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, United States
- Department
of Pathology, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Nicholas P. Farrell
- Department
of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284, United States
- Massey
Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| |
Collapse
|
14
|
Shakibi S, Onck PR, Van der Giessen E. A One-Bead-Per-Saccharide (1BPS) Model for Glycosaminoglycans. J Chem Theory Comput 2023; 19:5491-5502. [PMID: 37459601 PMCID: PMC10448712 DOI: 10.1021/acs.jctc.3c00238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Indexed: 08/23/2023]
Abstract
Glycosaminoglycans (GAGs) are polysaccharide compounds that play key roles in various biological processes. GAGs are important structural components of cartilage and the extracellular matrix of the brain. Due to the large size of these polysaccharides, coarse-grained approaches are indispensable for modeling these biopolymers. We develop a one-bead-per-saccharide model of chondroitin sulfates and hyaluronic acid based on an existing three-bead-per-saccharide coarse-grained model. Our coarse graining is carried out by using iterative Boltzmann inversion (IBI), including an additional coupling potential to incorporate the correlation between dihedral angles. The predictions of the model are verified against those of the existing three-bead-per-saccharin model and the experimental radius of gyration for hyaluronic acid.
Collapse
Affiliation(s)
- Saber Shakibi
- Micromechanics of Materials, Zernike
Institute for Advanced Materials, University
of Groningen, 9747 AG Groningen, The Netherlands
| | - Patrick R. Onck
- Micromechanics of Materials, Zernike
Institute for Advanced Materials, University
of Groningen, 9747 AG Groningen, The Netherlands
| | - Erik Van der Giessen
- Micromechanics of Materials, Zernike
Institute for Advanced Materials, University
of Groningen, 9747 AG Groningen, The Netherlands
| |
Collapse
|
15
|
Christian JM, Zoepfl M, Johnson WE, Ginsburg E, Peterson EJ, Hampton JD, Farrell NP. Glycosaminoglycan-directed cobalt complexes. J Inorg Biochem 2023; 245:112254. [PMID: 37182504 DOI: 10.1016/j.jinorgbio.2023.112254] [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: 03/15/2023] [Revised: 04/20/2023] [Accepted: 05/08/2023] [Indexed: 05/16/2023]
Abstract
The biological activity of the 6+ Co containing Werner's Complex has been described and mechanistic considerations suggest that the highly anionic glycosaminoglycans (heparan sulfate, HS, GAGs) are implicated in this activity [Paiva et al. 2021]. To examine in detail the molecular basis of Werner's Complex biological properties we have examined a selection of simple mononuclear Co3+ compounds for their interactions with HS and Fondaparinux (FPX). FPX is a highly sulfated synthetic pentasaccharide used as a model HS substrate [Mangrum et al. 2014, Peterson et al. 2017]. The Co complexes were chosen to be formally substitution-inert and/or have the potential for covalent binding to the biomolecule. Using both indirect competitive inhibition assays and direct mass spectrometric assays, formally substitution-inert complexes bound to FPX with protection from multiple sulfate loss in the gas phase through metalloshielding. Covalent binding of Co-Cl complexes as in [CoCl(NH3)5]2+ and cis-[CoCl2(en)2]+ was confirmed by mass spectrometry. Interestingly, the former complex was shown to be an effective inhibitor of bacterial heparinase enzyme activity and to inhibit heparanase-dependent cellular invasion through the extracellular matrix (ECM). Pursuing the theme of metalloglycomics, we have observed the hitherto unappreciated biological activity of the simple [CoCl(NH3)5]2+ compound, a staple of most inorganic chemistry lab curricula.
Collapse
Affiliation(s)
- Jessica M Christian
- Department of Chemistry, Virginia Commonwealth University, 1001 W Main St, Richmond, VA 23284, United States of America
| | - Mary Zoepfl
- Department of Chemistry, Virginia Commonwealth University, 1001 W Main St, Richmond, VA 23284, United States of America
| | - Wyatt E Johnson
- Massey Cancer Center, Virginia Commonwealth University, 1300 E Marshall Street, Richmond, VA 23298-0037, United States of America
| | - Eric Ginsburg
- Department of Chemistry, Virginia Commonwealth University, 1001 W Main St, Richmond, VA 23284, United States of America
| | - Erica J Peterson
- Massey Cancer Center, Virginia Commonwealth University, 1300 E Marshall Street, Richmond, VA 23298-0037, United States of America
| | - J David Hampton
- Massey Cancer Center, Virginia Commonwealth University, 1300 E Marshall Street, Richmond, VA 23298-0037, United States of America
| | - Nicholas P Farrell
- Department of Chemistry, Virginia Commonwealth University, 1001 W Main St, Richmond, VA 23284, United States of America; Massey Cancer Center, Virginia Commonwealth University, 1300 E Marshall Street, Richmond, VA 23298-0037, United States of America.
| |
Collapse
|
16
|
Hogwood J, Mulloy B, Lever R, Gray E, Page CP. Pharmacology of Heparin and Related Drugs: An Update. Pharmacol Rev 2023; 75:328-379. [PMID: 36792365 DOI: 10.1124/pharmrev.122.000684] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 02/17/2023] Open
Abstract
Heparin has been used extensively as an antithrombotic and anticoagulant for close to 100 years. This anticoagulant activity is attributed mainly to the pentasaccharide sequence, which potentiates the inhibitory action of antithrombin, a major inhibitor of the coagulation cascade. More recently it has been elucidated that heparin exhibits anti-inflammatory effect via interference of the formation of neutrophil extracellular traps and this may also contribute to heparin's antithrombotic activity. This illustrates that heparin interacts with a broad range of biomolecules, exerting both anticoagulant and nonanticoagulant actions. Since our previous review, there has been an increased interest in these nonanticoagulant effects of heparin, with the beneficial role in patients infected with SARS2-coronavirus a highly topical example. This article provides an update on our previous review with more recent developments and observations made for these novel uses of heparin and an overview of the development status of heparin-based drugs. SIGNIFICANCE STATEMENT: This state-of-the-art review covers recent developments in the use of heparin and heparin-like materials as anticoagulant, now including immunothrombosis observations, and as nonanticoagulant including a role in the treatment of SARS-coronavirus and inflammatory conditions.
Collapse
Affiliation(s)
- John Hogwood
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (B.M., E.G., C.P.P.); National Institute for Biological Standards and Control, South Mimms, Hertfordshire, United Kingdom (J.H., E.G.) and School of Pharmacy, University College London, London, United Kingdom (R.L.)
| | - Barbara Mulloy
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (B.M., E.G., C.P.P.); National Institute for Biological Standards and Control, South Mimms, Hertfordshire, United Kingdom (J.H., E.G.) and School of Pharmacy, University College London, London, United Kingdom (R.L.)
| | - Rebeca Lever
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (B.M., E.G., C.P.P.); National Institute for Biological Standards and Control, South Mimms, Hertfordshire, United Kingdom (J.H., E.G.) and School of Pharmacy, University College London, London, United Kingdom (R.L.)
| | - Elaine Gray
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (B.M., E.G., C.P.P.); National Institute for Biological Standards and Control, South Mimms, Hertfordshire, United Kingdom (J.H., E.G.) and School of Pharmacy, University College London, London, United Kingdom (R.L.)
| | - Clive P Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (B.M., E.G., C.P.P.); National Institute for Biological Standards and Control, South Mimms, Hertfordshire, United Kingdom (J.H., E.G.) and School of Pharmacy, University College London, London, United Kingdom (R.L.)
| |
Collapse
|
17
|
Direct thrombin inhibitors as alternatives to heparin to preserve lung growth and function in a murine model of compensatory lung growth. Sci Rep 2022; 12:21117. [PMID: 36477689 PMCID: PMC9729628 DOI: 10.1038/s41598-022-25773-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Infants with congenital diaphragmatic hernia (CDH) may require cardiopulmonary bypass and systemic anticoagulation. Expeditious lung growth while on bypass is essential for survival. Previously, we demonstrated that heparin impairs lung growth and function in a murine model of compensatory lung growth (CLG). We investigated the effects of the direct thrombin inhibitors (DTIs) bivalirudin and argatroban. In vitro assays of lung endothelial cell proliferation and apoptosis were performed. C57BL/6 J mice underwent left pneumonectomy and subcutaneous implantation of osmotic pumps. Pumps were pre-loaded with normal saline (control), bivalirudin, argatroban, or heparin and outcomes were assessed on postoperative day 8. Heparin administration inhibited endothelial cell proliferation in vitro and significantly decreased lung volume in vivo, while bivalirudin and argatroban preserved lung growth. These findings correlated with changes in alveolarization on morphometric analysis. Treadmill exercise tolerance testing demonstrated impaired exercise performance in heparinized mice; bivalirudin/argatroban did not affect exercise tolerance. On lung protein analysis, heparin decreased angiogenic signaling which was not impacted by bivalirudin or argatroban. Together, this data supports the use of DTIs as alternatives to heparin for systemic anticoagulation in CDH patients on bypass. Based on this work, clinical studies on the impact of heparin and DTIs on CDH outcomes are warranted.
Collapse
|
18
|
Xiao Z, Fu D, Zhang L, Fan W, Shen X, Qi X. Bone healing study of alendronate combined with enoxaparin sodium bone cement in rabbits with bone defects. J Orthop Surg Res 2022; 17:431. [PMID: 36175933 PMCID: PMC9524070 DOI: 10.1186/s13018-022-03330-y] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/21/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND To observe the effect of enoxaparin sodium-polymethyl methacrylate (ES-PMMA) bone cement supplemented with alendronate (AN) on bone repair of bone defects in New Zealand rabbits. METHODS Twenty-seven New Zealand rabbits were randomly divided into ES/AN, ES-PMMA and PMMA groups, with a total of 27 New Zealand rabbits. The drugs loaded in 40 g bone cement powder were as follows: ES/AN group 8000 AxaIU enoxaparin (ES) and 200 mg alendronate (AN), ES-PMMA group 8000 AxaIU enoxaparin (ES), PMMA group without drugs. A bone defect model with a length of 10 mm and a diameter of 5 mm was made from the left tibia of rabbits, and the prepared bone cement was placed in the tibia defect. At 4 weeks, 8 weeks and 12 weeks after the operation, 3 rabbits in each group were sacrificed, and left tibia samples were collected for histological scoring, HE staining and Masson staining. Bone mineral density and new bone volume were measured by imaging, and the related data were processed by one-way ANOVA and least significance difference (LSD) post hoc test. RESULTS (1) Bone mineral density (BMD, mg/mm3) around the bone defect: at the 4th week, BMD in the ES/AN group was higher than that in the PMMA group; at the 8th week, the BMD in the ES/AN group was significantly higher than that in the other two groups; and at the 12th week, the BMD in the ES/AN group was significantly higher than that in the other two groups. (2) New bone volume (BV, mm3): at the 4th week, BV in the ES/AN group was significantly higher than that in the other two groups, BV in the ES/AN group was significantly higher than that in the other two groups at the 8th and 12th weeks, and BV in the ES-PMMA group was higher than that in the PMMA group. (3) Histological score: at the 4th and 8th weeks, the histological score of the ES/AN group was higher than that of the PMMA group, and at the 12th week, the histological score of the ES/AN group was higher than that of the other two groups. (4) Cortical bone thickness (μm): at the 4th, 8th and 12th weeks, the cortical bone thickness in the ES/AN group was higher than that in the other two groups, and the cortical bone thickness in the ES-PMMA group was higher than that in the PMMA group. (5) The percentage of mature area of new bone in the ES/AN group was higher than that in the other two groups at the 4th week, and at the 8th and 12th weeks, the percentage of mature area of new bone in the ES/AN group and ES-PMMA group was significantly higher than that in the PMMA group. CONCLUSION (1) Enoxaparin sodium bone cement supplemented with alendronate was superior to enoxaparin sodium bone cement and PMMA bone cement in promoting bone repair of tibial bone defects in New Zealand rabbits. (2) Enoxaparin sodium bone cement is superior to PMMA bone cement in promoting bone repair, showing a certain osteogenic potential.
Collapse
Affiliation(s)
- Zhihang Xiao
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050035, People's Republic of China
| | - Dehao Fu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Li Zhang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050035, People's Republic of China
| | - Weiye Fan
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050035, People's Republic of China
| | - Xiaoyu Shen
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050035, People's Republic of China
| | - Xiangbei Qi
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050035, People's Republic of China.
| |
Collapse
|
19
|
Ritter GS, Dolgova EV, Petrova DD, Efremov YR, Proskurina AS, Potter EA, Ruzanova VS, Kirikovich SS, Levites EV, Taranov OS, Ostanin AA, Chernykh ER, Kolchanov NA, Bogachev SS. The new general biological property of stem-like tumor cells Part I. Peculiarities of the process of the double-stranded DNA fragments internalization into stem-like tumor cells. Front Genet 2022; 13:954395. [PMID: 36159968 PMCID: PMC9492886 DOI: 10.3389/fgene.2022.954395] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/15/2022] [Indexed: 11/19/2022] Open
Abstract
Stem-like tumor cells of ascites carcinoma Krebs-2 and Epstein-Barr virus-induced B-lymphoma were shown to possess the innate capability of binding and internalizing the TAMRA-labeled double-stranded DNA (dsDNA) probe. The process of binding and internalizing is rather complicated and composed of the following successive stages: 1) initiating electrostatic interaction and contact of a negatively charged dsDNA molecule with a positively charged molecule(s) on the surface of a stem-like tumor cell; 2) binding of the dsDNA probe to a tumor stem cell surface protein(s) via the formation of a strong chemical/molecular bond; and 3) the very internalization of dsDNA into the cell. Binding of DNA to cell surface proteins is determined by the presence of heparin/polyanion-binding sites within the protein structure, which can be competitively blocked by heparin and/or dextran sulfate, wherein heparin blocks only the binding, while dextran sulfate abrogates both binding and internalization. The abrogation of internalization by dextran sulfate implies the role of scavenger receptors in this process. Cells were shown to uptake DNA in amounts constituting ∼0.008% of the haploid genome. Inhibitors of caveolae-dependent internalization abrogate the DNA uptake in Krebs-2 cells, and inhibitors of the clathrin/caveolar mechanism block the internalization in B-lymphoma cells. In the present report, it is shown for the first time that in contrast to the majority of committed tumor cells, stem-like tumor cells of Krebs-2 and B-lymphoma carry a general positive charge on their surface.
Collapse
Affiliation(s)
- Genrikh S. Ritter
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Evgeniya V. Dolgova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Daria D. Petrova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Yaroslav R. Efremov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- Novosibirsk National Research State University, Novosibirsk, Russia
| | - Anastasia S. Proskurina
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Ekaterina A. Potter
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Vera S. Ruzanova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- Novosibirsk National Research State University, Novosibirsk, Russia
| | - Svetlana S. Kirikovich
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Evgeniy V. Levites
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Oleg S. Taranov
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, Russia
| | - Alexandr A. Ostanin
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Elena R. Chernykh
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Nikolay A. Kolchanov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Sergey S. Bogachev
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
20
|
Field CJ, Perez AM, Samet T, Ricles V, Iovine MK, Lowe-Krentz LJ. Involvement of transmembrane protein 184a during angiogenesis in zebrafish embryos. Front Physiol 2022; 13:845407. [PMID: 36117693 PMCID: PMC9478037 DOI: 10.3389/fphys.2022.845407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
Angiogenesis, the outgrowth of new blood vessels from existing vasculature, is critical during development, tissue formation, and wound healing. In response to vascular endothelial growth factors (VEGFs), endothelial cells are activated to proliferate and move towards the signal, extending the vessel. These events are directed by VEGF-VEGF receptor (Vegfr2) signal transduction, which in turn is modulated by heparan sulfate proteoglycans (HSPGs). HSPGs are glycoproteins covalently attached to HS glycosaminoglycan chains. Transmembrane protein 184a (Tmem184a) has been recently identified as a heparin receptor, which is believed to bind heparan sulfate chains in vivo. Therefore, Tmem184a has the potential to fine-tune interactions between VEGF and HS, modulating Vegfr2-dependent angiogenesis. The function of Tmem184a has been investigated in the regenerating zebrafish caudal fin, but its role has yet to be evaluated during developmental angiogenesis. Here we provide insights into how Tmem184a contributes to the proper formation of the vasculature in zebrafish embryos. First, we find that knockdown of Tmem184a causes a reduction in the number of intact intersegmental vessels (ISVs) in the zebrafish embryo. This phenotype mimics that of vegfr2b knockout mutants, which have previously been shown to exhibit severe defects in ISV development. We then test the importance of HS interactions by removing the binding domain within the Tmem184a protein, which has a negative effect on angiogenesis. Tmem184a is found to act synergistically with Vegfr2b, indicating that the two gene products function in a common pathway to modulate angiogenesis. Moreover, we find that knockdown of Tmem184a leads to an increase in endothelial cell proliferation but a decrease in the amount of VE-cadherin present. Together, these findings suggest that Tmem184a is necessary for ISVs to organize into mature, complete vessels.
Collapse
|
21
|
The Choice of Anticoagulant Influences the Characteristics of Bone Marrow Aspirate Concentrate and Mesenchymal Stem Cell Bioactivity In Vitro. Stem Cells Int 2022; 2022:8259888. [PMID: 35910535 PMCID: PMC9337942 DOI: 10.1155/2022/8259888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 11/28/2022] Open
Abstract
Bone marrow aspirate concentrate (BMC) is commonly used as a therapeutic agent to resolve orthopedic injuries, using its unique cellularity to reduce inflammation and prime the region for repair. The aspiration of the bone marrow is performed using either sodium citrate (SC) or heparin sodium (HS) as an anticoagulant and processed via centrifugation to concentrate the cellular constituents. To date, the consideration of the impact of the two commonly used anticoagulants on the mesenchymal stem/stromal cell (MSC) population has been overlooked. The current study assesses the differences in the BMCs produced using 15% SC and HS at 1,000 U/mL or 100 U/mL final v./v. as an anticoagulant using in vitro metrics including total nucleated cell counts (TNC) and viability, the ability for mesenchymal stromal/stem cells (MSCs) to establish colony-forming units with fibroblast morphology (CFU-f), and cytokine expression profile of the MSC cultures. Our findings demonstrate that HS-derived BMC cultures result in higher CFU-f formation and CFU-f frequency at both concentrations assessed compared to SC-derived BMC cultures. In addition, there were significant differences in 27% (7 of 26) of the cytokines quantified in HS-derived BMC cultures compared to SC-derived BMC cultures with implications for MSC plasticity and self-renewal.
Collapse
|
22
|
Glycosaminoglycan interaction networks and databases. Curr Opin Struct Biol 2022; 74:102355. [DOI: 10.1016/j.sbi.2022.102355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/14/2022]
|
23
|
Roy R, Jonniya NA, Kar P. Effect of Sulfation on the Conformational Dynamics of Dermatan Sulfate Glycosaminoglycan: A Gaussian Accelerated Molecular Dynamics Study. J Phys Chem B 2022; 126:3852-3866. [PMID: 35594147 DOI: 10.1021/acs.jpcb.2c01807] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Glycosaminoglycans (GAGs) are anionic biopolymers present on cell surfaces as a part of proteoglycans. The biological activities of GAGs depend on the sulfation pattern. In our study, we have considered three octadecasaccharide dermatan sulfate (DS) chains with increasing order of sulfation (dp6s, dp7s, and dp12s) to illuminate the role of sulfation on the GAG units and its chain conformation through 10 μs-long Gaussian accelerated molecular dynamics simulations. DS is composed of repeating disaccharide units of iduronic acid (IdoA) and N-acetylgalactosamine (N-GalNAc). Here, N-GalNAc is linked to IdoA via β(1-4), while IdoA is linked to N-GalNAc through α(1-3). With the increase in sulfation, the DS structure becomes more rigid and linear, as is evident from the distribution of root-mean-square deviations (RMSDs) and end-to-end distances. The tetrasaccharide linker region of the main chain shows a rigid conformation in terms of the glycosidic linkage. We have observed that upon sulfation (i.e., dp12s), the ring flip between two chair forms vanished for IdoA. The dynamic cross-correlation analysis reveals that the anticorrelation motions in dp12s are reduced significantly compared to dp6s or dp7s. An increase in sulfation generates relatively more stable hydrogen-bond networks, including water bridging with the neighboring monosaccharides. Despite the favorable linear structures of the GAG chains, our study also predicts few significant bendings related to the different puckering states, which may play a notable role in the function of the DS. The relation between the global conformation with the micro-level parameters such as puckering and water-mediated hydrogen bonds shapes the overall conformational space of GAGs. Overall, atomistic details of the DS chain provided in this study will help understand their functional and mechanical roles, besides developing new biomaterials.
Collapse
Affiliation(s)
- Rajarshi Roy
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Indore 453552, Madhya Pradesh, India
| | - Nisha Amarnath Jonniya
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Indore 453552, Madhya Pradesh, India
| | - Parimal Kar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Indore 453552, Madhya Pradesh, India
| |
Collapse
|
24
|
Kocak FZ, Yar M, Rehman IU. Hydroxyapatite-Integrated, Heparin- and Glycerol-Functionalized Chitosan-Based Injectable Hydrogels with Improved Mechanical and Proangiogenic Performance. Int J Mol Sci 2022; 23:ijms23105370. [PMID: 35628172 PMCID: PMC9140455 DOI: 10.3390/ijms23105370] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 02/04/2023] Open
Abstract
The investigation of natural bioactive injectable composites to induce angiogenesis during bone regeneration has been a part of recent minimally invasive regenerative medicine strategies. Our previous study involved the development of in situ-forming injectable composite hydrogels (Chitosan/Hydroxyapatite/Heparin) for bone regeneration. These hydrogels offered facile rheology, injectability, and gelation at 37 °C, as well as promising pro-angiogenic abilities. In the current study, these hydrogels were modified using glycerol as an additive and a pre-sterile production strategy to enhance their mechanical strength. These modifications allowed a further pH increment during neutralisation with maintained solution homogeneity. The synergetic effect of the pH increment and further hydrogen bonding due to the added glycerol improved the strength of the hydrogels substantially. SEM analyses showed highly cross-linked hydrogels (from high-pH solutions) with a hierarchical interlocking pore morphology. Hydrogel solutions showed more elastic flow properties and incipient gelation times decreased to just 2 to 3 min at 37 °C. Toluidine blue assay and SEM analyses showed that heparin formed a coating at the top layer of the hydrogels which contributed anionic bioactive surface features. The chick chorioallantoic membrane (CAM) assay confirmed significant enhancement of angiogenesis with chitosan-matrixed hydrogels comprising hydroxyapatite and small quantities of heparin (33 µg/mL) compared to basic chitosan hydrogels.
Collapse
Affiliation(s)
- Fatma Z. Kocak
- Engineering-Architecture Faculty, Metallurgy and Material Engineering, Nevsehir Haci Bektas Veli University, Nevsehir 50300, Turkey;
- Engineering Department, Lancaster University, Lancaster LA1 4YW, UK
| | - Muhammad Yar
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS University Islamabad, Lahore Campus, Lahore 54000, Pakistan;
| | - Ihtesham U. Rehman
- Engineering-Architecture Faculty, Metallurgy and Material Engineering, Nevsehir Haci Bektas Veli University, Nevsehir 50300, Turkey;
- Correspondence:
| |
Collapse
|
25
|
Corti F, Ristori E, Rivera-Molina F, Toomre D, Zhang J, Mihailovic J, Zhuang ZW, Simons M. Syndecan-2 selectively regulates VEGF-induced vascular permeability. NATURE CARDIOVASCULAR RESEARCH 2022; 1:518-528. [PMID: 36212522 PMCID: PMC9544384 DOI: 10.1038/s44161-022-00064-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 04/06/2022] [Indexed: 02/03/2023]
Abstract
Vascular endothelial growth factor (VEGF)- driven increase in vascular permeability is a key feature of many disease states associated with inflammation and ischemic injury, contributing significantly to morbidity and mortality in these settings. Despite its importance, no specific regulators that preferentially control VEGF-dependent increase in permeability versus its other biological activities, have been identified. Here we report that a proteoglycan Syndecan-2 (Sdc2) regulates the interaction between a transmembrane phosphatase DEP1 and VEGFR2 by controlling cell surface levels of DEP1. In the absence of Sdc2 or the presence of an antibody that blocks Sdc2-DEP1 interaction, increased plasma membrane DEP1 levels promote selective dephosphorylation of the VEGFR2 Y951 site that is involved in permeability control. Either an endothelial-specific Sdc2 deletion or a treatment with an anti-Sdc2 antibody result in a highly significant reduction in stroke size due to a decrease in intracerebral edema.
Collapse
Affiliation(s)
- F Corti
- Yale Cardiovascular Research Center Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - E Ristori
- Yale Cardiovascular Research Center Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - F Rivera-Molina
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - D Toomre
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - J Zhang
- Yale Cardiovascular Research Center Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - J Mihailovic
- Department of Radiology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Z W Zhuang
- Yale Cardiovascular Research Center Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - M Simons
- Yale Cardiovascular Research Center Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| |
Collapse
|
26
|
Rosa NMP, Ferreira FHDC, Farrell NP, Costa LAS. Substitution-inert polynuclear platinum complexes and Glycosaminoglycans: A molecular dynamics study of its non-covalent interactions. J Inorg Biochem 2022; 232:111811. [DOI: 10.1016/j.jinorgbio.2022.111811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/17/2022] [Accepted: 03/25/2022] [Indexed: 10/18/2022]
|
27
|
Production, characteristics and applications of microbial heparinases. Biochimie 2022; 198:109-140. [DOI: 10.1016/j.biochi.2022.03.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/03/2022] [Accepted: 03/28/2022] [Indexed: 12/26/2022]
|
28
|
Reyna-Urrutia VA, González-González AM, Rosales-Ibáñez R. Compositions and Structural Geometries of Scaffolds Used in the Regeneration of Cleft Palates: A Review of the Literature. Polymers (Basel) 2022; 14:polym14030547. [PMID: 35160534 PMCID: PMC8840587 DOI: 10.3390/polym14030547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
Cleft palate (CP) is one of the most common birth defects, presenting a multitude of negative impacts on the health of the patient. It also leads to increased mortality at all stages of life, economic costs and psychosocial effects. The embryological development of CP has been outlined thanks to the advances made in recent years due to biomolecular successions. The etiology is broad and combines certain environmental and genetic factors. Currently, all surgical interventions work off the principle of restoring the area of the fissure and aesthetics of the patient, making use of bone substitutes. These can involve biological products, such as a demineralized bone matrix, as well as natural–synthetic polymers, and can be supplemented with nutrients or growth factors. For this reason, the following review analyzes different biomaterials in which nutrients or biomolecules have been added to improve the bioactive properties of the tissue construct to regenerate new bone, taking into account the greatest limitations of this approach, which are its use for bone substitutes for large areas exclusively and the lack of vascularity. Bone tissue engineering is a promising field, since it favors the development of porous synthetic substitutes with the ability to promote rapid and extensive vascularization within their structures for the regeneration of the CP area.
Collapse
|
29
|
Zhang S, Yan H, Ma X, Zheng W, Wang W. Effects of different routes of heparin on instant blood-mediated inflammatory reaction after portal vein islet transplantation. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2022; 47:1-7. [PMID: 35545357 PMCID: PMC10930478 DOI: 10.11817/j.issn.1672-7347.2022.200993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Indexed: 06/15/2023]
Abstract
OBJECTIVES Heparin is mainly used as an anticoagulant in clinic, and it also has a certain anti-inflammatory effect. At present, after portal vein islet transplantation in diabetic patients, heparin is mainly infused through the peripheral veins of the limbs to achieve the purpose of anticoagulation and protection of the graft, rather than through the portal vein. In this study, animal experiments were conducted to investigate the effect of heparin infusion via the portal vein and marginal ear vein on the instant blood-mediated inflammatory reaction (IBMIR) after portal vein islet transplantation, which is the choice of anticoagulation methods for clinical islet transplantation to provide a basis for decision-making. METHODS A total of 50 neonatal pigs (Xeno-1 type, 3-5 days) were selected. Islets were isolated and purified from the pancreas of neonatal pigs. Ten non-diabetic Landrace pigs (1.5-2.0 months) served as recipients, and 12 000 IEQ/kg neonatal porcine islets were transplanted into the liver through the portal vein. All recipients received bolus injection of 50 U/kg of heparin 10 minutes before transplantation. After the bolus injection of heparin, the experimental group received heparin via the portal vein [10 U/(kg·h), 5 recipients], and the control group received heparin via the marginal ear vein [10 U/(kg·h), 5 recipients]. The superior vena cava blood was collected from the 2 groups pre-operation at 1, 3, 24 h post-operation of the transplantation. The portal vein blood was collected from the experimental group at 1 and 3 h after the transplantation as well. The levels of complement C3a, C5a, thrombin-antithrombin complex (TAT), β-thromboglobulin (β-TG), and D-dimer as well as activated partial thromboplastin time (APTT) in superior vena cava blood from 1 and 3 h post-transplantation were detected in the 2 groups, and the levels of anti-Xa and anti-IIa in the portal vein and superior vena cava blood from 1 and 3 h post-transplantation in the experimental group were detected. Twenty four hours after the transplantation, the liver tissues in the 2 groups were collected for pathological examination to observe the inflammatory cell infiltration and peripheral thrombosis around the islets graft in liver. RESULTS Before transplantation, there was no statistically significant difference in C3a, C5a, TAT, β-TG, D-dimer levels and APTT between the 2 groups (all P>0.05). At 1 and 3 h after transplantation, the C3a, TAT, and D-dimer levels in the experimental group were significant decreased than those in the control groups (all P<0.05), and at 3 h after transplantation the C5a was significant decreased than that in the control group (P<0.05). At 1 and 3 h after transplantation, the anti-Xa and anti-IIa levels in the portal vein blood were significantly increased than those in the superior vena cava blood in the experimental group (all P<0.05). Pathological results showed the presence of islet cell clusters in the liver blood vessels. The thrombus formation and neutrophil infiltration around islet graft was not obvious in the experimental group, while massive thrombus formation and neutrophil infiltration in the control group. CONCLUSIONS Compared with marginal ear vein infusion of heparin, the direct infusion of heparin in the portal vein has a certain inhibitory effect on complement system, coagulation system activation and inflammatory cell infiltration in portal vein islet transplantation, which may attenuate the occurrence of IBMIR.
Collapse
Affiliation(s)
- Shengwang Zhang
- Department of Radiology, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Haixiong Yan
- Department of Radiology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Xiaoqian Ma
- Department of Radiology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Wei Zheng
- Department of Radiology, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Wei Wang
- Department of Radiology, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| |
Collapse
|
30
|
Yamada K, Yoshida K. Multiple subcellular localizations and functions of protein kinase Cδ in liver cancer. World J Gastroenterol 2022; 28:188-198. [PMID: 35110944 PMCID: PMC8776529 DOI: 10.3748/wjg.v28.i2.188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/25/2021] [Accepted: 12/31/2021] [Indexed: 02/06/2023] Open
Abstract
Protein kinase Cδ (PKCδ) is a member of the PKC family, and its implications have been reported in various biological and cancerous processes, including cell proliferation, cell death, tumor suppression, and tumor progression. In liver cancer cells, accumulating reports show the bi-functional regulation of PKCδ in cell death and survival. PKCδ function is defined by various factors, such as phosphorylation, catalytic domain cleavage, and subcellular localization. PKCδ has multiple intracellular distribution patterns, ranging from the cytosol to the nucleus. We recently found a unique extracellular localization of PKCδ in liver cancer and its growth factor-like function in liver cancer cells. In this review, we first discuss the structural features of PKCδ and then focus on the functional diversity of PKCδ based on its subcellular localization, such as the nucleus, cell surface, and extracellular space. These findings improve our knowledge of PKCδ involvement in the progression of liver cancer.
Collapse
Affiliation(s)
- Kohji Yamada
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Kiyotsugu Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| |
Collapse
|
31
|
Cholenic acid derivative UniPR1331 impairs tumor angiogenesis via blockade of VEGF/VEGFR2 in addition to Eph/ephrin. Cancer Gene Ther 2022; 29:908-917. [PMID: 34426652 PMCID: PMC9293752 DOI: 10.1038/s41417-021-00379-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/12/2021] [Accepted: 08/10/2021] [Indexed: 12/11/2022]
Abstract
Angiogenesis, the formation of new blood vessels from preexisting ones, is crucial for tumor growth and metastatization, and is considered a promising therapeutic target. Unfortunately, drugs directed against a specific proangiogenic growth factor or receptor turned out to be of limited benefit for oncology patients, likely due to the high biochemical redundancy of the neovascularization process. In this scenario, multitarget compounds that are able to simultaneously tackle different proangiogenic pathways are eagerly awaited. UniPR1331 is a 3β-hydroxy-Δ5-cholenic acid derivative, which is already known to inhibit Eph-ephrin interaction. Here, we employed an analysis pipeline consisting of molecular modeling and simulation, surface plasmon resonance spectrometry, biochemical assays, and endothelial cell models to demonstrate that UniPR1331 directly interacts with the vascular endothelial growth factor receptor 2 (VEGFR2) too. The binding of UniPR1331 to VEGFR2 prevents its interaction with the natural ligand vascular endothelial growth factor and subsequent autophosphorylation, signal transduction, and in vitro proangiogenic activation of endothelial cells. In vivo, UniPR1331 inhibits tumor cell-driven angiogenesis in zebrafish. Taken together, these data shed light on the pleiotropic pharmacological effect of UniPR1331, and point to Δ5-cholenic acid as a promising molecular scaffold for the development of multitarget antiangiogenic compounds.
Collapse
|
32
|
Farooq M, Khan AW, Kim MS, Choi S. The Role of Fibroblast Growth Factor (FGF) Signaling in Tissue Repair and Regeneration. Cells 2021; 10:cells10113242. [PMID: 34831463 PMCID: PMC8622657 DOI: 10.3390/cells10113242] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/10/2021] [Accepted: 11/16/2021] [Indexed: 02/06/2023] Open
Abstract
Fibroblast growth factors (FGFs) are a large family of secretory molecules that act through tyrosine kinase receptors known as FGF receptors. They play crucial roles in a wide variety of cellular functions, including cell proliferation, survival, metabolism, morphogenesis, and differentiation, as well as in tissue repair and regeneration. The signaling pathways regulated by FGFs include RAS/mitogen-activated protein kinase (MAPK), phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)–protein kinase B (AKT), phospholipase C gamma (PLCγ), and signal transducer and activator of transcription (STAT). To date, 22 FGFs have been discovered, involved in different functions in the body. Several FGFs directly or indirectly interfere with repair during tissue regeneration, in addition to their critical functions in the maintenance of pluripotency and dedifferentiation of stem cells. In this review, we summarize the roles of FGFs in diverse cellular processes and shed light on the importance of FGF signaling in mechanisms of tissue repair and regeneration.
Collapse
Affiliation(s)
- Mariya Farooq
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (M.F.); (A.W.K.); (M.S.K.)
| | - Abdul Waheed Khan
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (M.F.); (A.W.K.); (M.S.K.)
| | - Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (M.F.); (A.W.K.); (M.S.K.)
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (M.F.); (A.W.K.); (M.S.K.)
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon 16502, Korea
- Correspondence:
| |
Collapse
|
33
|
Xie J, Wan J, Tang X, Li W, Peng B. Heparin modification improves the re-endothelialization and angiogenesis of decellularized kidney scaffolds through antithrombosis and anti-inflammation in vivo. Transl Androl Urol 2021; 10:3656-3668. [PMID: 34733661 PMCID: PMC8511541 DOI: 10.21037/tau-21-703] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/16/2021] [Indexed: 01/03/2023] Open
Abstract
Background Constructing tissue-engineered kidneys using decellularized kidney scaffolds (DKS) has attracted widespread attention as it is expected to be the key to solving the shortage of donor kidneys. However, thrombosis and the host inflammatory response are unfavorable factors that hider the re-endothelialization and vascularization of the decellularized scaffolds. Methods Heparin was immobilized into the DKS using the method of 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide/N-hydroxysuccinimide (EDC/NHS) activation. Fourier-transform infrared (FTIR) spectra were used to verify the heparinization of DKS. Human umbilical vein endothelial cells (HUVECs) were seeded and cultured in the DKS, then the sliced scaffolds were transplanted subcutaneously into nude mouse. Scanning electron microscopy and a series of histochemical stains including hematoxylin and eosin (H&E), elastic Verhöeff-Van Gieson (EVG), Sirius red, Masson’s trichrome, and toluidine blue (TB) staining were used for morphological characterization. The qRT-PCR analysis, immunohistochemistry (IHC), and immunofluorescence (IF) staining were used to determine the expression of related molecular markers. Results The rat DKS completely retained the extracellular matrix and heparinized modification. The H&E staining results showed there were more HUVECs covering the internal surfaces of tubular structures in the HEP-DKS group compared with the DKS group. The IF analysis results revealed that CD31, Ki67, and CD206 had higher positive rates in HUVECs in the HEP-DKS group compared to the DKS group. Both groups of scaffolds showed blood vessel formation via H&E staining, and there were more blood vessels in the HEP-DKS group compared with the native DKS group (P<0.05). The qRT-PCR results showed that the levels of IL-1β, IL-6, and TNF-α in the HEP-DKS group were significantly lower than those of the native DKS group, while the expression level of IL-10 was significantly higher than that in the native DKS group (P<0.05). Conclusions Heparin modification improves the re-endothelialization and vascular regeneration of the DKS through anticoagulation in vitro and in vivo. The anti-inflammatory effect of heparin on the transplanted host was initially confirmed, and it is considered that this effect may play a non-negligible role in promoting DKS re-endothelialization and angiogenesis. Heparinized DKS is therefore a promising candidate for kidney tissue engineering.
Collapse
Affiliation(s)
- Jinbo Xie
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jian Wan
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuemin Tang
- Department of Nephrology, Wuhu No. 1 People's Hospital, Wuhu, China
| | - Wei Li
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bo Peng
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
34
|
The Chemokine-Based Peptide, CXCL9(74-103), Inhibits Angiogenesis by Blocking Heparan Sulfate Proteoglycan-Mediated Signaling of Multiple Endothelial Growth Factors. Cancers (Basel) 2021; 13:cancers13205090. [PMID: 34680238 PMCID: PMC8534003 DOI: 10.3390/cancers13205090] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Major angiogenic growth factors activate downstream signaling cascades by interacting with both receptor tyrosine kinases (RTKs) and cell surface proteoglycans, such as heparan sulfate proteoglycans (HSPGs). As current anti-angiogenesis regimens in cancer are often faced with resistance, alternative therapeutic strategies are highly needed. The aim of our study was to investigate the impact on angiogenic signaling when we interfered with growth factor-HSPG interactions using a CXCL9 chemokine-derived peptide with high affinity for HS. Abstract Growth factors such as vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF) and epidermal growth factor (EGF) are important angiogenesis-mediating factors. They exert their effects not only through their respective receptor tyrosine kinases (RTKs), but they also require molecular pairing with heparan sulfate proteoglycans (HSPGs). Angiogenic growth factors and their signaling pathways are commonly targeted in current anti-angiogenic cancer therapies but have unfortunately insufficient impact on patient survival. Considering their obvious role in pathological angiogenesis, HS-targeting drugs have become an appealing new strategy. Therefore, we aimed to reduce angiogenesis through interference with growth factor-HS binding and downstream signaling using a CXCL9-derived peptide with a high affinity for glycosaminoglycans (GAGs), CXCL9(74-103). We showed that CXCL9(74-103) reduced EGF-, VEGF165- and FGF-2-mediated angiogenic processes in vitro, such as endothelial cell proliferation, chemotaxis, adhesion and sprouting, without exerting cell toxicity. CXCL9(74-103) interfered with growth factor signaling in diverse ways, e.g., by diminishing VEGF165 binding to HS and by direct association with FGF-2. The dependency of CXCL9(74-103) on HS for binding to HMVECs and for exerting its anti-angiogenic activity was also demonstrated. In vivo, CXCL9(74-103) attenuated neovascularization in the Matrigel plug assay, the corneal cauterization assay and in MDA-MB-231 breast cancer xenografts. Additionally, CXCL9(74-103) reduced vascular leakage in the retina of diabetic rats. In contrast, CXCL9(86-103), a peptide with low GAG affinity, showed no overall anti-angiogenic activity. Altogether, our results indicate that CXCL9(74-103) reduces angiogenesis by interfering with multiple HS-dependent growth factor signaling pathways.
Collapse
|
35
|
Methods for Assessing the Effects of Xylosides on Angiogenesis. Methods Mol Biol 2021. [PMID: 34626409 DOI: 10.1007/978-1-0716-1398-6_45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Xylosides are small synthetic molecules consisting of a xylose molecule attached to an aglycone group and serve as primers in the assembly of core protein free glycosaminoglycans using cellular machinery. Synthetic xylosides hold great promise in many biomedical applications and as therapeutics. Recent advances in the study of xylosides have opened up the possibility of developing xylosides as therapeutics to achieve a desirable biological outcome through their selective priming and inhibitory activities toward glycosaminoglycan biosynthesis. The approach described, herein, will serve as a general strategy to comprehensively screen xylosides and evaluate their ability to promote or inhibit angiogenesis, a critical biological process that is dysregulated in over 70 human diseases.
Collapse
|
36
|
Revuelta J, Fraile I, Monterrey DT, Peña N, Benito-Arenas R, Bastida A, Fernández-Mayoralas A, García-Junceda E. Heparanized chitosans: towards the third generation of chitinous biomaterials. MATERIALS HORIZONS 2021; 8:2596-2614. [PMID: 34617543 DOI: 10.1039/d1mh00728a] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The functionalization of chitosans is an emerging research area in the design of solutions for a wide range of biomedical applications. In particular, the modification of chitosans to incorporate sulfate groups has generated great interest since they show structural similarity to heparin and heparan sulfates. Most of the biomedical applications of heparan sulfates are derived from their ability to bind different growth factors and other proteins, as through these interactions they can modulate the cellular response. This review aims to summarize the most recent advances in the synthesis, and structural and physicochemical characterization of heparanized chitosan, a remarkably interesting family of polysaccharides that have demonstrated the ability to mimic heparan sulfates as ligands for different proteins, thereby exerting their biological activity by mimicking the function of these glycosaminoglycans.
Collapse
Affiliation(s)
- Julia Revuelta
- BioGlycoChem Group, Departamento de Química Bio-Orgánica, Instituto de Química Orgánica General, CSIC (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Isabel Fraile
- BioGlycoChem Group, Departamento de Química Bio-Orgánica, Instituto de Química Orgánica General, CSIC (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Dianelis T Monterrey
- BioGlycoChem Group, Departamento de Química Bio-Orgánica, Instituto de Química Orgánica General, CSIC (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Nerea Peña
- BioGlycoChem Group, Departamento de Química Bio-Orgánica, Instituto de Química Orgánica General, CSIC (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Raúl Benito-Arenas
- BioGlycoChem Group, Departamento de Química Bio-Orgánica, Instituto de Química Orgánica General, CSIC (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Agatha Bastida
- BioGlycoChem Group, Departamento de Química Bio-Orgánica, Instituto de Química Orgánica General, CSIC (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Alfonso Fernández-Mayoralas
- BioGlycoChem Group, Departamento de Química Bio-Orgánica, Instituto de Química Orgánica General, CSIC (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Eduardo García-Junceda
- BioGlycoChem Group, Departamento de Química Bio-Orgánica, Instituto de Química Orgánica General, CSIC (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain.
| |
Collapse
|
37
|
Rattila S, Kleefeldt F, Ballesteros A, Beltrame JS, L Ribeiro M, Ergün S, Dveksler G. Pro-angiogenic effects of pregnancy-specific glycoproteins in endothelial and extravillous trophoblast cells. Reproduction 2021; 160:737-750. [PMID: 33065549 DOI: 10.1530/rep-20-0169] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 08/20/2020] [Indexed: 01/23/2023]
Abstract
We previously reported that binding to heparan sulfate (HS) is required for the ability of the placentally secreted pregnancy-specific glycoprotein 1 (PSG1) to induce endothelial tubulogenesis. PSG1 is composed of four immunoglobulin-like domains but which domains of the protein bind to HS remains unknown. To analyze the interaction of PSG1 with HS, we generated several recombinant proteins, including the individual domains, chimeric proteins between two PSG1 domains, and mutants. Using flow cytometric and surface plasmon resonance studies, we determined that the B2 domain of PSG1 binds to HS and that the positively charged amino acids encompassed between amino acids 43-59 are required for this interaction. Furthermore, we showed that the B2 domain of PSG1 is required for the increase in the formation of tubes by endothelial cells (EC) including a human endometrial EC line and two extravillous trophoblast (EVT) cell lines and for the pro-angiogenic activity of PSG1 observed in an aortic ring assay. PSG1 enhanced the migration of ECs while it increased the expression of matrix metalloproteinase-2 in EVTs, indicating that the pro-angiogenic effect of PSG1 on these two cell types may be mediated by different mechanisms. Despite differences in amino acid sequence, we observed that all human PSGs bound to HS proteoglycans and confirmed that at least two other members of the family, PSG6 and PSG9, induce tube formation. These findings contribute to a better understanding of the pro-angiogenic activity of human PSGs and strongly suggest conservation of this function among all PSG family members.
Collapse
Affiliation(s)
- Shemona Rattila
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | - Florian Kleefeldt
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Angela Ballesteros
- Molecular Physiology and Biophysics Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Jimena S Beltrame
- Laboratory of Physiology and Pharmacology of Reproduction, Centre for Pharmacological and Botanical Studies (CONICET - School of Medicine, University of Buenos Aires), Buenos Aires, Argentina
| | - Maria L Ribeiro
- Laboratory of Physiology and Pharmacology of Reproduction, Centre for Pharmacological and Botanical Studies (CONICET - School of Medicine, University of Buenos Aires), Buenos Aires, Argentina
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Gabriela Dveksler
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
38
|
Paiva REF, Peterson EJ, Malina J, Zoepfl M, Hampton JD, Johnson WE, Graminha A, Ourahmane A, McVoy MA, Brabec V, Berners‐Price SJ, Farrell NP. On the Biology of Werner's Complex. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202105019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Raphael E. F. Paiva
- Institute for Glycomics Griffith University Gold Coast Campus Southport Qld. 4222 Australia
| | - Erica J. Peterson
- Department of Chemistry Virginia Commonwealth University Richmond VA 23284-2006 USA
- Massey Cancer Center Virginia Commonwealth University Richmond VA 23298-0037 USA
| | - Jaroslav Malina
- Institute of Biophysics Czech Academy of Sciences Kralovopolska 135 61265 Brno Czech Republic
| | - Mary Zoepfl
- Department of Chemistry Virginia Commonwealth University Richmond VA 23284-2006 USA
| | - J. David Hampton
- Massey Cancer Center Virginia Commonwealth University Richmond VA 23298-0037 USA
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University Richmond Virginia 23298-0033 USA
| | - Wyatt E. Johnson
- Department of Chemistry Virginia Commonwealth University Richmond VA 23284-2006 USA
| | - Angelica Graminha
- Department of Chemistry Virginia Commonwealth University Richmond VA 23284-2006 USA
| | - Amine Ourahmane
- Department of Pediatrics Virginia Commonwealth University Richmond VA 23298-0163 USA
| | - Michael A. McVoy
- Department of Pediatrics Virginia Commonwealth University Richmond VA 23298-0163 USA
| | - Viktor Brabec
- Institute of Biophysics Czech Academy of Sciences Kralovopolska 135 61265 Brno Czech Republic
| | - Susan J. Berners‐Price
- Institute for Glycomics Griffith University Gold Coast Campus Southport Qld. 4222 Australia
| | - Nicholas P. Farrell
- Institute for Glycomics Griffith University Gold Coast Campus Southport Qld. 4222 Australia
- Department of Chemistry Virginia Commonwealth University Richmond VA 23284-2006 USA
- Massey Cancer Center Virginia Commonwealth University Richmond VA 23298-0037 USA
| |
Collapse
|
39
|
de Paiva REF, Peterson EJ, Malina J, Zoepfl M, Hampton JD, Johnson WE, Graminha A, Ourahmane A, McVoy MA, Brabec V, Berners-Price SJ, Farrell NP. On the Biology of Werner's Complex. Angew Chem Int Ed Engl 2021; 60:17123-17130. [PMID: 34105220 PMCID: PMC8464317 DOI: 10.1002/anie.202105019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/30/2021] [Indexed: 11/05/2022]
Abstract
Werner's Complex, as a cationic coordination complex (CCC), has hitherto unappreciated biological properties derived from its binding affinity to highly anionic biomolecules such as glycosaminoglycans (GAGs) and nucleic acids. Competitive inhibitor and spectroscopic assays confirm the high affinity to GAGs heparin, heparan sulfate (HS), and its pentasaccharide mimetic Fondaparinux (FPX). Functional consequences of this affinity include inhibition of FPX cleavage by bacterial heparinase and mammalian heparanase enzymes with inhibition of cellular invasion and migration. Werner's Complex is a very efficient condensing agent for DNA and tRNA. In proof-of-principle for translational implications, it is demonstrated to display antiviral activity against human cytomegalovirus (HCMV) at micromolar concentrations with promising selectivity. Exploitation of non-covalent hydrogen-bonding and electrostatic interactions has motivated the unprecedented discovery of these properties, opening new avenues of research for this iconic compound.
Collapse
Affiliation(s)
- Raphael E F de Paiva
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, Qld., 4222, Australia
| | - Erica J Peterson
- Department of Chemistry, Virginia Commonwealth University, Richmond, VA, 23284-2006, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298-0037, USA
| | - Jaroslav Malina
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, 61265, Brno, Czech Republic
| | - Mary Zoepfl
- Department of Chemistry, Virginia Commonwealth University, Richmond, VA, 23284-2006, USA
| | - J David Hampton
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298-0037, USA
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, 23298-0033, USA
| | - Wyatt E Johnson
- Department of Chemistry, Virginia Commonwealth University, Richmond, VA, 23284-2006, USA
| | - Angelica Graminha
- Department of Chemistry, Virginia Commonwealth University, Richmond, VA, 23284-2006, USA
| | - Amine Ourahmane
- Department of Pediatrics, Virginia Commonwealth University, Richmond, VA, 23298-0163, USA
| | - Michael A McVoy
- Department of Pediatrics, Virginia Commonwealth University, Richmond, VA, 23298-0163, USA
| | - Viktor Brabec
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, 61265, Brno, Czech Republic
| | - Susan J Berners-Price
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, Qld., 4222, Australia
| | - Nicholas P Farrell
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, Qld., 4222, Australia
- Department of Chemistry, Virginia Commonwealth University, Richmond, VA, 23284-2006, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298-0037, USA
| |
Collapse
|
40
|
Genua M, Garçon LA, Sergeeva YN, Saesen E, Musnier B, Buhot A, Billon M, Gout E, Sadir R, Lortat-Jacob H, Le Narvor C, Bonnaffé D, Livache T, Hou Y. Discrimination of deletion to point cytokine mutants based on an array of cross-reactive receptors mimicking protein recognition by heparan sulfate. Anal Bioanal Chem 2021; 414:551-559. [PMID: 34258651 DOI: 10.1007/s00216-021-03516-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/15/2021] [Accepted: 06/29/2021] [Indexed: 10/20/2022]
Abstract
Differential sensing of proteins based on cross-reactive arrays and pattern recognition is a promising technique for the detection and identification of proteins. In this study, a rational biomimetic strategy has been used to prepare sensing materials capable of discriminating structurally similar proteins, such as deletion and point mutants of a cytokine, by mimicking the biological properties of heparan sulfate (HS). Using the self-assembly of two disaccharides, lactose and sulfated lactose at various ratios on the surface of a chip, an array of combinatorial cross-reactive receptors has been prepared. Coupling with surface plasmon resonance imaging (SPRi), the obtained cross-reactive array is very efficient for protein sensing. It is able to detect HS binding proteins (HSbps) such as IFNγ at nanomolar concentrations. Moreover, such a system is capable of discriminating between IFNγ and its mutants with good selectivity.
Collapse
Affiliation(s)
- Maria Genua
- Université Grenoble Alpes, CEA, CNRS, IRIG-SyMMES, 38000, Grenoble, France
| | | | - Yulia N Sergeeva
- Université Grenoble Alpes, CEA, CNRS, IRIG-SyMMES, 38000, Grenoble, France
| | - Els Saesen
- Institut de Biologie Structurale, UMR 5075, Université Grenoble Alpes, CNRS, CEA, 38000, Grenoble, France
| | - Benjamin Musnier
- Université Grenoble Alpes, CEA, CNRS, IRIG-SyMMES, 38000, Grenoble, France
| | - Arnaud Buhot
- Université Grenoble Alpes, CEA, CNRS, IRIG-SyMMES, 38000, Grenoble, France
| | - Martial Billon
- Université Grenoble Alpes, CEA, CNRS, IRIG-SyMMES, 38000, Grenoble, France
| | - Evelyne Gout
- Institut de Biologie Structurale, UMR 5075, Université Grenoble Alpes, CNRS, CEA, 38000, Grenoble, France
| | - Rabia Sadir
- Institut de Biologie Structurale, UMR 5075, Université Grenoble Alpes, CNRS, CEA, 38000, Grenoble, France
| | - Hugues Lortat-Jacob
- Institut de Biologie Structurale, UMR 5075, Université Grenoble Alpes, CNRS, CEA, 38000, Grenoble, France
| | - Christine Le Narvor
- Université Paris-Saclay, CNRS, Institut de chimie moléculaire et des matériaux d'Orsay, 91405, Orsay, France
| | - David Bonnaffé
- Université Paris-Saclay, CNRS, Institut de chimie moléculaire et des matériaux d'Orsay, 91405, Orsay, France
| | - Thierry Livache
- Université Grenoble Alpes, CEA, CNRS, IRIG-SyMMES, 38000, Grenoble, France
| | - Yanxia Hou
- Université Grenoble Alpes, CEA, CNRS, IRIG-SyMMES, 38000, Grenoble, France.
| |
Collapse
|
41
|
A Bittersweet Computational Journey among Glycosaminoglycans. Biomolecules 2021; 11:biom11050739. [PMID: 34063530 PMCID: PMC8156566 DOI: 10.3390/biom11050739] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 01/22/2023] Open
Abstract
Glycosaminoglycans (GAGs) are linear polysaccharides. In proteoglycans (PGs), they are attached to a core protein. GAGs and PGs can be found as free molecules, associated with the extracellular matrix or expressed on the cell membrane. They play a role in the regulation of a wide array of physiological and pathological processes by binding to different proteins, thus modulating their structure and function, and their concentration and availability in the microenvironment. Unfortunately, the enormous structural diversity of GAGs/PGs has hampered the development of dedicated analytical technologies and experimental models. Similarly, computational approaches (in particular, molecular modeling, docking and dynamics simulations) have not been fully exploited in glycobiology, despite their potential to demystify the complexity of GAGs/PGs at a structural and functional level. Here, we review the state-of-the art of computational approaches to studying GAGs/PGs with the aim of pointing out the “bitter” and “sweet” aspects of this field of research. Furthermore, we attempt to bridge the gap between bioinformatics and glycobiology, which have so far been kept apart by conceptual and technical differences. For this purpose, we provide computational scientists and glycobiologists with the fundamentals of these two fields of research, with the aim of creating opportunities for their combined exploitation, and thereby contributing to a substantial improvement in scientific knowledge.
Collapse
|
42
|
Litvinova LS, Yurova KA, Khaziakhmatova OG, Khlusova MY, Malashchenko VV, Shunkin EO, Todosenko NM, Norkin IK, Ivanov PA, Khlusov IA. Osteogenic and Angiogenic Properties of Heparin as a System for Delivery of Biomolecules for Bone Bioengineering: a Brief Critical Review. BIOCHEMISTRY MOSCOW-SUPPLEMENT SERIES B-BIOMEDICAL CHEMISTRY 2021; 15:147-152. [PMID: 34007413 PMCID: PMC8120488 DOI: 10.1134/s1990750821020050] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/16/2020] [Accepted: 10/29/2020] [Indexed: 01/04/2023]
Abstract
The review considers complex, controversial, and individual effects of heparin and its derivatives on the bone and circulatory systems in dependence of the dose, the state of the cells and tissues of the recipient. General data on the anticoagulant activity of heparin and its derivatives are presented; special attention is paid to the effect of heparin on mesenchymal cells and tissues and its role in angiogenesis. We also discuss the ability of heparin to bind osteogenic and angiogenic biomolecules in the context of the development of systems for their delivery and sustained controlled release and propose a schematic representation of the positive and side effects of heparin as a delivery system for biomolecules in tissue engineering.
Collapse
Affiliation(s)
- L S Litvinova
- Center for Immunology and Cellular Biotechnology of the Immanuel Kant Baltic Federal University, ul. Gaidarа 6, 236041 Kaliningrad, Russia
| | - K A Yurova
- Center for Immunology and Cellular Biotechnology of the Immanuel Kant Baltic Federal University, ul. Gaidarа 6, 236041 Kaliningrad, Russia
| | - O G Khaziakhmatova
- Center for Immunology and Cellular Biotechnology of the Immanuel Kant Baltic Federal University, ul. Gaidarа 6, 236041 Kaliningrad, Russia
| | - M Yu Khlusova
- Division of Pathophysiology, Siberian State Medical University, ul. Uchebnaya 39, 634050 Tomsk, Russia
| | - V V Malashchenko
- Center for Immunology and Cellular Biotechnology of the Immanuel Kant Baltic Federal University, ul. Gaidarа 6, 236041 Kaliningrad, Russia
| | - E O Shunkin
- Center for Immunology and Cellular Biotechnology of the Immanuel Kant Baltic Federal University, ul. Gaidarа 6, 236041 Kaliningrad, Russia
| | - N M Todosenko
- Center for Immunology and Cellular Biotechnology of the Immanuel Kant Baltic Federal University, ul. Gaidarа 6, 236041 Kaliningrad, Russia
| | - I K Norkin
- Center for Immunology and Cellular Biotechnology of the Immanuel Kant Baltic Federal University, ul. Gaidarа 6, 236041 Kaliningrad, Russia
| | - P A Ivanov
- Center for Immunology and Cellular Biotechnology of the Immanuel Kant Baltic Federal University, ul. Gaidarа 6, 236041 Kaliningrad, Russia
| | - I A Khlusov
- Department of Morphology and General Pathology, Siberian State Medical University, Moskovsky Trakt 2, bldg. 7, 634050 Tomsk, Russia.,Research School of Chemistry and Applied Biomedical Sciences National Research Tomsk Polytechnic University, ul. Lenina 43-A, 634034 Tomsk, Russia
| |
Collapse
|
43
|
Lisztes E, Mező E, Demeter F, Horváth L, Bősze S, István Tóth B, Borbás A, Herczeg M. Synthesis and Cell Growth Inhibitory Activity of Six Non-glycosaminoglycan-Type Heparin-Analogue Trisaccharides. ChemMedChem 2021; 16:1467-1476. [PMID: 33433040 PMCID: PMC8247843 DOI: 10.1002/cmdc.202000917] [Citation(s) in RCA: 4] [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: 11/24/2020] [Revised: 01/11/2021] [Indexed: 12/27/2022]
Abstract
The design and synthesis of heparin mimetics with high anticancer activity but no anticoagulant activity is an important task in medicinal chemistry. Herein, we present the efficient synthesis of five Glc-GlcA-Glc-sequenced and one Glc-IdoA-Glc-sequenced non-glycosaminoglycan, heparin-related trisaccharides with various sulfation/sulfonylation and methylation patterns. The cell growth inhibitory effects of the compounds were tested against four cancerous human cell lines and two non-cancerous cell lines. Two d-glucuronate-containing tetra-O-sulfated, partially methylated trisaccharides displayed remarkable and selective inhibitory effects on the growth of ovary carcinoma (A2780) and melanoma (WM35) cells. Methyl substituents on the glucuronide unit proved to be detrimental, whereas acetyl substituents were beneficial to the cytostatic activity of the sulfated derivatives.
Collapse
Affiliation(s)
- Erika Lisztes
- Department of PhysiologyUniversity of Debrecen PO Box 224012DebrecenHungary
| | - Erika Mező
- Department of Pharmaceutical ChemistryUniversity of DebrecenEgyetem tér 14032DebrecenHungary
| | - Fruzsina Demeter
- Department of Pharmaceutical ChemistryUniversity of DebrecenEgyetem tér 14032DebrecenHungary
- Doctoral School of ChemistryUniversity of DebrecenEgyetem tér 14032DebrecenHungary
- MTA-DE Molecular Recognition and Interaction Research Group, ELKHUniversity of DebrecenEgyetem tér 14032DebrecenHungary
| | - Lilla Horváth
- MTA-ELTE Research Group of Peptide ChemistryEötvös Loránd UniversityPázmány Péter sétány 1/a1117BudapestHungary
| | - Szilvia Bősze
- MTA-ELTE Research Group of Peptide ChemistryEötvös Loránd UniversityPázmány Péter sétány 1/a1117BudapestHungary
| | - Balázs István Tóth
- Department of PhysiologyUniversity of Debrecen PO Box 224012DebrecenHungary
| | - Anikó Borbás
- Department of Pharmaceutical ChemistryUniversity of DebrecenEgyetem tér 14032DebrecenHungary
| | - Mihály Herczeg
- Department of Pharmaceutical ChemistryUniversity of DebrecenEgyetem tér 14032DebrecenHungary
- MTA-DE Research Group for Oligosaccharide Chemistry, ELKHEgyetem tér 14032DebrecenHungary
| |
Collapse
|
44
|
Maszota-Zieleniak M, Marcisz M, Kogut MM, Siebenmorgen T, Zacharias M, Samsonov SA. Evaluation of replica exchange with repulsive scaling approach for docking glycosaminoglycans. J Comput Chem 2021; 42:1040-1053. [PMID: 33768554 DOI: 10.1002/jcc.26496] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/19/2021] [Accepted: 01/27/2021] [Indexed: 12/14/2022]
Abstract
Glycosaminoglycans (GAGs), long linear periodic anionic polysaccharides, are key molecules in the extracellular matrix (ECM). Therefore, deciphering their role in the biologically relevant context is important for fundamental understanding of the processes ongoing in ECM and for establishing new strategies in the regenerative medicine. Although GAGs represent a number of computational challenges, molecular docking is a powerful tool for analysis of their interactions. Despite the recent development of GAG-specific docking approaches, there is plenty of room for improvement. Here, replica exchange molecular dynamics with repulsive scaling (REMD-RS) recently proved to be a successful approach for protein-protein complexes, was applied to dock GAGs. In this method, effective pairwise radii are increased in different Hamiltonian replicas. REMD-RS is shown to be an attractive alternative to classical docking approaches for GAGs. This work contributes to setting up of GAG-specific computational protocols and provides new insights into the nature of these biological systems.
Collapse
Affiliation(s)
| | | | | | - Till Siebenmorgen
- Physics Department, Technical University of Munich, Garching, Germany
| | - Martin Zacharias
- Physics Department, Technical University of Munich, Garching, Germany
| | | |
Collapse
|
45
|
Sterling JD, Jiang W, Botello-Smith WM, Luo YL. Ion Pairing and Dielectric Decrement in Glycosaminoglycan Brushes. J Phys Chem B 2021; 125:2771-2780. [PMID: 33662212 DOI: 10.1021/acs.jpcb.0c11571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cell-surface polysaccharides are essential to many aspects of physiology, serving as a highly conserved evolutionary feature of life and as an important part of the innate immune system in mammals. Here, as simplified biophysical models of these sugar coatings, we present results of molecular dynamics simulations of hyaluronic acid and heparin brushes that show important effects of ion pairing, water dielectric decrease, and coion exclusion. As in prior studies of macromolecular crowding under physiologically relevant salt concentrations, our results show equilibria with electroneutrality attained through screening and pairing of brush anionic charges by monovalent cations at the atomistic detail. Most surprising is the reversal of the Donnan potential obtained from both nonpolarizable and Drude polarizable force fields, in contrast to what would be expected based on electrostatic Boltzmann partitioning alone. Water dielectric decrement within the brush domain is also associated with Born hydration-driven cation exclusion from the brush. We observe that the primary partition energy attracting cations to attain brush electroneutrality is the ion pairing or salt-bridge energy. Potassium and sodium pairings to glycosaminoglycan carboxylates and sulfates show similar abundance of contact-pairing and solvent-separated pairing. We conclude that in these crowded macromolecular brushes, ion-pairing, Born-hydration, and electrostatic potential energies all contribute to attain electroneutrality and should therefore contribute in mean-field models to accurately represent brush electrostatics.
Collapse
Affiliation(s)
- James D Sterling
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, 535 Watson Dr., Claremont, California 91711, United States
| | - Wenjuan Jiang
- College of Pharmacy, Western University of Health Sciences, 309 E. Second Street, Pomona, California 91766, United States
| | - Wesley M Botello-Smith
- College of Pharmacy, Western University of Health Sciences, 309 E. Second Street, Pomona, California 91766, United States
| | - Yun L Luo
- College of Pharmacy, Western University of Health Sciences, 309 E. Second Street, Pomona, California 91766, United States
| |
Collapse
|
46
|
Gorle AK, Haselhorst T, Katner SJ, Everest-Dass AV, Hampton JD, Peterson EJ, Koblinski JE, Katsuta E, Takabe K, von Itzstein M, Berners-Price SJ, Farrell NP. Conformational Modulation of Iduronic Acid-Containing Sulfated Glycosaminoglycans by a Polynuclear Platinum Compound and Implications for Development of Antimetastatic Platinum Drugs. Angew Chem Int Ed Engl 2021; 60:3283-3289. [PMID: 33174390 PMCID: PMC7902481 DOI: 10.1002/anie.202013749] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Indexed: 12/19/2022]
Abstract
1 H NMR spectroscopic studies on the 1:1 adduct of the pentasaccharide Fondaparinux (FPX) and the substitution-inert polynuclear platinum complex TriplatinNC show significant modulation of geometry around the glycosidic linkages of the FPX constituent monosaccharides. FPX is a valid model for the highly sulfated cell signalling molecule heparan sulfate (HS). The conformational ratio of the 1 C4 :2 S0 forms of the FPX residue IdoA(2S) is altered from ca. 35:65 (free FPX) to ca. 75:25 in the adduct; the first demonstration of a small molecule affecting conformational changes on a HS oligosaccharide. Functional consequences of such binding are suggested to be inhibition of HS cleavage in MDA-MB-231 triple-negative breast cancer (TNBC) cells. We further describe inhibition of metastasis by TriplatinNC in the TNBC 4T1 syngeneic tumour model. Our work provides insight into a novel approach for design of platinum drugs (and coordination compounds in general) with intrinsic anti-metastatic potential.
Collapse
Affiliation(s)
- Anil K. Gorle
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland, 4222, Australia
| | - Thomas Haselhorst
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland, 4222, Australia
| | - Samantha J. Katner
- Department of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284-2006, USA
- Department of Biochemistry, Chemistry and Geology, Minnesota State University, Mankato, Mankato, Minnesota 56001, USA
| | - Arun V. Everest-Dass
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland, 4222, Australia
| | - James D. Hampton
- Department of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284-2006, USA
| | - Erica J. Peterson
- Department of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284-2006, USA
| | - Jennifer E. Koblinski
- Department of Pathology, Division of Cellular and Molecular Pathogenesis, Virginia Commonwealth University, Richmond, Virginia 23284-2006, USA
| | - Eriko Katsuta
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, University at Buffalo, Buffalo, New York, 14203, USA
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, University at Buffalo, Buffalo, New York, 14203, USA
| | - Mark von Itzstein
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland, 4222, Australia
| | - Susan J. Berners-Price
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland, 4222, Australia
| | - Nicholas P. Farrell
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, Queensland, 4222, Australia
- Department of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284-2006, USA
| |
Collapse
|
47
|
Litvinova LS, Yurova KA, Khaziakhmatova OG, Khlusova MY, Malashchenko VV, Shunkin EO, Todosenko NM, Norkin IK, Ivanov PA, Khlusov IA. [Osteogenic and angiogenic properties of heparin as a system of biomolecule delivery for bone bioengineering: a brief critical review]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2021; 66:431-436. [PMID: 33372899 DOI: 10.18097/pbmc20206606431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The review discusses the complex, ambiguous and individual effects of heparin and its derivatives on the bone and circulatory systems, in dependence of the dosage, the state of the cells and tissues of recipients. General data on the anticoagulant activity of heparin and its derivatives are presented; aspects of the effect of heparin on mesenchymal cells and tissues and its role in angiogenesis are considered in details. Particular attention is paid to the ability of heparin to bind osteogenic and angiogenic biomolecules: thus us especially important for the development of systems for their delivery and sustained controlled release. A schematic representation of the positive and side effects of heparin as a delivery system for biomolecules in tissue engineering is proposed.
Collapse
Affiliation(s)
- L S Litvinova
- Center for Immunology and Cellular Biotechnology of the Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - K A Yurova
- Center for Immunology and Cellular Biotechnology of the Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - O G Khaziakhmatova
- Center for Immunology and Cellular Biotechnology of the Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - M Yu Khlusova
- Division of Pathophysiology Siberian State Medical University, Tomsk, Russia
| | - V V Malashchenko
- Center for Immunology and Cellular Biotechnology of the Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - E O Shunkin
- Center for Immunology and Cellular Biotechnology of the Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - N M Todosenko
- Center for Immunology and Cellular Biotechnology of the Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - I K Norkin
- Center for Immunology and Cellular Biotechnology of the Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - P A Ivanov
- Center for Immunology and Cellular Biotechnology of the Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - I A Khlusov
- Department of Morphology and General Pathology Siberian State Medical University, Tomsk, Russia; Research School of Chemistry and Applied Biomedical Sciences National Research Tomsk Polytechnic University, Tomsk, Russia
| |
Collapse
|
48
|
Oussoren E, Wagenmakers MAEM, Link B, van der Meijden JC, Pijnappel WWMP, Ruijter GJG, Langeveld M, van der Ploeg AT. Hip disease in Mucopolysaccharidoses and Mucolipidoses: A review of mechanisms, interventions and future perspectives. Bone 2021; 143:115729. [PMID: 33130340 DOI: 10.1016/j.bone.2020.115729] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 11/20/2022]
Abstract
The hips are frequently involved in inheritable diseases which affect the bones. The clinical and radiological presentation of these diseases may be very similar to common hip disorders as developmental dysplasia of the hip, osteoarthritis and avascular necrosis, so the diagnosis may be easily overlooked and treatment may be suboptimal. Mucopolysaccharidosis (MPS) and Mucolipidosis (ML II and III) are lysosomal storage disorders with multisystemic involvement. Characteristic skeletal abnormalities, known as dysostosis multiplex, are common in MPS and ML and originate from intra-lysosomal storage of glycosaminoglycans in cells of the cartilage, bones and ligaments. The hip joint is severely affected in MPS and ML. Hip pathology results in limitations in mobility and pain from young age, and negatively affects quality of life. In order to better understand the underlying process that causes hip disease in MPS and ML, this review first describes the normal physiological (embryonic) hip joint development, including the interplay between the acetabulum and the femoral head. In the second part the factors contributing to altered hip morphology and function in MPS and ML are discussed, such as abnormal development of the pelvic- and femoral bones (which results in altered biomechanical forces) and inflammation. In the last part of this review therapeutic options and future perspectives are addressed.
Collapse
Affiliation(s)
- Esmee Oussoren
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Margreet A E M Wagenmakers
- Department of Internal Medicine, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Bianca Link
- Division of Metabolism, Connective Tissue Unit, University Children's Hospital Zurich, Zurich, Switzerland.
| | - Jan C van der Meijden
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - W W M Pim Pijnappel
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Clinical Genetics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - George J G Ruijter
- Department of Clinical Genetics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Mirjam Langeveld
- Department of Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.
| | - Ans T van der Ploeg
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Clinical Genetics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
49
|
Yamada K, Oikawa T, Kizawa R, Motohashi S, Yoshida S, Kumamoto T, Saeki C, Nakagawa C, Shimoyama Y, Aoki K, Tachibana T, Saruta M, Ono M, Yoshida K. Unconventional Secretion of PKCδ Exerts Tumorigenic Function via Stimulation of ERK1/2 Signaling in Liver Cancer. Cancer Res 2020; 81:414-425. [PMID: 33318039 DOI: 10.1158/0008-5472.can-20-2009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/02/2020] [Accepted: 11/18/2020] [Indexed: 11/16/2022]
Abstract
Expression of human protein kinase C delta (PKCδ) protein has been linked to many types of cancers. PKCδ is known to be a multifunctional PKC family member and has been rigorously studied as an intracellular signaling molecule. Here we show that PKCδ is a secretory protein that regulates cell growth of liver cancer. Full-length PKCδ was secreted to the extracellular space in living liver cancer cells under normal cell culture conditions and in xenograft mouse models. Patients with liver cancer showed higher levels of serum PKCδ than patients with chronic hepatitis or liver cirrhosis or healthy individuals. In liver cancer cells, PKCδ secretion was executed in an endoplasmic reticulum (ER)-Golgi-independent manner, and the inactivation status of cytosolic PKCδ was required for its secretion. Furthermore, colocalization studies showed that extracellular PKCδ was anchored on the cell surface of liver cancer cells via association with glypican 3, a liver cancer-related heparan sulfate proteoglycan. Addition of exogenous PKCδ activated IGF-1 receptor (IGF1R) activation and subsequently enhanced activation of ERK1/2, which led to accelerated cell growth in liver cancer cells. Conversely, treatment with anti-PKCδ antibody attenuated activation of both IGF1R and ERK1/2 and reduced cell proliferation and spheroid formation of liver cancer cells and tumor growth in xenograft mouse models. This study demonstrates the presence of PKCδ at the extracellular space and the function of PKCδ as a growth factor and provides a rationale for the extracellular PKCδ-targeting therapy of liver cancer. SIGNIFICANCE: PKCδ secretion from liver cancer cells behaves as a humoral growth factor that contributes to cell growth via activation of proliferative signaling molecules, which may be potential diagnostic or therapeutic targets.
Collapse
Affiliation(s)
- Kohji Yamada
- Department of Biochemistry, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Tsunekazu Oikawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Ryusuke Kizawa
- Department of Biochemistry, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Saya Motohashi
- Department of Biochemistry, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Saishu Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Tomotaka Kumamoto
- Department of Biochemistry, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Chisato Saeki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Chika Nakagawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Yuya Shimoyama
- Department of Biochemistry, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Katsuhiko Aoki
- Department of Biochemistry, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Toshiaki Tachibana
- Core Research Facilities for Basic Science, Research Center for Medical Sciences, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Masayuki Saruta
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Masaya Ono
- Department of Clinical Proteomics, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Kiyotsugu Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan.
| |
Collapse
|
50
|
Gorle AK, Haselhorst T, Katner SJ, Everest‐Dass AV, Hampton JD, Peterson EJ, Koblinski JE, Katsuta E, Takabe K, Itzstein M, Berners‐Price SJ, Farrell NP. Conformational Modulation of Iduronic Acid‐Containing Sulfated Glycosaminoglycans by a Polynuclear Platinum Compound and Implications for Development of Antimetastatic Platinum Drugs. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202013749] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Anil K. Gorle
- Institute for Glycomics Griffith University Gold Coast Campus Southport Queensland 4222 Australia
| | - Thomas Haselhorst
- Institute for Glycomics Griffith University Gold Coast Campus Southport Queensland 4222 Australia
| | - Samantha J. Katner
- Department of Chemistry Virginia Commonwealth University Richmond VA 23284-2006 USA
- Department of Biochemistry, Chemistry and Geology Minnesota State University Mankato, Mankato MN 56001 USA
| | - Arun V. Everest‐Dass
- Institute for Glycomics Griffith University Gold Coast Campus Southport Queensland 4222 Australia
| | - James D. Hampton
- Department of Chemistry Virginia Commonwealth University Richmond VA 23284-2006 USA
- Massey Cancer Center Virginia Commonwealth University Richmond VA 23298-0037 USA
| | - Erica J. Peterson
- Department of Chemistry Virginia Commonwealth University Richmond VA 23284-2006 USA
- Massey Cancer Center Virginia Commonwealth University Richmond VA 23298-0037 USA
| | - Jennifer E. Koblinski
- Massey Cancer Center Virginia Commonwealth University Richmond VA 23298-0037 USA
- Department of Pathology Division of Cellular and Molecular Pathogenesis Virginia Commonwealth University Richmond VA 23284-2006 USA
| | - Eriko Katsuta
- Department of Surgical Oncology Roswell Park Comprehensive Cancer Center University at Buffalo Buffalo NY 14203 USA
| | - Kazuaki Takabe
- Department of Surgical Oncology Roswell Park Comprehensive Cancer Center University at Buffalo Buffalo NY 14203 USA
| | - Mark Itzstein
- Institute for Glycomics Griffith University Gold Coast Campus Southport Queensland 4222 Australia
| | - Susan J. Berners‐Price
- Institute for Glycomics Griffith University Gold Coast Campus Southport Queensland 4222 Australia
| | - Nicholas P. Farrell
- Institute for Glycomics Griffith University Gold Coast Campus Southport Queensland 4222 Australia
- Department of Chemistry Virginia Commonwealth University Richmond VA 23284-2006 USA
- Massey Cancer Center Virginia Commonwealth University Richmond VA 23298-0037 USA
| |
Collapse
|