1
|
Ribeiro B, Castro AL, Pilão S, Ribeiro C, Barbosa MA, Santos SG, Gonçalves RM. Enhanced collagen deposition in chondrogenic inflammation with Ibuprofen-specifically loaded chitosan/poly-gamma-glutamic acid nanoparticles. Int J Pharm 2025; 676:125591. [PMID: 40228612 DOI: 10.1016/j.ijpharm.2025.125591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Osteoarthritis (OA) is characterized by degeneration of articular cartilage, related to a pro-inflammatory environment. Different anti-inflammatory drugs have been proposed to treat OA symptoms, namely those from the group of non-steroidal anti-inflammatory drugs (NSAID), but they carry several drawbacks and are not able to repair/regenerate cartilage tissues. In this study, Chitosan (Ch) and Poly-Glutamic acid (PGA) were combined with Ibuprofen, a NSAID widely used in OA, for drug delivery. Ibuprofen-Ch/PGA nanoparticles (Ibuf-NPs) were produced by co-acervation method, and ability to control inflammation and promote chondrogenesis under a pro-inflammatory setting, was evaluated in vitro. To model OA, human bone marrow-derived mesenchymal stem/stromal cells (MSCs) were cultured in 3D pellets for up to 14 days in chondrogenic medium supplemented with pro-inflammatory IL-1β (10 ng/mL). Physicochemical characterization of Ibuf-Ch/PGA NPs, drug release, and anti-inflammatory potential of the Ibuf-NPs were evaluated in IL-1β-supplemented MSC pellets. Cartilage ECM was then characterized by histology/immunohistochemistry (IHC). Ibuf-Ch/PGA NPs (200 nm, 0.2 PdI, 20 mV charge) were obtained, with high drug entrapment efficiency (>90 %), and fast release at physiological pH. Both free drug and Ibuf-Ch/PGA NPs significantly reduce PGE2 in IL-1β-stimulated MSC pellets, but only Ibuf-NPs significantly stimulated collagen deposition, specifically collagen type 2. Overall, this study emphasizes a synergic potential of the NSAID Ibuprofen and Ch/PGA NPs to promote collagen deposition during chondrogenesis under pro-inflammatory conditions, opening new horizons in NSAID-based therapies to modulate inflammation in the context of OA.
Collapse
Affiliation(s)
- B Ribeiro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; FEUP - Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - A L Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - S Pilão
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; FEUP - Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - C Ribeiro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ISEP - Instituto Superior de Engenharia do Porto, Rua Dr. António Bernardino de Almeida, 431, 4249-015 Porto, Portugal
| | - M A Barbosa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - S G Santos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - R M Gonçalves
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
2
|
El-Saadony MT, Saad AM, Alkafaas SS, Dladla M, Ghosh S, Elkafas SS, Hafez W, Ezzat SM, Khedr SA, Hussien AM, Fahmy MA, Elesawi IE, Salem HM, Mohammed DM, Abd El-Mageed TA, Ahmed AE, Mosa WFA, El-Tarabily MK, AbuQamar SF, El-Tarabily KA. Chitosan, derivatives, and its nanoparticles: Preparation, physicochemical properties, biological activities, and biomedical applications - A comprehensive review. Int J Biol Macromol 2025:142832. [PMID: 40187443 DOI: 10.1016/j.ijbiomac.2025.142832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 03/17/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Chitosan, derived from the deacetylation of chitin, is the second most widely used natural polymer, valued for its nontoxic, biocompatible, and biodegradable properties. These attributes have driven extensive research into diverse applications of chitosan and various derivatives. The key characteristics of chitosan muco-adhesion, permeability enhancement, drug release modulation, and antimicrobial activity are primarily due to its amino and hydroxyl groups. However, the limited solubility of raw chitosan in water and most organic solvents has posed challenges for broader application. Numerous chemically modified derivatives have been developed to address these inadequacies with improved physical and chemical properties. Among these derivatives, chitosan nanoparticles have emerged as versatile drug carriers with precise release kinetics and the capacity for targeted delivery, greatly enhancing drug efficacy and safety profiles for therapeutic applications. Due to these unique physicochemical properties, chitosan and chitosan nanoparticles are promising for improved drug delivery, vaccine administration, transplantation, gene therapy, and diagnostics. This review examines the physicochemical properties and bioactivities of chitosan and chitosan nanoparticles, emphasizing their broad-ranging biomedical applications.
Collapse
Affiliation(s)
- Mohamed T El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt.
| | - Ahmed M Saad
- Biochemistry Department, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| | - Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Mthokozisi Dladla
- Department of Genetics, Faculty of Natural and Agricultural Sciences, University of the Free State, Bloemfontein 9301, South Africa
| | - Soumya Ghosh
- Department of Genetics, Faculty of Natural and Agricultural Sciences, University of the Free State, Bloemfontein 9301, South Africa
| | - Sara Samy Elkafas
- Production Engineering and Mechanical Design Department, Faculty of Engineering, Menofia University, Menofia 32511, Egypt; Faculty of Control System and Robotics, Information Technologies, Mechanics and Optics (ITMO) University, Saint-Petersburg 191002, Russia
| | - Wael Hafez
- Medical Research Division, Department of Internal Medicine, The National Research Centre, Cairo 11511, Egypt
| | - Salma Mohamed Ezzat
- Department of Chemistry, Division of Biochemistry, Faculty of Science, Tanta University, Tanta 44511, Egypt
| | - Sohila A Khedr
- Industrial Biotechnology Department, Faculty of Science, Tanta University, Tanta 31733, Egypt
| | - Aya Misbah Hussien
- Biotechnology Department at Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Mohamed A Fahmy
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| | - Ibrahim Eid Elesawi
- Biochemistry Department, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| | - Heba M Salem
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt; Department of Diseases of Birds, Rabbits, Fish & Their Care & Wildlife, School of Veterinary Medicine, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Dina Mostafa Mohammed
- Nutrition and Food Sciences Department, National Research Centre, Dokki, Giza 12622, Egypt
| | - Taia A Abd El-Mageed
- Department of Soils and Water, Faculty of Agriculture, Fayoum University, Fayoum 63514, Egypt
| | - Ahmed Ezzat Ahmed
- Department of Biology, College of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - Walid F A Mosa
- Plant Production Department (Horticulture-Pomology), Faculty of Agriculture, Saba Basha, Alexandria University, Alexandria 21531, Egypt
| | | | - Synan F AbuQamar
- Department of Biology, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates.
| | - Khaled A El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates.
| |
Collapse
|
3
|
Duymaz D, Kebabci AO, Kizilel S. Harnessing the immunomodulatory potential of chitosan and its derivatives for advanced biomedical applications. Int J Biol Macromol 2025; 307:142055. [PMID: 40090654 DOI: 10.1016/j.ijbiomac.2025.142055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 02/21/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025]
Abstract
The success of biomaterial applications in medicine, particularly in tissue engineering, relies on achieving a balance between promoting tissue regeneration and controlling the immune response. Due to its natural origin, high biocompatibility, and versatility, chitosan has emerged as a promising biomaterial especially for immunomodulation purposes. Immunomodulation, refers to the deliberate alteration of the immune system's activity to achieve a desired therapeutic effect either by enhancing or suppressing the function of specific immune cells, signaling pathways, or cytokine production. This modulation opens up the unlimited possibilities for the use of biomaterials, especially about the use of natural polymers such as chitosan. Although numerous chitosan-based immunoregulatory strategies have been demonstrated over the past two decades, the lack of in-depth exploration hinders the full potential of strategies that include chitosan and its derivatives in biomedical applications. Thus, in this review, the possible immunomodulatory effects of chitosan, chitosan derivatives and their potential combined with various agents and therapies are investigated in detail. Moreover, this report includes agents for localized immune response control, chitosan-based strategies with complementary immunomodulatory properties to create synergistic effects that will influence the success of cell therapies for enhanced tissue acceptance and regeneration. Finally, the challenges and outlook of chitosan-based therapies as a powerful tool for improving immunomodulatory applications are discussed for paving the way for further studies.
Collapse
Affiliation(s)
- Doğukan Duymaz
- Chemical and Biological Engineering, Koç University, Sariyer, İstanbul, Turkiye
| | - Aybaran O Kebabci
- Chemical and Biological Engineering, Koç University, Sariyer, İstanbul, Turkiye
| | - Seda Kizilel
- Chemical and Biological Engineering, Koç University, Sariyer, İstanbul, Turkiye.
| |
Collapse
|
4
|
Zhu H, Ren J, Wang X, Qin W, Xie Y. Targeting skeletal interoception: a novel mechanistic insight into intervertebral disc degeneration and pain management. J Orthop Surg Res 2025; 20:159. [PMID: 39940003 PMCID: PMC11823264 DOI: 10.1186/s13018-025-05577-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/05/2025] [Indexed: 02/14/2025] Open
Abstract
Despite being a leading cause of chronic pain and disability, the underlying mechanisms of intervertebral disc (IVD) degeneration (IVDD) remain unclear. Emerging evidence suggests that mechanosensation (the ability of the skeletal system to perceive mechanical and biochemical signals) mediated by abnormal mechanical loading plays a critical role in the regulation of IVD health. This review examines the complex interactions amongIVDs, intraosseous sensory mechanisms, and the central nervous system (CNS), with a particular focus on the roles of pathways such as PGE2/EP4, Wnt/β-catenin, and NF-κB. This review elucidates the manner in which mechanical stress and aberrant signaling disrupt the homeostasis of the nucleus pulposus (NP), cartilaginous endplate (CEP) and annulus fibrosus (AF), thereby driving degeneration and exacerbating pain. Furthermore, targeted therapeutic strategies, including the modulation of skeletal interoception and dynamic mechanical loading, present novel avenues for reversing IVDD progression. By integrating skeletal biology with spinal pathology, this work offers a novel perspective on the pathogenesis of IVDD and identifies promising strategies for clinical intervention. These findings highlight the potential of targeting skeletal interoception to mitigate IVDD and associated pain, paving the way for innovative, mechanism-driven therapies.
Collapse
Affiliation(s)
- Houcheng Zhu
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610000, China
| | - JianHang Ren
- Affiliated Yongchuan Hospital of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 402160, China
| | - Xiangjin Wang
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610000, China
| | - Wenjing Qin
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610000, China
| | - Yong Xie
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610000, China.
| |
Collapse
|
5
|
Costa MP, Abdu JOC, Machado Resende Guedes MC, Sarcinelli MA, Fabri RL, Pittella F, Macedo GC, Vilela FMP, Rocha HVA, Tavares GD. Dexamethasone-loaded chitosan-decorated PLGA nanoparticles: A step forward in attenuating the COVID-19 cytokine storm? Colloids Surf B Biointerfaces 2025; 246:114359. [PMID: 39522287 DOI: 10.1016/j.colsurfb.2024.114359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
This study aims to develop and characterize poly (lactic-co-glycolic acid) (PLGA) nanoparticles decorated with chitosan (CS) for the encapsulation of dexamethasone (DEX) (NP-DEX-CS), targeting improved efficacy in the treatment of severe acute respiratory syndrome (SARS) associated with COVID-19. The nanoparticles were systematically characterized for size, zeta potential (ZP), morphology, encapsulation efficiency, and in vitro drug release. Incorporation of CS resulted in significant modifications in the nanoparticles' physical properties, notably an increase in size (from 207.3 ± 6.7 nm to 264.4 ± 4.4 nm) and a shift in ZP to positive values (from -11.8 ±1.4 mV to +30.0 ± 1,6 mV). The NP-DEX-CS formulation achieved a high encapsulation efficiency (∼79 %) and a drug loading capacity of 6.53 ± 0.02 %.In addition, the in vitro release rate of DEX from NP-DEX-CS was lower compared to undecorated nanoparticles, with a reduction from approximately 64-37 % within 24 h. Microscopy analyses revealed a smoother surface on the CS-decorated nanoparticles. FTIR and XRD analyses confirmed successful chitosan coating and DEX encapsulation. The CS coating enhanced the tolerability of J774.A1 cells to the nanoparticles, particularly evident at the highest concentration (400ug/mL), resulting in a cell viability ≥70 %. Importantly, the NP-DEX-CS significantly reduced levels of nitric oxide and inflammatory cytokines (IL-1, IL-6, IL-12, and TNF-α). These findings suggest that CS-decorated PLGA nanoparticles hold promise as an effective dexamethasone delivery system for treating SARS related to COVID-19.
Collapse
Affiliation(s)
- Mirsiane Pascoal Costa
- Postgraduate Program in Pharmaceutical Science, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| | | | - Maria Clara Machado Resende Guedes
- Postgraduate Program in Biological Sciences, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| | | | - Rodrigo Luiz Fabri
- Postgraduate Program in Pharmaceutical Science, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| | - Frederico Pittella
- Postgraduate Program in Pharmaceutical Science, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil; Faculty of Pharmacy, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| | - Gilson Costa Macedo
- Postgraduate Program in Biological Sciences, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| | - Fernanda Maria Pinto Vilela
- Postgraduate Program in Pharmaceutical Science, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil; Faculty of Pharmacy, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| | | | - Guilherme Diniz Tavares
- Postgraduate Program in Pharmaceutical Science, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil; Faculty of Pharmacy, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| |
Collapse
|
6
|
Kolpek DJ, Kim J, Mohammed H, Gensel JC, Park J. Physicochemical Property Effects on Immune Modulating Polymeric Nanoparticles: Potential Applications in Spinal Cord Injury. Int J Nanomedicine 2024; 19:13357-13374. [PMID: 39691455 PMCID: PMC11649979 DOI: 10.2147/ijn.s497859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024] Open
Abstract
Nanoparticles (NPs) offer promising potential as therapeutic agents for inflammation-related diseases, owing to their capabilities in drug delivery and immune modulation. In preclinical studies focusing on spinal cord injury (SCI), polymeric NPs have demonstrated the ability to reprogram innate immune cells. This reprogramming results in redirecting immune cells away from the injury site, downregulating pro-inflammatory signaling, and promoting a regenerative environment post-injury. However, to fully understand the mechanisms driving these effects and maximize therapeutic efficacy, it is crucial to assess NP interactions with innate immune cells. This review examines how the physicochemical properties of polymeric NPs influence their modulation of the immune system. To achieve this, the review delves into the roles played by innate immune cells in SCI and investigates how various NP properties influence cellular interactions and subsequent immune modulation. Key NP properties such as size, surface charge, molecular weight, shape/morphology, surface functionalization, and polymer composition are thoroughly examined. Furthermore, the review establishes connections between these properties and their effects on the immunomodulatory functions of NPs. Ultimately, this review suggests that leveraging NPs and their physicochemical properties could serve as a promising therapeutic strategy for treating SCI and potentially other inflammatory diseases.
Collapse
Affiliation(s)
- Daniel J Kolpek
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Jaechang Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Hisham Mohammed
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - John C Gensel
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Jonghyuck Park
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
7
|
Zhang H, Yang X, Huang Y, Li Y, Hu Q, Wei Q, Xu W, Ding W, Guo Y, Shen JW. Reviving Intervertebral Discs: Treating Degeneration Using Advanced Delivery Systems. Mol Pharm 2024; 21:373-392. [PMID: 38252032 DOI: 10.1021/acs.molpharmaceut.3c00579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Intervertebral disc degeneration (IVDD) is commonly associated with many spinal problems, such as low back pain, and significantly impacts a patient's quality of life. However, current treatments for IVDD, which include conservative and surgical methods, are limited in their ability to fully address degeneration. To combat IVDD, delivery-system-based therapy has received extensive attention from researchers. These delivery systems can effectively deliver therapeutic agents for IVDD, overcoming the limitations of these agents, reducing leakage and increasing local concentration to inhibit IVDD or promote intervertebral disc (IVD) regeneration. This review first briefly introduces the structure and function of the IVD, and the related pathophysiology of IVDD. Subsequently, the roles of drug-based and bioactive-substance-based delivery systems in IVDD are highlighted. The former includes natural source drugs, nonsteroidal anti-inflammatory drugs, steroid medications, and other small molecular drugs. The latter includes chemokines, growth factors, interleukin, and platelet-rich plasma. Additionally, gene-based and cell-based delivery systems are briefly involved. Finally, the limitations and future development of the combination of therapeutic agents and delivery systems in the treatment of IVDD are discussed, providing insights for future research.
Collapse
Affiliation(s)
- Hong Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Xiaorong Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yiheng Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yue Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Quan Hu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Qiaolin Wei
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Weixing Xu
- Department of Orthopedics, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province 310012, China
| | - Weiguo Ding
- Department of Orthopedics, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province 310012, China
| | - Yong Guo
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jia-Wei Shen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| |
Collapse
|
8
|
Álvarez K, Rojas M. Nanoparticles targeting monocytes and macrophages as diagnostic and therapeutic tools for autoimmune diseases. Heliyon 2023; 9:e19861. [PMID: 37810138 PMCID: PMC10559248 DOI: 10.1016/j.heliyon.2023.e19861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 08/16/2023] [Accepted: 09/04/2023] [Indexed: 10/10/2023] Open
Abstract
Autoimmune diseases are chronic conditions that result from an inadequate immune response to self-antigens and affect many people worldwide. Their signs, symptoms, and clinical severity change throughout the course of the disease, therefore the diagnosis and treatment of autoimmune diseases are major challenges. Current diagnostic tools are often invasive and tend to identify the issue at advanced stages. Moreover, the available treatments for autoimmune diseases do not typically lead to complete remission and are associated with numerous side effects upon long-term usage. A promising strategy is the use of nanoparticles that can be used as contrast agents in diagnostic imaging techniques to detect specific cells present at the inflammatory infiltrates in tissues that are not easily accessible by biopsy. In addition, NPs can be designed to deliver drugs to a cell population or tissue. Considering the significant role played by monocytes in the development of chronic inflammatory conditions and their emergence as a target for extracorporeal monitoring and precise interventions, this review focuses on recent advancements in nanoparticle-based strategies for diagnosing and treating autoimmune diseases, with a particular emphasis on targeting monocyte populations.
Collapse
Affiliation(s)
- Karen Álvarez
- Grupo de Inmunología Celular e Inmunogenética, Sede de Investigación Universitaria (SIU), Universidad de Antioquia (UDEA), Colombia
| | - Mauricio Rojas
- Grupo de Inmunología Celular e Inmunogenética, Sede de Investigación Universitaria (SIU), Universidad de Antioquia (UDEA), Colombia
- Unidad de Citometría de Flujo, Sede de Investigación Universitaria (SIU), Universidad de Antioquia (UDEA), Colombia
| |
Collapse
|
9
|
Shnayder NA, Ashhotov AV, Trefilova VV, Novitsky MA, Medvedev GV, Petrova MM, Narodova EA, Kaskaeva DS, Chumakova GA, Garganeeva NP, Lareva NV, Al-Zamil M, Asadullin AR, Nasyrova RF. High-Tech Methods of Cytokine Imbalance Correction in Intervertebral Disc Degeneration. Int J Mol Sci 2023; 24:13333. [PMID: 37686139 PMCID: PMC10487844 DOI: 10.3390/ijms241713333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
An important mechanism for the development of intervertebral disc degeneration (IDD) is an imbalance between anti-inflammatory and pro-inflammatory cytokines. Therapeutic and non-therapeutic approaches for cytokine imbalance correction in IDD either do not give the expected result, or give a short period of time. This explains the relevance of high-tech medical care, which is part of specialized care and includes the use of new resource-intensive methods of treatment with proven effectiveness. The aim of the review is to update knowledge about new high-tech methods based on cytokine imbalance correction in IDD. It demonstrates promise of new approaches to IDD management in patients resistant to previously used therapies, including: cell therapy (stem cell implantation, implantation of autologous cultured cells, and tissue engineering); genetic technologies (gene modifications, microRNA, and molecular inducers of IDD); technologies for influencing the inflammatory cascade in intervertebral discs mediated by abnormal activation of inflammasomes; senolytics; exosomal therapy; and other factors (hypoxia-induced factors; lysyl oxidase; corticostatin; etc.).
Collapse
Affiliation(s)
- Natalia A. Shnayder
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia; (A.V.A.); (V.V.T.)
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (M.M.P.); (E.A.N.); (D.S.K.)
| | - Azamat V. Ashhotov
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia; (A.V.A.); (V.V.T.)
| | - Vera V. Trefilova
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia; (A.V.A.); (V.V.T.)
- Department of Neurology, Hospital for War Veterans, 193079 Saint Petersburg, Russia;
| | - Maxim A. Novitsky
- Department of Neurology, Hospital for War Veterans, 193079 Saint Petersburg, Russia;
| | - German V. Medvedev
- R.R. Vreden National Medical Research Center for Traumatology and Orthopedics, 195427 Saint-Petersburg, Russia;
| | - Marina M. Petrova
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (M.M.P.); (E.A.N.); (D.S.K.)
| | - Ekaterina A. Narodova
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (M.M.P.); (E.A.N.); (D.S.K.)
| | - Daria S. Kaskaeva
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (M.M.P.); (E.A.N.); (D.S.K.)
| | - Galina A. Chumakova
- Department of Therapy and General Medical Practice with a Course of Postgraduate Professional Education, Altai State Medical University, 656038 Barnaul, Russia;
| | - Natalia P. Garganeeva
- Department of General Medical Practice and Outpatient Therapy, Siberian State Medical University, 634050 Tomsk, Russia;
| | - Natalia V. Lareva
- Department of Therapy of Faculty of Postgraduate Education, Chita State Medical Academy, 672000 Chita, Russia;
| | - Mustafa Al-Zamil
- Department of Physiotherapy, Faculty of Continuing Medical Education, Peoples’ Friendship University of Russia, 117198 Moscow, Russia;
| | - Azat R. Asadullin
- Department of Psychiatry and Addiction, Bashkir State Medical University, 450008 Ufa, Russia;
| | - Regina F. Nasyrova
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia; (A.V.A.); (V.V.T.)
- International Centre for Education and Research in Neuropsychiatry, Samara State Medical University, 443016 Samara, Russia
| |
Collapse
|
10
|
Anwar F, Naqvi S, Shams S, Sheikh RA, Al-Abbasi FA, Asseri AH, Baig MR, Kumar V. Nanomedicines: intervention in inflammatory pathways of cancer. Inflammopharmacology 2023; 31:1199-1221. [PMID: 37060398 PMCID: PMC10105366 DOI: 10.1007/s10787-023-01217-w] [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: 03/12/2023] [Accepted: 03/29/2023] [Indexed: 04/16/2023]
Abstract
Inflammation is a complex defense process that maintains tissue homeostasis. However, this complex cascade, if lasts long, may contribute to pathogenesis of several diseases. Chronic inflammation has been exhaustively studied in the last few decades, for its contribution in development and progression of cancer. The intrinsic limitations of conventional anti-inflammatory and anti-cancer therapies triggered the development of nanomedicines for more effective and safer therapies. Targeting inflammation and tumor cells by nanoparticles, encapsulated with active therapeutic agents, offers a promising outcome with patient survival. Considerable technological success has been achieved in this field through exploitation of tumor microenvironment, and recognition of molecules overexpressed on endothelial cells or macrophages, through enhanced vascular permeability, or by rendering biomimetic approach to nanoparticles. This review focusses on the inflammatory pathways in progression of a tumor, and advancement in nanotechnologies targeting these pathways. We also aim to identify the gaps that hinder the successful clinical translation of nanotherapeutics with further clinical studies that will allow oncologist to precisely identify the patients who may be benefited from nanotherapy at time when promotion or progression of tumor initiates. It is postulated that the nanomedicines, in near future, will shift the paradigm of cancer treatment and improve patient survival.
Collapse
Affiliation(s)
- Firoz Anwar
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia.
| | - Salma Naqvi
- Department of Biomedical Sciences, College of Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Saiba Shams
- School of Pharmaceutical Education & Research, (Deemed to be University), New Delhi, 110062, India
| | - Ryan Adnan Sheikh
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Fahad A Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Amer H Asseri
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Mirza Rafi Baig
- Department of Clinical Pharmacy & Pharmacotherapeutics. Dubai Pharmacy College for Girls, Po Box 19099, Dubai, United Arab Emirates
| | - Vikas Kumar
- Natural Product Drug Discovery Laboratory, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Sam Higginbottom Institute of Agriculture, Technology & Sciences, Allahabad, Uttar Pradesh, India.
| |
Collapse
|
11
|
Inhibition of miR-652-3p Regulates Lipid Metabolism and Inflammatory Cytokine Secretion of Macrophages to Alleviate Atherosclerosis by Improving TP53 Expression. Mediators Inflamm 2022; 2022:9655097. [PMID: 36248191 PMCID: PMC9568360 DOI: 10.1155/2022/9655097] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/30/2022] [Indexed: 11/26/2022] Open
Abstract
Purpose The aim was to elucidate the regulatory function of miR-652-3p on lipid metabolism and inflammatory cytokine secretion of macrophages in atherosclerosis. Methods miR-652-3p level in atherosclerosis patients, ox-LDL-treated macrophages, and their controls were monitored by Q-PCR. After ox-LDL treatment and miR-652-3p mimic, si-TP53 and their controls transfection, ELISA, and Q-PCR assays were used to detect IL-1ß, IL-6, and TNF-α levels. oil red O staining was processed to verify cholesterol accumulation. CE/TC and lipid metabolism were also detected. The protein levels of ABCA1, ABCG1, PPARα, CRT1, ADRP, and ALBP were detected by western blot assay. Based on the TargetScan database, the TP53 3′UTR region had complementary bases with miR-652-3p, which was also verified by dual-luciferase reporter gene assay. Finally, the regulation of miR-652-3p and TP53 was confirmed by rescue assay in atherosclerosis. Results miR-652-3p is highly expressed in atherosclerosis, miR-652-3p inhibitor decreased IL-1β, IL-6, and TNF-α expression after ox-LDL treatment. Knockdown of miR-652-3p reduces foam formation in ox-LDL-treated macrophages. miR-652-3p inhibitor ameliorates cholesterol accumulation and lipid metabolism disorder. miR-652-3p negatively regulated TP53 in atherosclerosis. Si-TP53 rescued the effect of miR-652 inhibitor in atherosclerosis. Conclusion miR-652-3p regulates the lipid metabolism of macrophages to alleviate atherosclerosis by inhibiting TP53 expression. It might be a potential target for atherosclerosis treatment.
Collapse
|
12
|
Novel Bile Salt Stabilized Vesicles-Mediated Effective Topical Delivery of Diclofenac Sodium: A New Therapeutic Approach for Pain and Inflammation. Pharmaceuticals (Basel) 2022; 15:ph15091106. [PMID: 36145327 PMCID: PMC9506322 DOI: 10.3390/ph15091106] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
The oral delivery of diclofenac sodium (DNa), a non-steroidal analgesic, anti-inflammatory drug, is associated with various gastrointestinal side effects. The aim of the research was to appraise the potential of transdermal delivery of DNa using bilosomes as a vesicular carrier (BSVC) in inflamed paw edema. DNa-BSVCs were elaborated using a thin-film hydration technique and optimized using a 31.22 multilevel categoric design with Design Expert® software 10 software (Stat-Ease, Inc., Minneapolis, MI, USA). The effect of formulation variables on the physicochemical properties of BSVC, as well as the optimal formulation selection, was investigated. The BSVCs were evaluated for various parameters including entrapment efficiency (EE%), vesicle size (VS), zeta potential (ZP) and permeation studies. The optimized BSVC was characterized for in vitro release, Fourier transform infrared spectroscopy (FTIR), transmission electron microscopy (TEM) and incorporated into hydrogel base. The optimized DNa-BSVC gel effectiveness was assessed in vivo using carrageenan-induced paw edema animal model via cyclooxygenase 2 (COX-2), interleukin 6 (IL-6), Hemooxygenase 1 (HO-1) and nuclear factor-erythroid factor2-related factor 2 (Nfr-2) that potentiate anti-inflammatory and anti-oxidant activity coupled with histopathological investigation. The resulting vesicles presented VS from 120.4 ± 0.65 to 780.4 ± 0.99 nm, EE% from 61.7 ± 3.44 to 93.2 ± 2.21%, ZP from −23.8 ± 2.65 to −82.1 ± 12.63 mV and permeation from 582.9 ± 32.14 to 1350.2 ± 45.41 µg/cm2. The optimized BSVCs were nano-scaled spherical vesicles with non-overlapped bands of their constituents in the FTIR. Optimized formulation has superior skin permeability ex vivo approximately 2.5 times greater than DNa solution. Furthermore, histological investigation discovered that the formed BSVC had no skin irritating properties. It was found that DNa-BSVC gel suppressed changes in oxidative inflammatory mediators (COX-2), IL-6 and consequently enhanced Nrf2 and HO-1 levels. Moreover, reduction of percent of paw edema by about three-folds confirmed histopathological alterations. The results revealed that the optimized DNa-BSVC could be a promising transdermal drug delivery system to boost anti-inflammatory efficacy of DNa by enhancing the skin permeation of DNa and suppressing the inflammation of rat paw edema.
Collapse
|
13
|
Guimarães CF, Marques AP, Reis RL. Pushing the Natural Frontier: Progress on the Integration of Biomaterial Cues toward Combinatorial Biofabrication and Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105645. [PMID: 35419887 DOI: 10.1002/adma.202105645] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 03/14/2022] [Indexed: 06/14/2023]
Abstract
The engineering of fully functional, biological-like tissues requires biomaterials to direct cellular events to a near-native, 3D niche extent. Natural biomaterials are generally seen as a safe option for cell support, but their biocompatibility and biodegradability can be just as limited as their bioactive/biomimetic performance. Furthermore, integrating different biomaterial cues and their final impact on cellular behavior is a complex equation where the outcome might be very different from the sum of individual parts. This review critically analyses recent progress on biomaterial-induced cellular responses, from simple adhesion to more complex stem cell differentiation, looking at the ever-growing possibilities of natural materials modification. Starting with a discussion on native material formulation and the inclusion of cell-instructive cues, the roles of shape and mechanical stimuli, the susceptibility to cellular remodeling, and the often-overlooked impact of cellular density and cell-cell interactions within constructs, are delved into. Along the way, synergistic and antagonistic combinations reported in vitro and in vivo are singled out, identifying needs and current lessons on the development of natural biomaterial libraries to solve the cell-material puzzle efficiently. This review brings together knowledge from different fields envisioning next-generation, combinatorial biomaterial development toward complex tissue engineering.
Collapse
Affiliation(s)
- Carlos F Guimarães
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Alexandra P Marques
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
14
|
V. Lima B, Oliveira MJ, Barbosa MA, Gonçalves RM, Castro F. Harnessing chitosan and poly-(γ-glutamic acid)-based biomaterials towards cancer immunotherapy. MATERIALS TODAY ADVANCES 2022; 15:100252. [DOI: 10.1016/j.mtadv.2022.100252] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
15
|
Li C, Zheng X, Hu M, Jia M, Jin R, Nie Y. Recent progress in therapeutic strategies and biomimetic nanomedicines for rheumatoid arthritis treatment. Expert Opin Drug Deliv 2022; 19:883-898. [PMID: 35760767 DOI: 10.1080/17425247.2022.2094364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is an autoimmune systemic disease in which inflammatory and immune cells accumulate in inflamed joints. Researchers aimed at the characteristics of RA to achieve the effect of treating RA through different therapeutic strategies, and have used various endogenous materials to design drug-loaded nanoparticles that can target RA by binding to cell adhesion molecules or chemokines. In some cases, the nanoparticles can respond to the characteristics of the microenvironment. AREAS COVERED This article reviews the recent advances in the treatment of RA from two aspects of therapeutic strategies and delivery strategies. Therapeutic strategies mainly include neutralization of inflammatory factors, promotion of inflammatory cell apoptosis, ROS scavenger, immunosuppression, and bone tissue repair. The drug delivery strategy is mainly described from two aspects: chemically functionalized biomimetic nanoparticles and endogenous nanoparticles. EXPERT OPINION Biomimetic NPs may be effective drug carriers for targeted RA treatment. NPs can reduce the clearance of mononuclear phagocytes, prolong the blood circulation time, and improve the targeting ability. With the deepening of research, more and more biomimetic NPs have entered the clinical trial stage. However, safe and scalable preparation methods are needed to improve their clinical applicability.
Collapse
Affiliation(s)
- Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Xiu Zheng
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Mei Hu
- Pharmacy Laboratory, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Ming Jia
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Yu Nie
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| |
Collapse
|
16
|
Morales X, Cortés-Domínguez I, Ortiz-de-Solorzano C. Modeling the Mechanobiology of Cancer Cell Migration Using 3D Biomimetic Hydrogels. Gels 2021; 7:17. [PMID: 33673091 PMCID: PMC7930983 DOI: 10.3390/gels7010017] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/29/2021] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
Understanding how cancer cells migrate, and how this migration is affected by the mechanical and chemical composition of the extracellular matrix (ECM) is critical to investigate and possibly interfere with the metastatic process, which is responsible for most cancer-related deaths. In this article we review the state of the art about the use of hydrogel-based three-dimensional (3D) scaffolds as artificial platforms to model the mechanobiology of cancer cell migration. We start by briefly reviewing the concept and composition of the extracellular matrix (ECM) and the materials commonly used to recreate the cancerous ECM. Then we summarize the most relevant knowledge about the mechanobiology of cancer cell migration that has been obtained using 3D hydrogel scaffolds, and relate those discoveries to what has been observed in the clinical management of solid tumors. Finally, we review some recent methodological developments, specifically the use of novel bioprinting techniques and microfluidics to create realistic hydrogel-based models of the cancer ECM, and some of their applications in the context of the study of cancer cell migration.
Collapse
Affiliation(s)
| | | | - Carlos Ortiz-de-Solorzano
- IDISNA, Ciberonc and Solid Tumors and Biomarkers Program, Center for Applied Medical Research, University of Navarra, 31008 Pamplona, Spain; (X.M.); (I.C.-D.)
| |
Collapse
|
17
|
Li Z, Bratlie KM. Fibroblasts treated with macrophage conditioned medium results in phenotypic shifts and changes in collagen organization. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 122:111915. [PMID: 33641908 DOI: 10.1016/j.msec.2021.111915] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/30/2020] [Accepted: 01/23/2021] [Indexed: 01/08/2023]
Abstract
In tissue regeneration, the goal is to regenerate tissue similar to what was damaged or missing while preventing fibrotic scarring, which may lead to decreased mechanical strength and dissimilar tissue characteristics compared to native tissue. We believe collagen orientation plays a critical role in wound contraction and scarring and that it is modulated by myofibroblasts. We used macrophage conditioned medium to simulate complex events that can influence the fibroblast phenotype during the wound healing process. In addition to examining the effect of macrophage phenotype on fibroblasts, we inhibited focal adhesion kinase (FAK), Rho-associated protein kinase (ROCK), and myosin II for fibroblasts cultured on both tissue culture plastic and methacrylated gellan gum to understand how different pathways and materials influence fibroblast responses. Collagen orientation, α-SMA expression, focal adhesion area, and cell migration were altered by inhibition of FAK, ROCK, or myosin II and macrophage phenotype, along with the substrate. An increase in either focal adhesion area or α-smooth muscle actin (α-SMA) expression correlated with an aligned collagen orientation. Gellan gum hydrogels upregulated α-SMA expression in ROCK inhibited conditioned media and downregulated the FAK area in FAK and ROCK inhibited conditioned media. Myosin II had no impact on the α-SMA expression on the substrate compared to coverslip except for M2 conditioned medium. Gellan gum hydrogel significantly increased cell migration under FAK and Myosin II mediated conditioned media and unconditioned media. Collectively, our study examined how macrophage phenotype influences fibroblast response, which would be beneficial in controlling scar tissue formation.
Collapse
Affiliation(s)
- Zhuqing Li
- Department of Materials Science & Engineering, Iowa State University, Ames, IA 50011, USA
| | - Kaitlin M Bratlie
- Department of Materials Science & Engineering, Iowa State University, Ames, IA 50011, USA; Department of Chemical & Biological Engineering, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
18
|
Castro F, Pinto ML, Pereira CL, Serre K, Barbosa MA, Vermaelen K, Gärtner F, Gonçalves RM, De Wever O, Oliveira MJ. Chitosan/γ-PGA nanoparticles-based immunotherapy as adjuvant to radiotherapy in breast cancer. Biomaterials 2020; 257:120218. [DOI: 10.1016/j.biomaterials.2020.120218] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/05/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023]
|
19
|
Brusini R, Varna M, Couvreur P. Advanced nanomedicines for the treatment of inflammatory diseases. Adv Drug Deliv Rev 2020; 157:161-178. [PMID: 32697950 PMCID: PMC7369016 DOI: 10.1016/j.addr.2020.07.010] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/04/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023]
Abstract
Inflammation, a common feature of many diseases, is an essential immune response that enables survival and maintains tissue homeostasis. However, in some conditions, the inflammatory process becomes detrimental, contributing to the pathogenesis of a disease. Targeting inflammation by using nanomedicines (i.e. nanoparticles loaded with a therapeutic active principle), either through the recognition of molecules overexpressed onto the surface of activated macrophages or endothelial cells, or through enhanced vasculature permeability, or even through biomimicry, offers a promising solution for the treatment of inflammatory diseases. After providing a brief insight on the pathophysiology of inflammation and current therapeutic strategies, the review will discuss, at a pre-clinical stage, the main innovative nanomedicine approaches that have been proposed in the past five years for the resolution of inflammatory disorders, finally focusing on those currently in clinical trials.
Collapse
|
20
|
Yetisgin AA, Cetinel S, Zuvin M, Kosar A, Kutlu O. Therapeutic Nanoparticles and Their Targeted Delivery Applications. Molecules 2020; 25:E2193. [PMID: 32397080 PMCID: PMC7248934 DOI: 10.3390/molecules25092193] [Citation(s) in RCA: 409] [Impact Index Per Article: 81.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/24/2020] [Accepted: 04/26/2020] [Indexed: 12/12/2022] Open
Abstract
Nanotechnology offers many advantages in various fields of science. In this regard, nanoparticles are the essential building blocks of nanotechnology. Recent advances in nanotechnology have proven that nanoparticles acquire a great potential in medical applications. Formation of stable interactions with ligands, variability in size and shape, high carrier capacity, and convenience of binding of both hydrophilic and hydrophobic substances make nanoparticles favorable platforms for the target-specific and controlled delivery of micro- and macromolecules in disease therapy. Nanoparticles combined with the therapeutic agents overcome problems associated with conventional therapy; however, some issues like side effects and toxicity are still debated and should be well concerned before their utilization in biological systems. It is therefore important to understand the specific properties of therapeutic nanoparticles and their delivery strategies. Here, we provide an overview on the unique features of nanoparticles in the biological systems. We emphasize on the type of clinically used nanoparticles and their specificity for therapeutic applications, as well as on their current delivery strategies for specific diseases such as cancer, infectious, autoimmune, cardiovascular, neurodegenerative, ocular, and pulmonary diseases. Understanding of the characteristics of nanoparticles and their interactions with the biological environment will enable us to establish novel strategies for the treatment, prevention, and diagnosis in many diseases, particularly untreatable ones.
Collapse
Affiliation(s)
- Abuzer Alp Yetisgin
- Materials Science and Nano-Engineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey;
| | - Sibel Cetinel
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey;
| | - Merve Zuvin
- Mechatronics Engineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey; (M.Z.); (A.K.)
| | - Ali Kosar
- Mechatronics Engineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey; (M.Z.); (A.K.)
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabanci University, Istanbul 34956, Turkey
| | - Ozlem Kutlu
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey;
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabanci University, Istanbul 34956, Turkey
| |
Collapse
|
21
|
Modulation of the In Vivo Inflammatory Response by Pro- Versus Anti-Inflammatory Intervertebral Disc Treatments. Int J Mol Sci 2020; 21:ijms21051730. [PMID: 32138314 PMCID: PMC7084831 DOI: 10.3390/ijms21051730] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/18/2020] [Accepted: 02/27/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammation is central in intervertebral disc (IVD) degeneration/regeneration mechanisms, and its balance is crucial to maintain tissue homeostasis. This work investigates the modulation of local and systemic inflammatory response associated with IVD degeneration/herniation by administration of PRO- versus ANTI-inflammatory treatments. Chitosan/poly-γ-glutamic acid nanocomplexes, known as pro-inflammatory (PRO), and soluble diclofenac, a non-steroidal anti-inflammatory drug (ANTI), were intradiscally administered in a rat IVD injury model, 24 h after lesion. Two weeks after administration, a reduction of disc height accompanied by hernia formation was observed. In the PRO-inflammatory treated group, IL-1β, IL-6 and COX-2 IVD gene expression were upregulated, and loss of nucleus pulposus (NP) structure and composition was observed. Systemically, lower T-cell frequency was observed in the lymph nodes (LN) and spleen (SP) of the PRO group, together with an increase in CD4+ T cells subset in the blood (BL) and LN. In contrast, the ANTI-group had higher proteoglycans/collagen ratio and collagen type 2 content in the NP, while an increase in the frequency of myeloid cells, M1 macrophages and activated macrophages (MHCII+) was observed at the systemic level. Overall, this study illustrates the dynamics of local and systemic inflammatory and immune cell responses associated with intradiscal therapies, which will contribute to designing more successful immunomodulatory treatments for IVD degeneration.
Collapse
|
22
|
Zhang B, Cristescu R, Chrisey DB, Narayan RJ. Solvent-based Extrusion 3D Printing for the Fabrication of Tissue Engineering Scaffolds. Int J Bioprint 2020; 6:211. [PMID: 32596549 PMCID: PMC7294686 DOI: 10.18063/ijb.v6i1.211] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 12/02/2019] [Indexed: 11/23/2022] Open
Abstract
Three-dimensional (3D) printing has been emerging as a new technology for scaffold fabrication to overcome the problems associated with the undesirable microstructure associated with the use of traditional methods. Solvent-based extrusion (SBE) 3D printing is a popular 3D printing method, which enables incorporation of cells during the scaffold printing process. The scaffold can be customized by optimizing the scaffold structure, biomaterial, and cells to mimic the properties of natural tissue. However, several technical challenges prevent SBE 3D printing from translation to clinical use, such as the properties of current biomaterials, the difficulties associated with simultaneous control of multiple biomaterials and cells, and the scaffold-to-scaffold variability of current 3D printed scaffolds. In this review paper, a summary of SBE 3D printing for tissue engineering (TE) is provided. The influences of parameters such as ink biomaterials, ink rheological behavior, cross-linking mechanisms, and printing parameters on scaffold fabrication are considered. The printed scaffold structure, mechanical properties, degradation, and biocompatibility of the scaffolds are summarized. It is believed that a better understanding of the scaffold fabrication process and assessment methods can improve the functionality of SBE-manufactured 3D printed scaffolds.
Collapse
Affiliation(s)
- Bin Zhang
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27606, USA
| | - Rodica Cristescu
- National Institute for Lasers, Plasma and Radiation Physics, Lasers Department, P.O. Box MG-36, Bucharest-Magurele, Romania
| | - Douglas B Chrisey
- Department of Physics and Engineering Physics, Tulane University, New Orleans, LA, USA
| | - Roger J Narayan
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27606, USA
| |
Collapse
|
23
|
Nanomaterials for direct and indirect immunomodulation: A review of applications. Eur J Pharm Sci 2020; 142:105139. [DOI: 10.1016/j.ejps.2019.105139] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/14/2019] [Accepted: 11/03/2019] [Indexed: 01/03/2023]
|
24
|
Reichel D, Tripathi M, Perez JM. Biological Effects of Nanoparticles on Macrophage Polarization in the Tumor Microenvironment. Nanotheranostics 2019; 3:66-88. [PMID: 30662824 PMCID: PMC6328304 DOI: 10.7150/ntno.30052] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/02/2018] [Indexed: 12/11/2022] Open
Abstract
Biological interactions between tumor-associated macrophages (TAMs), cancer cells and other cells within the tumor microenvironment contribute to tumorigenesis, tumor growth, metastasis and therapeutic resistance. TAMs can remodel the tumor microenvironment to reduce growth barriers such as the dense extracellular matrix and shift tumors towards an immunosuppressive microenvironment that protects cancer cells from targeted immune responses. Nanoparticles can interrupt these biological interactions within tumors by altering TAM phenotypes through a process called polarization. Macrophage polarization within tumors can shift TAMs from a growth-promoting phenotype towards a cancer cell-killing phenotype that predicts treatment efficacy. Because many types of nanoparticles have been shown to preferentially accumulate within macrophages following systemic administration, there is considerable interest in identifying nanoparticle effects on TAM polarization, evaluating nanoparticle-induced TAM polarization effects on cancer treatment using drug-loaded nanoparticles and identifying beneficial types of nanoparticles for effective cancer treatment. In this review, the macrophage polarization effects of nanoparticles will be described based on their primary chemical composition. Because of their strong macrophage-polarizing and antitumor effects compared to other types of nanoparticles, the effects of iron oxide nanoparticles on macrophages will be discussed in detail. By comparing the macrophage polarization effects of various nanoparticle treatments reported in the literature, this review aims to both elucidate nanoparticle material effects on macrophage polarization and to provide insight into engineering nanoparticles with more beneficial immunological responses for cancer treatment.
Collapse
Affiliation(s)
- Derek Reichel
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Manisha Tripathi
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Current Address: Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - J. Manuel Perez
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| |
Collapse
|
25
|
Pinheiro A, Silva AM, Teixeira JH, Gonçalves RM, Almeida MI, Barbosa MA, Santos SG. Extracellular vesicles: intelligent delivery strategies for therapeutic applications. J Control Release 2018; 289:56-69. [PMID: 30261205 DOI: 10.1016/j.jconrel.2018.09.019] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles (EV), in particular exosomes, have been the object of intense research, due to their potential to mediate intercellular communication, modulating the phenotype of target cells. The natural properties and functions of EV are being exploited as biomarkers for disease diagnosis and prognosis, and as nano-bio-carriers for the development of new therapeutic strategies. EV have been particularly examined in the field of cancer, but are also increasingly investigated in other areas, like immune-related diseases and regenerative medicine. In this review, the therapeutic use of EV as drug delivery systems is described, balancing the advantages and drawbacks of different routes for their in vivo administration. Systemic and local delivery of EV are discussed, tackling the persisting difficulties in the assessment of their pharmacokinetics, pharmacodynamics and biodistribution in vivo. Finally, we discuss the future perspectives for incorporating EV into delivery systems and their use for an improved and controlled release of EV in vivo.
Collapse
Affiliation(s)
- Alice Pinheiro
- i3S - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; FEUP - Faculdade de Engenharia da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| | - Andreia M Silva
- i3S - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - José H Teixeira
- i3S - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Raquel M Gonçalves
- i3S - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Maria I Almeida
- i3S - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Mário A Barbosa
- i3S - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Susana G Santos
- i3S - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal.
| |
Collapse
|
26
|
Farjadian F, Moghoofei M, Mirkiani S, Ghasemi A, Rabiee N, Hadifar S, Beyzavi A, Karimi M, Hamblin MR. Bacterial components as naturally inspired nano-carriers for drug/gene delivery and immunization: Set the bugs to work? Biotechnol Adv 2018; 36:968-985. [PMID: 29499341 PMCID: PMC5971145 DOI: 10.1016/j.biotechadv.2018.02.016] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/20/2018] [Accepted: 02/26/2018] [Indexed: 12/28/2022]
Abstract
Drug delivery is a rapidly growing area of research motivated by the nanotechnology revolution, the ideal of personalized medicine, and the desire to reduce the side effects of toxic anti-cancer drugs. Amongst a bewildering array of different nanostructures and nanocarriers, those examples that are fundamentally bio-inspired and derived from natural sources are particularly preferred. Delivery of vaccines is also an active area of research in this field. Bacterial cells and their components that have been used for drug delivery, include the crystalline cell-surface layer known as "S-layer", bacterial ghosts, bacterial outer membrane vesicles, and bacterial products or derivatives (e.g. spores, polymers, and magnetic nanoparticles). Considering the origin of these components from potentially pathogenic microorganisms, it is not surprising that they have been applied for vaccines and immunization. The present review critically summarizes their applications focusing on their advantages for delivery of drugs, genes, and vaccines.
Collapse
Affiliation(s)
- Fatemeh Farjadian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohsen Moghoofei
- Department of Microbiology, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soroush Mirkiani
- Biomaterials Laboratory, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Amir Ghasemi
- Department of Materials Science and Engineering, Sharif University of Technology, Tehran, Iran
| | - Navid Rabiee
- Department of Chemistry, Shahid Beheshti University, Tehran, Iran
| | - Shima Hadifar
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | - Ali Beyzavi
- Koch institute of MIT, 500 Main Street, Cambridge, MA, USA
| | - Mahdi Karimi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA; Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
27
|
Castro F, Pinto ML, Silva AM, Pereira CL, Teixeira GQ, Gomez-Lazaro M, Santos SG, Barbosa MA, Gonçalves RM, Oliveira MJ. Pro-inflammatory chitosan/poly(γ-glutamic acid) nanoparticles modulate human antigen-presenting cells phenotype and revert their pro-invasive capacity. Acta Biomater 2017; 63:96-109. [PMID: 28919508 DOI: 10.1016/j.actbio.2017.09.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/06/2017] [Accepted: 09/13/2017] [Indexed: 02/06/2023]
Abstract
Anticancer immune responses depend on efficient presentation of tumor antigens and co-stimulatory signals provided by antigen-presenting cells (APCs). However, it is described that immature dendritic cells (DCs) and macrophages at the tumor site may have an immunosuppressive profile, which limits the activity of effector T cells and supports tumor progression. Therapeutic targeting of these innate immune cells, either aiming at their elimination or re-polarization towards an immunostimulatory profile, has been pointed as an attractive approach to control tumor progression. In the present work, we assessed the potential of Chitosan (Ch)/Poly(γ-glutamic acid) (γ-PGA) nanoparticles (NPs) to modulate macrophages and DCs inflammatory profile and to impair their ability to promote cancer cell invasion. Interestingly, Ch/γ-PGA NPs, prepared by co-acervation method, induced an immunostimulatory DCs phenotype, enhancing the expression of the co-stimulatory molecules CD86, CD40 and HLA-DR, and the secretion of the pro-inflammatory cytokines TNF-α, IL-12p40 and IL-6. Furthermore, Ch/γ-PGA NPs re-educated IL-10-stimulated macrophages towards a pro-inflammatory profile, decreasing the expression of CD163 and promoting the secretion of IL-12p40 and TNF-α. These alterations in the immune cells phenotype promoted CD4+ and CD8+ T cell activation/proliferation and partially inhibited APCs' ability to induce colorectal cancer cell invasion. Overall, our findings open new perspectives on the use of Ch/γ-PGA NPs as an immunomodulatory therapy for antigen-presenting cells reprogramming, providing a new tool for anticancer therapies. STATEMENT OF SIGNIFICANCE The immune system is responsible to detect and destroy abnormal cells preventing the development of cancer. However, the immunosuppressive tumor microenvironment can compromise the immune response favoring tumor progression. Thus, immune system modulation towards an immunostimulatory profile can improve anticancer therapies. This research focus on the development of chitosan/poly(γ-glutamic acid) nanoparticles (NPs) to modulate human antigen-presenting cells (APCs) phenotype and to counteract their pro-invasive capacity. Interestingly, Ch/γ-PGA NPs had a prominent effect in inducing macrophages and dendritic cells immunostimulatory phenotype, thus favoring T cell proliferation and inhibiting colorectal cancer cell invasion. We propose that their combination with other immunomodulatory drugs or conventional anticancer therapies can improve patients' outcome.
Collapse
|
28
|
van Rijt S, Habibovic P. Enhancing regenerative approaches with nanoparticles. J R Soc Interface 2017; 14:20170093. [PMID: 28404870 PMCID: PMC5414913 DOI: 10.1098/rsif.2017.0093] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/27/2017] [Indexed: 12/13/2022] Open
Abstract
In this review, we discuss recent developments in the field of nanoparticles and their use in tissue regeneration approaches. Owing to their unique chemical properties and flexibility in design, nanoparticles can be used as drug delivery systems, to create novel features within materials or as bioimaging agents, or indeed these properties can be combined to create smart multifunctional structures. This review aims to provide an overview of this research field where the focus will be on nanoparticle-based strategies to stimulate bone regeneration; however, the same principles can be applied for other tissue and organ regeneration strategies. In the first section, nanoparticle-based methods for the delivery of drugs, growth factors and genetic material to promote tissue regeneration are discussed. The second section deals with the addition of nanoparticles to materials to create nanocomposites. Such materials can improve several material properties, including mechanical stability, biocompatibility and biological activity. The third section will deal with the emergence of a relatively new field of research using nanoparticles in advanced cell imaging and stem cell tracking approaches. As the development of nanoparticles continues, incorporation of this technology in the field of regenerative medicine will ultimately lead to new tools that can diagnose, track and stimulate the growth of new tissues and organs.
Collapse
Affiliation(s)
- Sabine van Rijt
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, The Netherlands
| | - Pamela Habibovic
- MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, The Netherlands
| |
Collapse
|
29
|
Antunes JC, Pereira CL, Teixeira GQ, Silva RV, Caldeira J, Grad S, Gonçalves RM, Barbosa MA. Poly(γ-glutamic acid) and poly(γ-glutamic acid)-based nanocomplexes enhance type II collagen production in intervertebral disc. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2017; 28:6. [PMID: 27885573 DOI: 10.1007/s10856-016-5787-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 01/12/2016] [Indexed: 06/06/2023]
Abstract
Intervertebral disc (IVD) degeneration often leads to low back pain, which is one of the major causes of disability worldwide, affecting more than 80% of the population. Although available treatments for degenerated IVD decrease symptoms' progression, they fail to address the underlying causes and to restore native IVD properties. Poly(γ-glutamic acid) (γ-PGA) has recently been shown to support the production of chondrogenic matrix by mesenchymal stem/stromal cells. γ-PGA/chitosan (Ch) nanocomplexes (NCs) have been proposed for several biomedical applications, showing advantages compared with either polymer alone. Hence, this study explores the potential of γ-PGA and γ-PGA/Ch NCs for IVD regeneration. Nucleotomised bovine IVDs were cultured ex vivo upon injection of γ-PGA (pH 7.4) and γ-PGA/Ch NCs (pH 5.0 and pH 7.4). Tissue metabolic activity and nucleus pulposus DNA content were significantly reduced when NCs were injected in acidic-buffered solution (pH 5.0). However, at pH 7.4, both γ-PGA and NCs promoted sulphated glycosaminoglycan production and significant type II collagen synthesis, as determined at the protein level. This study is a first proof of concept that γ-PGA and γ-PGA/Ch NCs promote recovery of IVD native matrix, opening new perspectives on the development of alternative therapeutic approaches for IVD degeneration.
Collapse
Affiliation(s)
- Joana C Antunes
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias s/n, Porto, 4200-465, Portugal
| | - Catarina Leite Pereira
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Graciosa Q Teixeira
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Ricardo V Silva
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
| | - Joana Caldeira
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
| | - Sibylle Grad
- AO Research Institute, Davos, Clavadelerstrasse 8, Davos, 7270, Switzerland
| | - Raquel M Gonçalves
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
| | - Mário A Barbosa
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal.
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal.
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal.
| |
Collapse
|
30
|
Boddupalli A, Zhu L, Bratlie KM. Methods for Implant Acceptance and Wound Healing: Material Selection and Implant Location Modulate Macrophage and Fibroblast Phenotypes. Adv Healthc Mater 2016; 5:2575-2594. [PMID: 27593734 DOI: 10.1002/adhm.201600532] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 06/17/2016] [Indexed: 12/12/2022]
Abstract
This review focuses on materials and methods used to induce phenotypic changes in macrophages and fibroblasts. Herein, we give a brief overview on how changes in macrophages and fibroblasts phenotypes are critical biomarkers for identification of implant acceptance, wound healing effectiveness, and are also essential for evaluating the regenerative capabilities of some hybrid strategies that involve the combination of natural and synthetic materials. The different types of cells present during the host response have been extensively studied for evaluating the reaction to different materials and there are varied material approaches towards fabrication of biocompatible substrates. We discuss how natural and synthetic materials have been used to engineer desirable outcomes in lung, heart, liver, skin, and musculoskeletal implants, and how certain properties such as rigidity, surface shape, and porosity play key roles in the progression of the host response. Several fabrication strategies are discussed to control the phenotype of infiltrating macrophages and fibroblasts: decellularization of scaffolds, surface coatings, implant shape, and pore size apart from biochemical signaling pathways that can inhibit or accelerate unfavorable host responses. It is essential to factor all the different design principles and material fabrication criteria for evaluating the choice of implant materials or regenerative therapeutic strategies.
Collapse
Affiliation(s)
- Anuraag Boddupalli
- Department of Chemical & Biological Engineering; Iowa State University; 2114 Sweeney Hall Ames IA 50011 USA
| | - Lida Zhu
- Department of Chemical & Biological Engineering; Iowa State University; 2114 Sweeney Hall Ames IA 50011 USA
| | - Kaitlin M. Bratlie
- Department of Chemical & Biological Engineering; Iowa State University; 2114 Sweeney Hall Ames IA 50011 USA
- Department of Materials Science & Engineering; Iowa State University; 2220 Hoover Hall Ames IA 50011 USA
- Division of Materials Science & Engineering; Ames National Laboratory; 126 Metals Development Ames IA 50011 USA
| |
Collapse
|
31
|
Teixeira GQ, Leite Pereira C, Castro F, Ferreira JR, Gomez-Lazaro M, Aguiar P, Barbosa MA, Neidlinger-Wilke C, Goncalves RM. Anti-inflammatory Chitosan/Poly-γ-glutamic acid nanoparticles control inflammation while remodeling extracellular matrix in degenerated intervertebral disc. Acta Biomater 2016; 42:168-179. [PMID: 27321188 DOI: 10.1016/j.actbio.2016.06.013] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 05/27/2016] [Accepted: 06/09/2016] [Indexed: 01/07/2023]
Abstract
UNLABELLED Intervertebral disc (IVD) degeneration is one of the most common causes of low back pain (LBP), the leading disorder in terms of years lived with disability. Inflammation can play a role in LPB, while impairs IVD regeneration. In spite of this, different inflammatory targets have been purposed in the context of IVD regeneration. Anti-inflammatory nanoparticles (NPs) of Chitosan and Poly-(γ-glutamic acid) with a non-steroidal anti-inflammatory drug, diclofenac (Df), were previously shown to counteract a pro-inflammatory response of human macrophages. Here, the effect of intradiscal injection of Df-NPs in degenerated IVD was evaluated. For that, Df-NPs were injected in a bovine IVD organ culture in pro-inflammatory/degenerative conditions, upon stimulation with needle-puncture and interleukin (IL)-1β. Df-NPs were internalized by IVD cells, down-regulating IL-6, IL-8, MMP1 and MMP3, and decreasing PGE2 production, compared with IL-1β-stimulated IVD punches. Interestingly, at the same time, Df-NPs promoted an up-regulation of extracellular matrix (ECM) proteins, namely collagen type II and aggrecan. Allover, this study suggests that IVD treatment with Df-NPs not only reduces inflammation, but also delays and/or decreases ECM degradation, opening perspectives to new intradiscal therapies for IVD degeneration, based on the modulation of inflammation. STATEMENT OF SIGNIFICANCE Degeneration of the IVD is an age-related progressive process considered to be the major cause of spine disorders. The pro-inflammatory environment and biomechanics of the degenerated IVD is a challenge for regenerative therapies. The novelty of this work is the intradiscal injection of an anti-inflammatory therapy based on Chitosan (Ch)/Poly-(γ-glutamic acid) (γ-PGA) nanoparticles (NPs) with an anti-inflammatory drug (diclofenac, Df), previously developed by us. This drug delivery system was tested in a pro-inflammatory/degenerative intervertebral disc ex vivo model. The main findings support the success of an anti-inflammatory therapy for degenerated IVD that not only reduces inflammation but also promotes native IVD matrix production.
Collapse
Affiliation(s)
- Graciosa Q Teixeira
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Institute of Orthopaedic Research and Biomechanics, University of Ulm, Helmholtzstrasse 14, 89081 Ulm, Germany; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Catarina Leite Pereira
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Flávia Castro
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Joana R Ferreira
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Maria Gomez-Lazaro
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Paulo Aguiar
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Mário A Barbosa
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Cornelia Neidlinger-Wilke
- Institute of Orthopaedic Research and Biomechanics, University of Ulm, Helmholtzstrasse 14, 89081 Ulm, Germany
| | - Raquel M Goncalves
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal.
| |
Collapse
|
32
|
Ruocco N, Costantini S, Guariniello S, Costantini M. Polysaccharides from the Marine Environment with Pharmacological, Cosmeceutical and Nutraceutical Potential. Molecules 2016; 21:molecules21050551. [PMID: 27128892 PMCID: PMC6273702 DOI: 10.3390/molecules21050551] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/18/2016] [Accepted: 04/22/2016] [Indexed: 01/29/2023] Open
Abstract
Carbohydrates, also called saccharides, are molecules composed of carbon, hydrogen, and oxygen. They are the most abundant biomolecules and essential components of many natural products and have attracted the attention of researchers because of their numerous human health benefits. Among carbohydrates the polysaccharides represent some of the most abundant bioactive substances in marine organisms. In fact, many marine macro- and microorganisms are good resources of carbohydrates with diverse applications due to their biofunctional properties. By acting on cell proliferation and cycle, and by modulating different metabolic pathways, marine polysaccharides (including mainly chitin, chitosan, fucoidan, carrageenan and alginate) also have numerous pharmaceutical activities, such as antioxidative, antibacterial, antiviral, immuno-stimulatory, anticoagulant and anticancer effects. Moreover, these polysaccharides have many general beneficial effects for human health, and have therefore been developed into potential cosmeceuticals and nutraceuticals. In this review we describe current advances in the development of marine polysaccharides for nutraceutical, cosmeceutical and pharmacological applications. Research in this field is opening new doors for harnessing the potential of marine natural products.
Collapse
Affiliation(s)
- Nadia Ruocco
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Napoli, Italy.
- Department of Biology, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, Via Cinthia, 80126 Napoli, Italy.
- Bio-Organic Chemistry Unit, Institute of Biomolecular Chemistry-CNR, Via Campi Flegrei 34, Pozzuoli, 80078 Naples, Italy.
| | - Susan Costantini
- CROM, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, 80131 Napoli, Italy.
| | - Stefano Guariniello
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università degli Studi di Napoli, 80131 Napoli, Italy.
| | - Maria Costantini
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Napoli, Italy.
| |
Collapse
|
33
|
Teixeira GQ, Boldt A, Nagl I, Pereira CL, Benz K, Wilke HJ, Ignatius A, Barbosa MA, Gonçalves RM, Neidlinger-Wilke C. A Degenerative/Proinflammatory Intervertebral Disc Organ Culture: An Ex Vivo Model for Anti-inflammatory Drug and Cell Therapy. Tissue Eng Part C Methods 2015; 22:8-19. [PMID: 26565141 DOI: 10.1089/ten.tec.2015.0195] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Resolution of intervertebral disc (IVD) degeneration-associated inflammation is a prerequisite for tissue regeneration and could possibly be achieved by strategies ranging from pharmacological to cell-based therapies. In this study, a proinflammatory disc organ culture model was established. Bovine caudal disc punches were needle punctured and additionally stimulated with lipopolysaccharide (10 μg/mL) or interleukin-1β (IL-1β, 10-100 ng/mL) for 48 h. Two intradiscal therapeutic approaches were tested: (i) a nonsteroidal anti-inflammatory drug, diclofenac (Df) and (ii) human mesenchymal stem/stromal cells (MSCs) embedded in an albumin/hyaluronan hydrogel. IL-1β-treated disc organ cultures showed a statistically significant upregulation of proinflammatory markers (IL-6, IL-8, prostaglandin E2 [PGE2]) and metalloproteases (MMP1, MMP3) expression, while extracellular matrix (ECM) proteins (collagen II, aggrecan) were significantly downregulated. The injection of the anti-inflammatory drug, Df, was able to reduce the levels of proinflammatory cytokines and MMPs and surprisingly increase ECM protein levels. These results point the intradiscal application of anti-inflammatory drugs as promising therapeutics for disc degeneration. In parallel, the immunomodulatory role of MSCs on this model was also evaluated. Although a slight downregulation of IL-6 and IL-8 expression could be found, the variability among the five donors tested was high, suggesting that the beneficial effect of these cells on disc degeneration needs to be further evaluated. The proinflammatory/degenerative IVD organ culture model established can be considered a suitable approach for testing novel therapeutic drugs, thus reducing the number of animals in in vivo experimentation. Moreover, this model can be used to address the cellular and molecular mechanisms that regulate inflammation in the IVD and their implications in tissue degeneration.
Collapse
Affiliation(s)
- Graciosa Q Teixeira
- 1 Institute of Orthopaedic Research and Biomechanics, Center for Musculoskeletal Research, University of Ulm , Ulm, Germany .,2 Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto, Portugal .,3 Instituto de Engenharia Biomédica (INEB), Universidade do Porto , Porto, Portugal .,4 Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto , Porto, Portugal
| | - Antje Boldt
- 1 Institute of Orthopaedic Research and Biomechanics, Center for Musculoskeletal Research, University of Ulm , Ulm, Germany
| | - Ines Nagl
- 1 Institute of Orthopaedic Research and Biomechanics, Center for Musculoskeletal Research, University of Ulm , Ulm, Germany
| | - Catarina Leite Pereira
- 2 Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto, Portugal .,3 Instituto de Engenharia Biomédica (INEB), Universidade do Porto , Porto, Portugal .,4 Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto , Porto, Portugal
| | - Karin Benz
- 5 Natural and Medical Sciences Institute (NMI), University of Tuebingen , Reutlingen, Germany
| | - Hans-Joachim Wilke
- 1 Institute of Orthopaedic Research and Biomechanics, Center for Musculoskeletal Research, University of Ulm , Ulm, Germany
| | - Anita Ignatius
- 1 Institute of Orthopaedic Research and Biomechanics, Center for Musculoskeletal Research, University of Ulm , Ulm, Germany
| | - Mário A Barbosa
- 2 Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto, Portugal .,3 Instituto de Engenharia Biomédica (INEB), Universidade do Porto , Porto, Portugal .,4 Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto , Porto, Portugal
| | - Raquel M Gonçalves
- 2 Instituto de Investigação e Inovação em Saúde, Universidade do Porto , Porto, Portugal .,3 Instituto de Engenharia Biomédica (INEB), Universidade do Porto , Porto, Portugal
| | - Cornelia Neidlinger-Wilke
- 1 Institute of Orthopaedic Research and Biomechanics, Center for Musculoskeletal Research, University of Ulm , Ulm, Germany
| |
Collapse
|