1
|
Yadav VK, Gupta R, Assiri AA, Uddin J, Ishaqui AA, Kumar P, Orayj KM, Tahira S, Patel A, Choudhary N. Role of Nanotechnology in Ischemic Stroke: Advancements in Targeted Therapies and Diagnostics for Enhanced Clinical Outcomes. J Funct Biomater 2025; 16:8. [PMID: 39852564 PMCID: PMC11766075 DOI: 10.3390/jfb16010008] [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: 11/13/2024] [Revised: 12/27/2024] [Accepted: 12/28/2024] [Indexed: 01/26/2025] Open
Abstract
Each year, the number of cases of strokes and deaths due to this is increasing around the world. This could be due to work stress, lifestyles, unhealthy food habits, and several other reasons. Currently, there are several traditional methods like thrombolysis and mechanical thrombectomy for managing strokes. The current approach has several limitations, like delayed diagnosis, limited therapeutic delivery, and risks of secondary injuries. So, there is a need for some effective and reliable methods for the management of strokes, which could help in early diagnosis followed by the treatment of strokes. Nanotechnology has played an immense role in managing strokes, and recently, it has emerged as a transformative solution offering innovative diagnostic tools and therapeutic strategies. Nanoparticles (NPs) belonging to several classes, including metallic (metallic and metal oxide), organic (lipids, liposome), and carbon, can cross the blood-brain barrier and may exhibit immense potential for managing various strokes. Moreover, these NPs have exhibited promise in improving imaging specificity and therapeutic delivery by precise drug delivery and real-time monitoring of treatment efficacy. Nanomaterials like cerium oxide (CeO2) and liposome-encapsulated agents have neuroprotective properties that reduce oxidative stress and promote neuroregeneration. In the present article, the authors have emphasized the significant advancements in the nanomedicine management of stroke, including NPs-based drug delivery systems, neuroprotective and neuroregenerative therapies, and multimodal imaging advancements.
Collapse
Affiliation(s)
- Virendra Kumar Yadav
- Marwadi University Research Center, Department of Microbiology, Faculty of Sciences, Marwadi University, Rajkot 360003, Gujarat, India
| | - Rachna Gupta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382021, Gujarat, India;
| | - Abdullah A. Assiri
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha 61441, Saudi Arabia; (A.A.A.); (A.A.I.); (K.M.O.)
| | - Jalal Uddin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 61441, Saudi Arabia;
| | - Azfar A. Ishaqui
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha 61441, Saudi Arabia; (A.A.A.); (A.A.I.); (K.M.O.)
| | - Pankaj Kumar
- Department of Environmental Science, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, Gujarat, India;
| | - Khalid M. Orayj
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha 61441, Saudi Arabia; (A.A.A.); (A.A.I.); (K.M.O.)
| | - Shazia Tahira
- Institute of Professional Psychology, Bahria University Karachi Campus, Karachi 75260, Pakistan;
- Department of Psychiatry, Jinnah Postgraduate Medical Centre, Karachi 75510, Pakistan
| | - Ashish Patel
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan 384265, Gujarat, India;
| | - Nisha Choudhary
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan 384265, Gujarat, India;
- Department of Lifesciences, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, Gujarat, India
| |
Collapse
|
2
|
Xiong Y, Mi BB, Shahbazi MA, Xia T, Xiao J. Microenvironment-responsive nanomedicines: a promising direction for tissue regeneration. Mil Med Res 2024; 11:69. [PMID: 39434177 PMCID: PMC11492517 DOI: 10.1186/s40779-024-00573-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/29/2024] [Indexed: 10/23/2024] Open
Abstract
Severe tissue defects present formidable challenges to human health, persisting as major contributors to mortality rates. The complex pathological microenvironment, particularly the disrupted immune landscape within these defects, poses substantial hurdles to existing tissue regeneration strategies. However, the emergence of nanobiotechnology has opened a new direction in immunomodulatory nanomedicine, providing encouraging prospects for tissue regeneration and restoration. This review aims to gather recent advances in immunomodulatory nanomedicine to foster tissue regeneration. We begin by elucidating the distinctive features of the local immune microenvironment within defective tissues and its crucial role in tissue regeneration. Subsequently, we explore the design and functional properties of immunomodulatory nanosystems. Finally, we address the challenges and prospects of clinical translation in nanomedicine development, aiming to propose a potent approach to enhance tissue regeneration through synergistic immune modulation and nanomedicine integration.
Collapse
Affiliation(s)
- Yuan Xiong
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bo-Bin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Mohammad-Ali Shahbazi
- Department of Biomaterials and Biomedical Technology, Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands.
| | - Tian Xia
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| | - Jun Xiao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
3
|
Cai W, Sun T, Qiu C, Sheng H, Chen R, Xie C, Kou L, Yao Q. Stable triangle: nanomedicine-based synergistic application of phototherapy and immunotherapy for tumor treatment. J Nanobiotechnology 2024; 22:635. [PMID: 39420366 PMCID: PMC11488210 DOI: 10.1186/s12951-024-02925-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 10/10/2024] [Indexed: 10/19/2024] Open
Abstract
In recent decades, cancer has posed a challenging obstacle that humans strive to overcome. While phototherapy and immunotherapy are two emerging therapies compared to traditional methods, they each have their advantages and limitations. These limitations include easy metastasis and recurrence, low response rates, and strong side effects. To address these issues, researchers have increasingly focused on combining these two therapies by utilizing a nano-drug delivery system due to its superior targeting effect and high drug loading rate, yielding remarkable results. The combination therapy demonstrates enhanced response efficiency and effectiveness, leading to a preparation that is highly targeted, responsive, and with low recurrence rates. This paper reviews several main mechanisms of anti-tumor effects observed in combination therapy based on the nano-drug delivery system over the last five years. Furthermore, the challenges and future prospects of this combination therapy are also discussed.
Collapse
Affiliation(s)
- Wenjing Cai
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325027, China
| | - Tuyue Sun
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Chenyu Qiu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325027, China
| | - Huixiang Sheng
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325027, China
| | - Congying Xie
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, Wenzhou, 325000, China.
- Zhejiang-Hong Kong Precision Theranostics of Thoracic Tumors Joint Laboratory, Wenzhou, 325000, China.
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325027, China.
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, Wenzhou, 325000, China.
- Zhejiang-Hong Kong Precision Theranostics of Thoracic Tumors Joint Laboratory, Wenzhou, 325000, China.
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
4
|
de Carvalho Lima EN, Octaviano ALM, Piqueira JRC, Diaz RS, Justo JF. Coronavirus and Carbon Nanotubes: Seeking Immunological Relationships to Discover Immunotherapeutic Possibilities. Int J Nanomedicine 2022; 17:751-781. [PMID: 35241912 PMCID: PMC8887185 DOI: 10.2147/ijn.s341890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/31/2022] [Indexed: 12/11/2022] Open
Abstract
Since December 2019, the world has faced an unprecedented pandemic crisis due to a new coronavirus disease, coronavirus disease-2019 (COVID-19), which has instigated intensive studies on prevention and treatment possibilities. Here, we investigate the relationships between the immune activation induced by three coronaviruses associated with recent outbreaks, with special attention to SARS-CoV-2, the causative agent of COVID-19, and the immune activation induced by carbon nanotubes (CNTs) to understand the points of convergence in immune induction and modulation. Evidence suggests that CNTs are among the most promising materials for use as immunotherapeutic agents. Therefore, this investigation explores new possibilities of effective immunotherapies for COVID-19. This study aimed to raise interest and knowledge about the use of CNTs as immunotherapeutic agents in coronavirus treatment. Thus, we summarize the most important immunological aspects of various coronavirus infections and describe key advances and challenges in using CNTs as immunotherapeutic agents against viral infections and the activation of the immune response induced by CNTs, which can shed light on the immunotherapeutic possibilities of CNTs.
Collapse
Affiliation(s)
- Elidamar Nunes de Carvalho Lima
- Telecommunication and Control Engineering Department, Polytechnic School of the University of São Paulo, São Paulo, Brazil
- Infectious Diseases Division, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
- Electronic Systems Engineering Department, Polytechnic School of the University of São Paulo, São Paulo, SP, CEP 05508-010, Brazil
| | - Ana Luiza Moraes Octaviano
- Telecommunication and Control Engineering Department, Polytechnic School of the University of São Paulo, São Paulo, Brazil
| | - José Roberto Castilho Piqueira
- Telecommunication and Control Engineering Department, Polytechnic School of the University of São Paulo, São Paulo, Brazil
| | - Ricardo Sobhie Diaz
- Infectious Diseases Division, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - João Francisco Justo
- Electronic Systems Engineering Department, Polytechnic School of the University of São Paulo, São Paulo, SP, CEP 05508-010, Brazil
| |
Collapse
|
5
|
Wei Q, Su Y, Xin H, Zhang L, Ding J, Chen X. Immunologically Effective Biomaterials. ACS APPLIED MATERIALS & INTERFACES 2021; 13:56719-56724. [PMID: 34797622 DOI: 10.1021/acsami.1c14781] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Immunoregulation represents a booming field of biomaterial design. The unique physical and chemical properties of biomaterials offer tremendous opportunities for development. Each of their parameters exerts immunogenic effects at the immune system, cellular, and molecular levels. Herein, the perspective summarizes the interaction of biomaterials with immune cells and the underlying mechanisms to control immunoregulation in a top-down manner, providing solid inspiration for biomedical applications of immunologically effective biomaterials.
Collapse
Affiliation(s)
- Qi Wei
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun 130033, P. R. China
| | - Yuanzhen Su
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, P. R. China
| | - Hua Xin
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun 130033, P. R. China
| | - Lening Zhang
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun 130033, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, 220 Handan Road, Shanghai 200433, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, P. R. China
| |
Collapse
|
6
|
Nkune NW, Simelane NWN, Montaseri H, Abrahamse H. Photodynamic Therapy-Mediated Immune Responses in Three-Dimensional Tumor Models. Int J Mol Sci 2021; 22:12618. [PMID: 34884424 PMCID: PMC8657498 DOI: 10.3390/ijms222312618] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022] Open
Abstract
Photodynamic therapy (PDT) is a promising non-invasive phototherapeutic approach for cancer therapy that can eliminate local tumor cells and produce systemic antitumor immune responses. In recent years, significant efforts have been made in developing strategies to further investigate the immune mechanisms triggered by PDT. The majority of in vitro experimental models still rely on the two-dimensional (2D) cell cultures that do not mimic a three-dimensional (3D) cellular environment in the human body, such as cellular heterogeneity, nutrient gradient, growth mechanisms, and the interaction between cells as well as the extracellular matrix (ECM) and therapeutic resistance to anticancer treatments. In addition, in vivo animal studies are highly expensive and time consuming, which may also show physiological discrepancies between animals and humans. In this sense, there is growing interest in the utilization of 3D tumor models, since they precisely mimic different features of solid tumors. This review summarizes the characteristics and techniques for 3D tumor model generation. Furthermore, we provide an overview of innate and adaptive immune responses induced by PDT in several in vitro and in vivo tumor models. Future perspectives are highlighted for further enhancing PDT immune responses as well as ideal experimental models for antitumor immune response studies.
Collapse
Affiliation(s)
| | | | | | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Johannesburg 2028, South Africa; (N.W.N.); (N.W.N.S.); (H.M.)
| |
Collapse
|
7
|
Ulloa LS, Perissinotto F, Rago I, Goldoni A, Santoro R, Pesce M, Casalis L, Scaini D. Carbon Nanotubes Substrates Alleviate Pro-Calcific Evolution in Porcine Valve Interstitial Cells. NANOMATERIALS 2021; 11:nano11102724. [PMID: 34685165 PMCID: PMC8538037 DOI: 10.3390/nano11102724] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 01/19/2023]
Abstract
The quest for surfaces able to interface cells and modulate their functionality has raised, in recent years, the development of biomaterials endowed with nanocues capable of mimicking the natural extracellular matrix (ECM), especially for tissue regeneration purposes. In this context, carbon nanotubes (CNTs) are optimal candidates, showing dimensions and a morphology comparable to fibril ECM constituents. Moreover, when immobilized onto surfaces, they demonstrated outstanding cytocompatibility and ease of chemical modification with ad hoc functionalities. In this study, we interface porcine aortic valve interstitial cells (pVICs) to multi-walled carbon nanotube (MWNT) carpets, investigating the impact of surface nano-morphology on cell properties. The results obtained indicate that CNTs significantly affect cell behavior in terms of cell morphology, cytoskeleton organization, and mechanical properties. We discovered that CNT carpets appear to maintain interfaced pVICs in a sort of “quiescent state”, hampering cell activation into a myofibroblasts-like phenotype morphology, a cellular evolution prodromal to Calcific Aortic Valve Disease (CAVD) and characterized by valve interstitial tissue stiffening. We found that this phenomenon is linked to CNTs’ ability to alter cell tensional homeostasis, interacting with cell plasma membranes, stabilizing focal adhesions and enabling a better strain distribution within cells. Our discovery contributes to shedding new light on the ECM contribution in modulating cell behavior and will open the door to new criteria for designing nanostructured scaffolds to drive cell functionality for tissue engineering applications.
Collapse
Affiliation(s)
- Luisa Severino Ulloa
- Dipartimento di Fisica, Università di Trieste, Piazzale Europa 1, 34127 Trieste, Italy; (L.S.U.); (F.P.); (I.R.)
| | - Fabio Perissinotto
- Dipartimento di Fisica, Università di Trieste, Piazzale Europa 1, 34127 Trieste, Italy; (L.S.U.); (F.P.); (I.R.)
| | - Ilaria Rago
- Dipartimento di Fisica, Università di Trieste, Piazzale Europa 1, 34127 Trieste, Italy; (L.S.U.); (F.P.); (I.R.)
| | - Andrea Goldoni
- Elettra-Sincrotrone Trieste S.C.p.A., Basovizza, 34149 Trieste, Italy;
| | - Rosaria Santoro
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (R.S.); (M.P.)
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (R.S.); (M.P.)
| | - Loredana Casalis
- Elettra-Sincrotrone Trieste S.C.p.A., Basovizza, 34149 Trieste, Italy;
- Correspondence: (L.C.); (D.S.)
| | - Denis Scaini
- Area di Neuroscienze, Scuola Internazionale Superiore di Studi Avanzati, Via Bonomea 265, 34136 Trieste, Italy
- Faculty of Medicine, Imperial College London, London W12 0NN, UK
- Correspondence: (L.C.); (D.S.)
| |
Collapse
|
8
|
de Carvalho Lima EN, Diaz RS, Justo JF, Castilho Piqueira JR. Advances and Perspectives in the Use of Carbon Nanotubes in Vaccine Development. Int J Nanomedicine 2021; 16:5411-5435. [PMID: 34408416 PMCID: PMC8367085 DOI: 10.2147/ijn.s314308] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 07/21/2021] [Indexed: 12/15/2022] Open
Abstract
Advances in nanobiotechnology have allowed the utilization of nanotechnology through nanovaccines. Nanovaccines are powerful tools for enhancing the immunogenicity of a specific antigen and exhibit advantages over other adjuvant approaches, with features such as expanded stability, prolonged release, decreased immunotoxicity, and immunogenic selectivity. We introduce recent advances in carbon nanotubes (CNTs) to induce either a carrier effect as a nanoplatform or an immunostimulatory effect. Several studies of CNT-based nanovaccines revealed that due to the ability of CNTs to carry immunogenic molecules, they can act as nonclassical vaccines, a quality not possessed by vaccines with traditional formulations. Therefore, adapting and modifying the physicochemical properties of CNTs for use in vaccines may additionally enhance their efficacy in inducing a T cell-based immune response. Accordingly, the purpose of this study is to renew and awaken interest in and knowledge of the safe use of CNTs as adjuvants and carriers in vaccines.
Collapse
Affiliation(s)
- Elidamar Nunes de Carvalho Lima
- Telecommunication and Control Engineering Department, Polytechnic School of the University of São Paulo, São Paulo, Brazil
- Infectious Diseases Division, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Ricardo Sobhie Diaz
- Infectious Diseases Division, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - João Francisco Justo
- Electronic Systems Engineering Department, Polytechnic School of the University of São Paulo, São Paulo, Brazil
| | - José Roberto Castilho Piqueira
- Telecommunication and Control Engineering Department, Polytechnic School of the University of São Paulo, São Paulo, Brazil
| |
Collapse
|
9
|
Alagarsamy KN, Mathan S, Yan W, Rafieerad A, Sekaran S, Manego H, Dhingra S. Carbon nanomaterials for cardiovascular theranostics: Promises and challenges. Bioact Mater 2021; 6:2261-2280. [PMID: 33553814 PMCID: PMC7829079 DOI: 10.1016/j.bioactmat.2020.12.030] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/15/2020] [Accepted: 12/31/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide. Heart attack and stroke cause irreversible tissue damage. The currently available treatment options are limited to "damage-control" rather than tissue repair. The recent advances in nanomaterials have offered novel approaches to restore tissue function after injury. In particular, carbon nanomaterials (CNMs) have shown significant promise to bridge the gap in clinical translation of biomaterial based therapies. This family of carbon allotropes (including graphenes, carbon nanotubes and fullerenes) have unique physiochemical properties, including exceptional mechanical strength, electrical conductivity, chemical behaviour, thermal stability and optical properties. These intrinsic properties make CNMs ideal materials for use in cardiovascular theranostics. This review is focused on recent efforts in the diagnosis and treatment of heart diseases using graphenes and carbon nanotubes. The first section introduces currently available derivatives of graphenes and carbon nanotubes and discusses some of the key characteristics of these materials. The second section covers their application in drug delivery, biosensors, tissue engineering and immunomodulation with a focus on cardiovascular applications. The final section discusses current shortcomings and limitations of CNMs in cardiovascular applications and reviews ongoing efforts to address these concerns and to bring CNMs from bench to bedside.
Collapse
Affiliation(s)
- Keshav Narayan Alagarsamy
- Regenerative Medicine Program, Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Sajitha Mathan
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), Department of Bioengineering, School of Chemical and Biotechnology, SASTRA University, Thanjavur, 613 401, Tamil Nadu, India
| | - Weiang Yan
- Regenerative Medicine Program, Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
- Section of Cardiac Surgery, Department of Surgery, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Alireza Rafieerad
- Regenerative Medicine Program, Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Saravanan Sekaran
- Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), Department of Bioengineering, School of Chemical and Biotechnology, SASTRA University, Thanjavur, 613 401, Tamil Nadu, India
| | - Hanna Manego
- Regenerative Medicine Program, Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Sanjiv Dhingra
- Regenerative Medicine Program, Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
10
|
Nanotechnology Facilitated Cultured Neuronal Network and Its Applications. Int J Mol Sci 2021; 22:ijms22115552. [PMID: 34074027 PMCID: PMC8197344 DOI: 10.3390/ijms22115552] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 12/16/2022] Open
Abstract
The development of a biomimetic neuronal network from neural cells is a big challenge for researchers. Recent advances in nanotechnology, on the other hand, have enabled unprecedented tools and techniques for guiding and directing neural stem cell proliferation and differentiation in vitro to construct an in vivo-like neuronal network. Nanotechnology allows control over neural stem cells by means of scaffolds that guide neurons to reform synaptic networks in suitable directions in 3D architecture, surface modification/nanopatterning to decide cell fate and stimulate/record signals from neurons to find out the relationships between neuronal circuit connectivity and their pathophysiological functions. Overall, nanotechnology-mediated methods facilitate precise physiochemical controls essential to develop tools appropriate for applications in neuroscience. This review emphasizes the newest applications of nanotechnology for examining central nervous system (CNS) roles and, therefore, provides an insight into how these technologies can be tested in vitro before being used in preclinical and clinical research and their potential role in regenerative medicine and tissue engineering.
Collapse
|
11
|
Hadidi N, Sharifnia Z, Eteghadi A, Shokrgozar MA, Mosaffa N. PEGylated single-walled carbon nanotubes as co-adjuvants enhance expression of maturation markers in monocyte-derived dendritic cells. Nanomedicine (Lond) 2021; 16:171-188. [PMID: 33560153 DOI: 10.2217/nnm-2020-0339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Aim: This study investigated the application of phospholipid-PEGylated single-walled carbon nanotubes (PL-PEG-SWCNTs) as a safe co-adjuvant for the commercial recombinant hepatitis B virus vaccine to enhance induction of monocyte-derived dendritic cells (MDDCs) differentiation and activation in vitro as an immune response initiator cell to prompt a long-term immune response after a single dose injection. Methods: Immature MDDCs were exposed to PL-PEG-SWCNTs alone and in combination with hepatitis B vaccine. Results & conclusion: Study results confirm the enhanced expression of maturation markers in human immature MDDCs after PL-PEG-SWCNT exposure. The results suggest that PL-PEG-SWCNT is an efficient co-adjuvant for the commercial recombinant hepatitis B virus vaccine to enhance dendritic cell response stimulation in vitro.
Collapse
Affiliation(s)
- Naghmeh Hadidi
- Department of Clinical Research & EM Microscope, Pasteur Institute of Iran (PII), Tehran 1316943551, Iran
| | - Zarin Sharifnia
- Department of Clinical Research & EM Microscope, Pasteur Institute of Iran (PII), Tehran 1316943551, Iran
| | - Atefeh Eteghadi
- Department of Clinical Research & EM Microscope, Pasteur Institute of Iran (PII), Tehran 1316943551, Iran
| | | | - Nariman Mosaffa
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 198396-3113, Iran
| |
Collapse
|
12
|
Mia MB, Saxena RK. Poly dispersed acid-functionalized single walled carbon nanotubes target activated T and B cells to suppress acute and chronic GVHD in mouse model. Immunol Lett 2020; 224:30-37. [PMID: 32504776 DOI: 10.1016/j.imlet.2020.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/05/2020] [Accepted: 05/12/2020] [Indexed: 12/27/2022]
Abstract
Graft versus host disease (GVHD) results from hyper-activation of transplanted lymphocytes against the host antigens. Bone marrow transplantation in humans as well as some cases of blood transfusion and organ transplantation are associated with a strong GVH reaction resulting in GVHD that in many cases may be fatal. We had previously shown that poly-dispersed acid-functionalized single-walled carbon nanotubes (AF-SWCNTs) specifically target activated T and B lymphocytes and kill them. In the present study, efficacy of AF-SWCNTs to suppress the GVH reaction was tested in the mouse model. Acute GVHD was induced in mice by administering intravenously 30 or 60 million spleen cells from a parental strain (C57bl/6 mouse, MHC haplotype H-2b) to host (C57bl/6 x Balb/c) F1 mice (MHC haplotype H-2b/d)and waiting for 8-10 days. Chronic GVHD was similarly induced by administration of 30 million parent spleen cells to F1 mice and waiting for a period of 60 days. Our results demonstrate a marked decline in splenomegaly and recovery of spleen T (both CD4 and CD8) and B cells in GVHD mice treated with AF-SWCNTs. AF-SWCNTs treatment also limited T and B cell proliferation by restricting S-phage of cell cycle. Generation of anti-host cytotoxic T cells (CTLs) was also markedly suppressed by AF-SWCNT treatment of acute GVHD mice, and a significant reduction in the generation of anti-host antibodies could also be demonstrated. Taken together, our results suggest that the AF-SWCNTs can be considered as a potential therapeutic agent for treating GVHD.
Collapse
Affiliation(s)
- Md Babu Mia
- Faculty of Life Sciences and Biotechnology, South Asian University, Akbar Bhawan, Chanakyapuri, New Delhi, 110021, India
| | - Rajiv K Saxena
- Faculty of Life Sciences and Biotechnology, South Asian University, Akbar Bhawan, Chanakyapuri, New Delhi, 110021, India.
| |
Collapse
|
13
|
Shields CW, Wang LLW, Evans MA, Mitragotri S. Materials for Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901633. [PMID: 31250498 DOI: 10.1002/adma.201901633] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/17/2019] [Indexed: 05/20/2023]
Abstract
Breakthroughs in materials engineering have accelerated the progress of immunotherapy in preclinical studies. The interplay of chemistry and materials has resulted in improved loading, targeting, and release of immunomodulatory agents. An overview of the materials that are used to enable or improve the success of immunotherapies in preclinical studies is presented, from immunosuppressive to proinflammatory strategies, with particular emphasis on technologies poised for clinical translation. The materials are organized based on their characteristic length scale, whereby the enabling feature of each technology is organized by the structure of that material. For example, the mechanisms by which i) nanoscale materials can improve targeting and infiltration of immunomodulatory payloads into tissues and cells, ii) microscale materials can facilitate cell-mediated transport and serve as artificial antigen-presenting cells, and iii) macroscale materials can form the basis of artificial microenvironments to promote cell infiltration and reprogramming are discussed. As a step toward establishing a set of design rules for future immunotherapies, materials that intrinsically activate or suppress the immune system are reviewed. Finally, a brief outlook on the trajectory of these systems and how they may be improved to address unsolved challenges in cancer, infectious diseases, and autoimmunity is presented.
Collapse
Affiliation(s)
- C Wyatt Shields
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - Lily Li-Wen Wang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Michael A Evans
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
14
|
Bhargava A, Mishra DK, Tiwari R, Lohiya NK, Goryacheva IY, Mishra PK. Immune cell engineering: opportunities in lung cancer therapeutics. Drug Deliv Transl Res 2020; 10:1203-1227. [PMID: 32172351 DOI: 10.1007/s13346-020-00719-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Engineered immune cells offer a prime therapeutic alternate for some aggressive and frequently occurring malignancies like lung cancer. These therapies were reported to result in tumor regression and overall improvement in patient survival. However, studies also suggest that the presence of cancer cell-induced immune-suppressive microenvironment, off-target toxicity, and difficulty in concurrent imaging are some prime impendent in the success of these approaches. The present article reviews the need and significance of the currently available immune cell-based strategies for lung cancer therapeutics. It also showcases the utility of incorporating nanoengineered strategies and details the available formulations of nanocarriers. In last, it briefly discussed the existing methods for nanoparticle fuctionalization and challenges in translating basic research to the clinics. Graphical Abstract.
Collapse
Affiliation(s)
- Arpit Bhargava
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Kamla Nehru Hospital,, Building (Gandhi Medical College Campus), Bhopal, Madhya Pradesh, 462001, India
| | | | - Rajnarayan Tiwari
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Kamla Nehru Hospital,, Building (Gandhi Medical College Campus), Bhopal, Madhya Pradesh, 462001, India
| | | | - Irina Yu Goryacheva
- Department of General and Inorganic Chemistry, Saratov State University, Saratov, Russian Federation
| | - Pradyumna Kumar Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Kamla Nehru Hospital,, Building (Gandhi Medical College Campus), Bhopal, Madhya Pradesh, 462001, India.
| |
Collapse
|
15
|
Rezaei R, Safaei M, Mozaffari HR, Moradpoor H, Karami S, Golshah A, Salimi B, Karami H. The Role of Nanomaterials in the Treatment of Diseases and Their Effects on the Immune System. Open Access Maced J Med Sci 2019; 7:1884-1890. [PMID: 31316678 PMCID: PMC6614262 DOI: 10.3889/oamjms.2019.486] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 12/12/2022] Open
Abstract
Nanotechnology has been widely exploited in recent years in various applications. Different sectors of medicine and treatment have also focused on the use of nanoproducts. One of the areas of interest in the treatment measures is the interaction between nanomaterials and immune system components. Engineered nanomaterials can stimulate the inhibition or enhancement of immune responses and prevent the detection ability of the immune system. Changes in immune function, in addition to the benefits, may also lead to some damage. Therefore, adequate assessment of the novel nanomaterials seems to be necessary before practical use in treatment. However, there is little information on the toxicological and biological effects of nanomaterials, especially on the potential ways of contacting and handling nanomaterials in the body and the body response to these materials. Extensive variation and different properties of nanomaterials have made it much more difficult to access their toxicological effects to the present. The present study aims to raise knowledge about the potential benefits and risks of using the nanomaterials on the immune system to design and safely employ these compounds in therapeutic purposes.
Collapse
Affiliation(s)
- Razieh Rezaei
- Advanced Dental Sciences Research Laboratory, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Medical Biology Research Centre, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohsen Safaei
- Advanced Dental Sciences Research Laboratory, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hamid Reza Mozaffari
- Medical Biology Research Centre, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Oral and Maxillofacial Medicine, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hedaiat Moradpoor
- Department of Prosthodontics, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sara Karami
- Students Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amin Golshah
- Department of Orthodontics, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Behroz Salimi
- School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hossein Karami
- School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
16
|
Wan Z, Shaheen S, Chau A, Zeng Y, Liu W. Imaging: Gear up for mechano-immunology. Cell Immunol 2019; 350:103926. [PMID: 31151736 DOI: 10.1016/j.cellimm.2019.103926] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 04/15/2019] [Accepted: 05/15/2019] [Indexed: 12/17/2022]
Abstract
Immune cells including B and T lymphocytes have a remarkable ability to sense the physical perturbations through their surface expressed receptors. At the advent of modern imaging technologies paired with biophysical methods, we have gained the understanding of mechanical forces exerted by immune cells to perform their functions. This review will go over the imaging techniques already being used to study mechanical forces in immune cells. We will also discuss the dire need for new modern technologies for future work.
Collapse
Affiliation(s)
- Zhengpeng Wan
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Samina Shaheen
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Alicia Chau
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Yingyue Zeng
- School of Life Science, Liaoning University, Shenyang 110036, China
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing 100084, China; Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing 100084, China.
| |
Collapse
|
17
|
Wu Y, Peng Y, Bohra H, Zou J, Ranjan VD, Zhang Y, Zhang Q, Wang M. Photoconductive Micro/Nanoscale Interfaces of a Semiconducting Polymer for Wireless Stimulation of Neuron-Like Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:4833-4841. [PMID: 30624894 DOI: 10.1021/acsami.8b19631] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
We report multiscale structured fibers and patterned films based on a semiconducting polymer, poly(3-hexylthiophene) (P3HT), as photoconductive biointerfaces to promote neuronal stimulation upon light irradiation. The micro/nanoscale structures of P3HT used for neuronal interfacing and stimulation include nanofibers with an average diameter of 100 nm, microfibers with an average diameter of about 1 μm, and lithographically patterned stripes with width of 3, 25, and 50 μm, respectively. The photoconductive effect of P3HT upon light irradiation provides electrical stimulation for neuronal differentiation and directed growth. Our results demonstrate that neurons on P3HT nanofibers showed a significantly higher total number of branches, while neurons grown on P3HT microfibers had longer and thinner neurites. Such a combination strategy of topographical and photoconductive stimulation can be applied to further enhance neuronal differentiation and directed growth. These photoconductive polymeric micro/nanostructures demonstrated their great potential for neural engineering and development of novel neural regenerative devices.
Collapse
Affiliation(s)
- Yingjie Wu
- School of Chemical and Biomedical Engineering , Nanyang Technological University , 62 Nanyang Drive , Singapore 637459 , Singapore
| | - Yanfen Peng
- School of Chemical and Biomedical Engineering , Nanyang Technological University , 62 Nanyang Drive , Singapore 637459 , Singapore
| | - Hassan Bohra
- School of Chemical and Biomedical Engineering , Nanyang Technological University , 62 Nanyang Drive , Singapore 637459 , Singapore
| | - Jianping Zou
- School of Electrical and Electronic Engineering , Nanyang Technological University , 50 Nanyang Avenue , Singapore 639798 , Singapore
| | - Vivek Damodar Ranjan
- School of Mechanical & Aerospace Engineering , Nanyang Technological University , 50 Nanyang Avenue , Singapore 639798 , Singapore
| | - Yilei Zhang
- School of Mechanical & Aerospace Engineering , Nanyang Technological University , 50 Nanyang Avenue , Singapore 639798 , Singapore
| | - Qing Zhang
- School of Electrical and Electronic Engineering , Nanyang Technological University , 50 Nanyang Avenue , Singapore 639798 , Singapore
| | - Mingfeng Wang
- School of Chemical and Biomedical Engineering , Nanyang Technological University , 62 Nanyang Drive , Singapore 637459 , Singapore
| |
Collapse
|
18
|
Musto M, Rauti R, Rodrigues AF, Bonechi E, Ballerini C, Kostarelos K, Ballerini L. 3D Organotypic Spinal Cultures: Exploring Neuron and Neuroglia Responses Upon Prolonged Exposure to Graphene Oxide. Front Syst Neurosci 2019; 13:1. [PMID: 30733671 PMCID: PMC6354065 DOI: 10.3389/fnsys.2019.00001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/03/2019] [Indexed: 11/13/2022] Open
Abstract
Graphene-based nanomaterials are increasingly engineered as components of biosensors, interfaces or drug delivery platforms in neuro-repair strategies. In these developments, the mostly used derivative of graphene is graphene oxide (GO). To tailor the safe development of GO nanosheets, we need to model in vitro tissue responses, and in particular the reactivity of microglia, a sub-population of neuroglia that acts as the first active immune response, when challenged by GO. Here, we investigated central nervous system (CNS) tissue reactivity upon long-term exposure to GO nanosheets in 3D culture models. We used the mouse organotypic spinal cord cultures, ideally suited for studying long-term interference with cues delivered at controlled times and concentrations. In cultured spinal segments, the normal presence, distribution and maturation of anatomically distinct classes of neurons and resident neuroglial cells are preserved. Organotypic explants were developed for 2 weeks embedded in fibrin glue alone or presenting GO nanosheets at 10, 25 and 50 μg/mL. We addressed the impact of such treatments on premotor synaptic activity monitored by patch clamp recordings of ventral interneurons. We investigated by immunofluorescence and confocal microscopy the accompanying glial responses to GO exposure, focusing on resident microglia, tested in organotypic spinal slices and in isolated neuroglia cultures. Our results suggest that microglia reactivity to accumulation of GO flakes, maybe due to active phagocytosis, may trim down synaptic activity, although in the absence of an effective activation of inflammatory response and in the absence of neuronal cell death.
Collapse
Affiliation(s)
- Mattia Musto
- Neuron Physiology and Technology Lab, International School for Advanced Studies (SISSA), Trieste, Italy
| | - Rossana Rauti
- Neuron Physiology and Technology Lab, International School for Advanced Studies (SISSA), Trieste, Italy
| | - Artur Filipe Rodrigues
- Nanomedicine Lab, Faculty of Biology, Medicine & Health and National Graphene Institute, University of Manchester, Manchester, United Kingdom
| | - Elena Bonechi
- Department NEUROFARBA, University of Florence, Florence, Italy
| | - Clara Ballerini
- Laboratory of Neuroimmunology, Dipartimento di Medicina Sperimentale e Clinica, University of Firenze, Firenze, Italy
| | - Kostas Kostarelos
- Nanomedicine Lab, Faculty of Biology, Medicine & Health and National Graphene Institute, University of Manchester, Manchester, United Kingdom
| | - Laura Ballerini
- Neuron Physiology and Technology Lab, International School for Advanced Studies (SISSA), Trieste, Italy
| |
Collapse
|
19
|
Tomić S, Ilić N, Kokol V, Gruden-Movsesijan A, Mihajlović D, Bekić M, Sofronić-Milosavljević L, Čolić M, Vučević D. Functionalization-dependent effects of cellulose nanofibrils on tolerogenic mechanisms of human dendritic cells. Int J Nanomedicine 2018; 13:6941-6960. [PMID: 30464452 PMCID: PMC6217907 DOI: 10.2147/ijn.s183510] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Cellulose nanofibrils (CNF) are attractive nanomaterials for various biomedical applications due to their excellent biocompatibility and biomimetic properties. However, their immunoregulatory properties are insufficiently investigated, especially in relation to their functionalization, which could cause problems during their clinical application. Methods Using a model of human dendritic cells (DC), which have a central role in the regulation of immune response, we investigated how differentially functionalized CNF, ie, native (n) CNF, 2,2,6,6-tetramethylpiperidine 1-oxyl radical-oxidized (c) CNF, and 3-aminopropylphosphoric acid-functionalized (APAc) CNF, affect DC properties, their viability, morphology, differentiation and maturation potential, and the capacity to regulate T cell-mediated immune response. Results Nontoxic doses of APAcCNF displayed the strongest inhibitory effects on DC differentiation, maturation, and T helper (Th) 1 and Th17 polarization capacity, followed by cCNF and nCNF, respectively. These results correlated with a specific pattern of regulatory cytokines production by APAcCNF-DC and their increased capacity to induce suppressive CD8+CD25+IL-10+ regulatory T cells in immunoglobulin-like transcript (ILT)-3- and ILT-4- dependent manner. In contrast, nCNF-DC induced predominantly suppressive CD4+CD25hiFoxP3hi regulatory T cells in indolamine 2,3-dioxygenase-1-dependent manner. Different tolerogenic properties of CNF correlated with their size and APA functionalization, as well as with different expression of CD209 and actin bundles at the place of contact with CNF. Conclusion The capacity to induce different types of DC-mediated tolerogenic immune responses by functionalized CNF opens new perspectives for their application as well-tolerated nanomaterials in tissue engineering and novel platforms for the therapy of inflammatory T cell-mediated pathologies.
Collapse
Affiliation(s)
- Sergej Tomić
- Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia, .,Institute for Medical Research, Medical Faculty of the Military Medical Academy, University of Defense, Belgrade, Serbia,
| | - Nataša Ilić
- Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia,
| | - Vanja Kokol
- Institute of Engineering Materials and Design, University of Maribor, Maribor, Slovenia
| | | | - Dušan Mihajlović
- Institute for Medical Research, Medical Faculty of the Military Medical Academy, University of Defense, Belgrade, Serbia,
| | - Marina Bekić
- Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia,
| | | | - Miodrag Čolić
- Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia, .,Institute for Medical Research, Medical Faculty of the Military Medical Academy, University of Defense, Belgrade, Serbia, .,Medical Faculty Foča, University of East Sarajevo, Foča, Bosnia and Herzegovina
| | - Dragana Vučević
- Institute for Medical Research, Medical Faculty of the Military Medical Academy, University of Defense, Belgrade, Serbia,
| |
Collapse
|
20
|
Sculpting neurotransmission during synaptic development by 2D nanostructured interfaces. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:2521-2532. [DOI: 10.1016/j.nano.2017.01.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 12/08/2016] [Accepted: 01/04/2017] [Indexed: 11/23/2022]
|
21
|
Yang L, Chueng STD, Li Y, Patel M, Rathnam C, Dey G, Wang L, Cai L, Lee KB. A biodegradable hybrid inorganic nanoscaffold for advanced stem cell therapy. Nat Commun 2018; 9:3147. [PMID: 30089775 PMCID: PMC6082841 DOI: 10.1038/s41467-018-05599-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 07/17/2018] [Indexed: 01/06/2023] Open
Abstract
Stem cell transplantation, as a promising treatment for central nervous system (CNS) diseases, has been hampered by crucial issues such as a low cell survival rate, incomplete differentiation, and limited neurite outgrowth in vivo. Addressing these hurdles, scientists have designed bioscaffolds that mimic the natural tissue microenvironment to deliver physical and soluble cues. However, several significant obstacles including burst release of drugs, insufficient cellular adhesion support, and slow scaffold degradation rate remain to be overcome before the full potential of bioscaffold-based stem-cell therapies can be realized. To this end, we developed a biodegradable nanoscaffold-based method for enhanced stem cell transplantation, differentiation, and drug delivery. These findings collectively support the therapeutic potential of our biodegradable hybrid inorganic (BHI) nanoscaffolds for advanced stem cell transplantation and neural tissue engineering.
Collapse
Affiliation(s)
- Letao Yang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Sy-Tsong Dean Chueng
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Ying Li
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ, 08854, USA
| | - Misaal Patel
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ, 08854, USA
| | - Christopher Rathnam
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Gangotri Dey
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Lu Wang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Li Cai
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ, 08854, USA
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA.
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ, 08854, USA.
- College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea.
| |
Collapse
|
22
|
Dong J, Ma Q. Type 2 Immune Mechanisms in Carbon Nanotube-Induced Lung Fibrosis. Front Immunol 2018; 9:1120. [PMID: 29872441 PMCID: PMC5972321 DOI: 10.3389/fimmu.2018.01120] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 05/03/2018] [Indexed: 01/29/2023] Open
Abstract
T helper (Th) 2-dependent type 2 immune pathways have been recognized as an important driver for the development of fibrosis. Upon stimulation, activated Th2 immune cells and type 2 cytokines interact with inflammatory and tissue repair functions to stimulate an overzealous reparative response to tissue damage, leading to organ fibrosis and destruction. In this connection, type 2 pathways are activated by a variety of insults and pathological conditions to modulate the response. Carbon nanotubes (CNTs) are nanomaterials with a wide range of applications. However, pulmonary exposure to CNTs causes a number of pathologic outcomes in animal lungs, dominated by inflammation and fibrosis. These findings, alongside the rapidly expanding production and commercialization of CNTs and CNT-containing materials in recent years, have raised concerns on the health risk of CNT exposure in humans. The CNT-induced pulmonary fibrotic lesions resemble those of human fibrotic lung diseases, such as idiopathic pulmonary fibrosis and pneumoconiosis, to a certain extent with regard to disease development and pathological features. In fibrotic scenarios, immune cells are activated including varying immune pathways, ranging from innate immune cell activation to autoimmune disease. These events often precede and/or accompany the occurrence of fibrosis. Upon CNT exposure, significant induction and activation of Th2 cells and type 2 cytokines in the lungs are observed. Moreover, type 2 pathways are shown to play important roles in promoting CNT-induced lung fibrosis by producing type 2 pro-fibrotic factors and inducing the reparative phenotypes of macrophages in response to CNTs. In light of the vastly increased demand for nanosafety and the apparent induction and multiple roles of type 2 immune pathways in lung fibrosis, we review the current literature on CNT-induced lung fibrosis, with a focus on the induction and activation of type 2 responses by CNTs and the stimulating function of type 2 signaling on pulmonary fibrosis development. These analyses provide new insights into the mechanistic understanding of CNT-induced lung fibrosis, as well as the potential of using type 2 responses as a monitoring target and therapeutic strategy for human fibrotic lung disease.
Collapse
Affiliation(s)
| | - Qiang Ma
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, United States
| |
Collapse
|
23
|
Ray A, Macwan I, Singh S, Silwal S, Patra P. A Computational Approach for Understanding the Interactions between Graphene Oxide and Nucleoside Diphosphate Kinase with Implications for Heart Failure. NANOMATERIALS 2018; 8:nano8020057. [PMID: 29360759 PMCID: PMC5853690 DOI: 10.3390/nano8020057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/13/2018] [Accepted: 01/20/2018] [Indexed: 01/05/2023]
Abstract
During a heart failure, an increased content and activity of nucleoside diphosphate kinase (NDPK) in the sarcolemmal membrane is responsible for suppressing the formation of the second messenger cyclic adenosine monophosphate (cAMP)-a key component required for calcium ion homeostasis for the proper systolic and diastolic functions. Typically, this increased NDPK content lets the surplus NDPK react with a mutated G protein in the beta-adrenergic signal transduction pathway, thereby inhibiting cAMP synthesis. Thus, it is thus that inhibition of NDPK may cause a substantial increase in adenylate cyclase activity, which in turn may be a potential therapy for end-stage heart failure patients. However, there is little information available about the molecular events at the interface of NDPK and any prospective molecule that may potentially influence its reactive site (His118). Here we report a novel computational approach for understanding the interactions between graphene oxide (GO) and NDPK. Using molecular dynamics, it is found that GO interacts favorably with the His118 residue of NDPK to potentially prevent its binding with adenosine triphosphate (ATP), which otherwise would trigger the phosphorylation of the mutated G protein. Therefore, this will result in an increase in cAMP levels during heart failure.
Collapse
Affiliation(s)
- Anushka Ray
- Nashua High School South, Nashua, NH 03062, USA.
| | - Isaac Macwan
- Department of Biomedical Engineering, University of Bridgeport, Bridgeport, CT 06604, USA.
| | - Shrishti Singh
- Department of Biomedical Engineering, University of Bridgeport, Bridgeport, CT 06604, USA.
| | - Sushila Silwal
- Department of Biomedical Engineering, University of Bridgeport, Bridgeport, CT 06604, USA.
| | - Prabir Patra
- Department of Biomedical Engineering, University of Bridgeport, Bridgeport, CT 06604, USA.
| |
Collapse
|
24
|
D'Elios MM, Aldinucci A, Amoriello R, Benagiano M, Bonechi E, Maggi P, Flori A, Ravagli C, Saer D, Cappiello L, Conti L, Valtancoli B, Bencini A, Menichetti L, Baldi G, Ballerini C. Myelin-specific T cells carry and release magnetite PGLA–PEG COOH nanoparticles in the mouse central nervous system. RSC Adv 2018; 8:904-913. [PMID: 35538965 PMCID: PMC9076978 DOI: 10.1039/c7ra11290d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 12/13/2017] [Indexed: 11/21/2022] Open
Abstract
Progress in nanotechnology has determined new strategies concerning drug delivery into the central nervous system for the treatment of degenerative and inflammatory diseases. To date, brain targeting through systemic drug administration, even in a nano-composition, is often unsuccessful. Therefore, we investigated the possibility of loading T lymphocytes with PGLA–PEG COOH magnetite nanoparticles (30 nm), which can be built up to easily bind drugs and monoclonal antibodies, and to exploit the ability of activated T cells to cross the blood–brain barrier and infiltrate the brain parenchyma. Iron oxide nanoparticles have been widely used in biomedical applications due to their theranostic properties and are therefore a well-established nanomaterial. The magnetite core is easily hybridized with polymeric compounds that may enhance the possibility of the nanoparticles entering cells with low phagocytic properties. Taking advantage of these material characteristics, after in vitro assessment of the viability and functionality of nano-loaded MOG35–55 specific T cells, we transferred cells containing the nano-cargo into naïve mice affected by experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis. By means of histological and immunohistological methods, we were able to identify the nano-loaded T cells in the central nervous system. Our data demonstrated that T cells containing nanomaterials hold the possibility of carrying and releasing nanoparticles in the brain. Magnetite nanoparticles enter non-phagocytic myelin-specific T cells and reach the central nervous system after in vivo transfer.![]()
Collapse
Affiliation(s)
- M. M. D'Elios
- Department of Clinical and Experimental Medicine
- University of Florence
- Italy
| | - A. Aldinucci
- Department of Neuroscience, Psychology, Drug and Child Health
- University of Florence
- Italy
| | - R. Amoriello
- Department of Neuroscience, Psychology, Drug and Child Health
- University of Florence
- Italy
| | - M. Benagiano
- Department of Clinical and Experimental Medicine
- University of Florence
- Italy
| | - E. Bonechi
- Department of Neuroscience, Psychology, Drug and Child Health
- University of Florence
- Italy
| | - P. Maggi
- Department of Neurology
- Hôpital CHU Brugmann-Université libre de Bruxelles-Bruxelles-Be
- Belgium
| | - A. Flori
- Fondazione CNR Regione Toscana G. Monasterio
- Pisa
- Italy
| | - C. Ravagli
- Research Center Colorobbia
- Cericol, Colorobbia Consulting
- Florence
- Italy
| | - D. Saer
- Research Center Colorobbia
- Cericol, Colorobbia Consulting
- Florence
- Italy
| | - L. Cappiello
- Research Center Colorobbia
- Cericol, Colorobbia Consulting
- Florence
- Italy
| | - L. Conti
- Department of Chemistry Ugo Schiff
- University of Florence
- Italy
| | - B. Valtancoli
- Department of Chemistry Ugo Schiff
- University of Florence
- Italy
| | - A. Bencini
- Department of Chemistry Ugo Schiff
- University of Florence
- Italy
| | | | - G. Baldi
- Research Center Colorobbia
- Cericol, Colorobbia Consulting
- Florence
- Italy
| | - C. Ballerini
- Department of Neuroscience, Psychology, Drug and Child Health
- University of Florence
- Italy
| |
Collapse
|
25
|
Abstract
In 2015, cancer was the cause of almost 22% of deaths worldwide. The high frequency of relapsing diseases and metastasis requires the development of new diagnostic and therapeutic approaches, and the use of nanomaterials is a promising tool for fighting cancer. Among the more extensively studied nanomaterials are carbon nanotubes (CNTs), synthesized as graphene sheets, whose spiral shape is varied in length and thickness. Their physicochemical features, such as the resistance to tension, and thermal and electrical conductivity, allow their application in several fields. In this review, we show evidence supporting the applicability of CNTs in biomedical practice as nanocarriers for drugs and immunomodulatory material, emphasizing their potential for use in cancer treatment.
Collapse
|
26
|
Ong LC, Tan YF, Tan BS, Chung FFL, Cheong SK, Leong CO. Single-walled carbon nanotubes (SWCNTs) inhibit heat shock protein 90 (HSP90) signaling in human lung fibroblasts and keratinocytes. Toxicol Appl Pharmacol 2017; 329:347-357. [DOI: 10.1016/j.taap.2017.06.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 06/17/2017] [Accepted: 06/30/2017] [Indexed: 12/15/2022]
|
27
|
Zhu W, Ye T, Lee SJ, Cui H, Miao S, Zhou X, Shuai D, Zhang LG. Enhanced neural stem cell functions in conductive annealed carbon nanofibrous scaffolds with electrical stimulation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 14:2485-2494. [PMID: 28552650 DOI: 10.1016/j.nano.2017.03.018] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 01/19/2017] [Accepted: 03/06/2017] [Indexed: 01/19/2023]
Abstract
Carbon-based nanomaterials have shown great promise in regenerative medicine because of their unique electrical, mechanical, and biological properties; however, it is still difficult to engineer 2D pure carbon nanomaterials into a 3D scaffold while maintaining its structural integrity. In the present study, we developed novel carbon nanofibrous scaffolds by annealing electrospun mats at elevated temperature. The resultant scaffold showed a cohesive structure and excellent mechanical flexibility. The graphitic structure generated by annealing renders superior electrical conductivity to the carbon nanofibrous scaffold. By integrating the conductive scaffold with biphasic electrical stimulation, neural stem cell proliferation was promoted associating with upregulated neuronal gene expression level and increased microtubule-associated protein 2 immunofluorescence, demonstrating an improved neuronal differentiation and maturation. The findings suggest that the integration of the conducting carbon nanofibrous scaffold and electrical stimulation may pave a new avenue for neural tissue regeneration.
Collapse
Affiliation(s)
- Wei Zhu
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, USA
| | - Tao Ye
- Department of Civil and Environmental Engineering, The George Washington University, Washington, DC, United States
| | - Se-Jun Lee
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, USA
| | - Haitao Cui
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, USA
| | - Shida Miao
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, USA
| | - Xuan Zhou
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, USA
| | - Danmeng Shuai
- Department of Civil and Environmental Engineering, The George Washington University, Washington, DC, United States
| | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, USA; Department of Biomedical Engineering, The George Washington University, Washington, DC, USA; Department of Medicine, The George Washington University, Washington, DC, USA.
| |
Collapse
|
28
|
Yang Y, Hu Y, Wang H. Targeting Antitumor Immune Response for Enhancing the Efficacy of Photodynamic Therapy of Cancer: Recent Advances and Future Perspectives. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:5274084. [PMID: 27672421 PMCID: PMC5031843 DOI: 10.1155/2016/5274084] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/04/2016] [Indexed: 02/06/2023]
Abstract
Photodynamic therapy (PDT) is a minimally invasive therapeutic strategy for cancer treatment, which can destroy local tumor cells and induce systemic antitumor immune response, whereas, focusing on improving direct cytotoxicity to tumor cells treated by PDT, there is growing interest in developing approaches to further explore the immune stimulatory properties of PDT. In this review we summarize the current knowledge of the innate and adaptive immune responses induced by PDT against tumors, providing evidence showing PDT facilitated-antitumor immunity. Various immunotherapeutic approaches on different cells are reviewed for their effectiveness in improving the treatment efficiency in concert with PDT. Future perspectives are discussed for further enhancing PDT efficiency via intracellular targetable drug delivery as well as optimized experimental model development associated with the study of antitumor immune response.
Collapse
Affiliation(s)
- Yamin Yang
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, 169 Sheng Tai West Road, Nanjing, Jiangsu 211106, China
| | - Yue Hu
- Department of Biological and Environmental Engineering, Cornell University, 120 Riley Robb, Ithaca, NY 14853, USA
| | - Hongjun Wang
- Department of Chemistry, Chemical Biology and Biomedical Engineering, Stevens Institute of Technology, Castle Point on Hudson, Hoboken, NJ 07030, USA
| |
Collapse
|
29
|
Tomić S, Kokol V, Mihajlović D, Mirčić A, Čolić M. Native cellulose nanofibrills induce immune tolerance in vitro by acting on dendritic cells. Sci Rep 2016; 6:31618. [PMID: 27558765 PMCID: PMC4997350 DOI: 10.1038/srep31618] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 07/20/2016] [Indexed: 12/22/2022] Open
Abstract
Cellulose nanofibrills (CNFs) are attractive biocompatible, natural nanomaterials for wide biomedical applications. However, the immunological mechanisms of CNFs have been poorly investigated. Considering that dendritic cells (DCs) are the key immune regulatory cells in response to nanomaterials, our aim was to investigate the immunological mechanisms of CNFs in a model of DC-mediated immune response. We found that non-toxic concentrations of CNFs impaired the differentiation, and subsequent maturation of human monocyte-derived (mo)-DCs. In a co-culture with CD4+T cells, CNF-treated mo-DCs possessed a weaker allostimulatory and T helper (Th)1 and Th17 polarizing capacity, but a stronger capacity to induce Th2 cells and CD4+CD25hiFoxP3hi regulatory T cells. This correlated with an increased immunoglobulin-like transcript-4 and indolamine dioxygenase-1 expression by CNF-treated mo-DCs, following the partial internalization of CNFs and the accumulation of CD209 and actin bundles at the place of contacts with CNFs. Cumulatively, we showed that CNFs are able to induce an active immune tolerance by inducing tolerogenic DCs, which could be beneficial for the application of CNFs in wound healing and chronic inflammation therapies.
Collapse
Affiliation(s)
- Sergej Tomić
- University of Defense, Medical Faculty of the Military Medical Academy, Institute for Medical Research, Belgrade, Serbia
| | - Vanja Kokol
- University of Maribor, Faculty of Mechanical Engineering, Institute for Engineering Materials and Design, Maribor, Slovenia
| | - Dušan Mihajlović
- University of Defense, Medical Faculty of the Military Medical Academy, Institute for Medical Research, Belgrade, Serbia
| | - Aleksandar Mirčić
- University of Belgrade, Institute of Histology and Embryology, School of Medicine, Belgrade, Serbia
| | - Miodrag Čolić
- University of Defense, Medical Faculty of the Military Medical Academy, Institute for Medical Research, Belgrade, Serbia.,University of Belgrade, Institute for Application of Nuclear Energy, Belgrade, Serbia
| |
Collapse
|
30
|
Aldinucci A, Bonechi E, Manuelli C, Nosi D, Masini E, Passani MB, Ballerini C. Histamine Regulates Actin Cytoskeleton in Human Toll-like Receptor 4-activated Monocyte-derived Dendritic Cells Tuning CD4+ T Lymphocyte Response. J Biol Chem 2016; 291:14803-14. [PMID: 27226579 DOI: 10.1074/jbc.m116.720680] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Indexed: 01/06/2023] Open
Abstract
Histamine, a major mediator in allergic diseases, differentially regulates the polarizing ability of dendritic cells after Toll-like receptor (TLR) stimulation, by not completely explained mechanisms. In this study we investigated the effects of histamine on innate immune reaction during the response of human monocyte-derived DCs (mDCs) to different TLR stimuli: LPS, specific for TLR4, and Pam3Cys, specific for heterodimer molecule TLR1/TLR2. We investigated actin remodeling induced by histamine together with mDCs phenotype, cytokine production, and the stimulatory and polarizing ability of Th0. By confocal microscopy and RT-PCR expression of Rac1/CdC42 Rho GTPases, responsible for actin remodeling, we show that histamine selectively modifies actin cytoskeleton organization induced by TLR4, but not TLR2 and this correlates with increased IL4 production and decreased IFNγ by primed T cells. We also demonstrate that histamine-induced cytoskeleton organization is at least in part mediated by down-regulation of small Rho GTPase CdC42 and the protein target PAK1, but not by down-regulation of Rac1. The presence and relative expression of histamine receptors HR1-4 and TLRs were determined as well. Independently of actin remodeling, histamine down-regulates IL12p70 and CXCL10 production in mDCs after TLR2 and TLR4 stimulation. We also observed a trend of IL10 up-regulation that, despite previous reports, did not reach statistical significance.
Collapse
Affiliation(s)
| | - Elena Bonechi
- From the Departments of Neuroscience, Psychology, Drugs and Child Health
| | | | | | - Emanuela Masini
- From the Departments of Neuroscience, Psychology, Drugs and Child Health
| | | | - Clara Ballerini
- From the Departments of Neuroscience, Psychology, Drugs and Child Health,
| |
Collapse
|
31
|
Kim TE, Kim CG, Kim JS, Jin S, Yoon S, Bae HR, Kim JH, Jeong YH, Kwak JY. Three-dimensional culture and interaction of cancer cells and dendritic cells in an electrospun nano-submicron hybrid fibrous scaffold. Int J Nanomedicine 2016; 11:823-35. [PMID: 27042051 PMCID: PMC4780731 DOI: 10.2147/ijn.s101846] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
An artificial three-dimensional (3D) culture system that mimics the tumor microenvironment in vitro is an essential tool for investigating the cross-talk between immune and cancer cells in tumors. In this study, we developed a 3D culture system using an electrospun poly(ε-caprolactone) (PCL) nanofibrous scaffold (NFS). A hybrid NFS containing an uninterrupted network of nano- and submicron-scale fibers (400 nm to 2 µm) was generated by deposition onto a stainless steel mesh instead of an aluminum plate. The hybrid NFS contained multiplanar pores in a 3D structure. Surface-seeded mouse CT26 colon cancer cells and bone marrow-derived dendritic cells (BM-DCs) were able to infiltrate the hybrid NFS within several hours. BM-DCs cultured on PCL nanofibers showed a baseline inactive form, and lipopolysaccharide (LPS)-activated BM-DCs showed increased expression of CD86 and major histocompatibility complex Class II. Actin and phosphorylated FAK were enriched where unstimulated and LPS-stimulated BM-DCs contacted the fibers in the 3D hybrid NFS. When BM-DCs were cocultured with mitoxantrone-treated CT26 cells in a 3D hybrid NFS, BM-DCs sprouted cytoplasm to, migrated to, synapsed with, and engulfed mitoxantrone-treated CT26 cancer cells, which were similar to the naturally occurring cross-talk between these two types of cells. The 3D hybrid NFS developed here provides a 3D structure for coculture of cancer and immune cells.
Collapse
Affiliation(s)
- Tae-Eon Kim
- Department of Pharmacology, School of Medicine, Ajou University, Suwon, South Korea; Department of Biomedical Sciences, The Graduate School, Ajou University, Suwon, South Korea; Immune Network Pioneer Research Center, Ajou University Medical Center, Suwon, South Korea
| | - Chang Gun Kim
- Department of Pharmacology, School of Medicine, Ajou University, Suwon, South Korea; Department of Biomedical Sciences, The Graduate School, Ajou University, Suwon, South Korea; Immune Network Pioneer Research Center, Ajou University Medical Center, Suwon, South Korea
| | - Jin Soo Kim
- Department of Mechanical Engineering, Korea Polytechnic University, Gyeonggi, South Korea
| | - Songwan Jin
- Department of Mechanical Engineering, Korea Polytechnic University, Gyeonggi, South Korea
| | - Sik Yoon
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, South Korea
| | - Hae-Rahn Bae
- Department of Physiology, College of Medicine, Dong-A University, Busan, South Korea
| | - Jeong-Hwa Kim
- School of Mechanical Engineering, Kyungpook National University, Daegu, South Korea; Department of Mechanical Engineering, Graduate School, Kyungpook National University, Daegu, South Korea
| | - Young Hun Jeong
- School of Mechanical Engineering, Kyungpook National University, Daegu, South Korea; Department of Mechanical Engineering, Graduate School, Kyungpook National University, Daegu, South Korea
| | - Jong-Young Kwak
- Department of Pharmacology, School of Medicine, Ajou University, Suwon, South Korea; Department of Biomedical Sciences, The Graduate School, Ajou University, Suwon, South Korea; Immune Network Pioneer Research Center, Ajou University Medical Center, Suwon, South Korea
| |
Collapse
|
32
|
Marchesan S, Melchionna M, Prato M. Wire Up on Carbon Nanostructures! How To Play a Winning Game. ACS NANO 2015; 9:9441-50. [PMID: 26390071 DOI: 10.1021/acsnano.5b04956] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Carbon nanotubes and graphene possess a unique extended π-system that makes them stand out among carbon nanostructures. The resulting electronic properties enable electron or charge flow along one or two directions, respectively, thus offering the opportunity to connect electronically different entities that come into contact, be they living cells or catalytic systems. Using these carbon nanostructures thus holds great promise in providing innovative solutions to address key challenges in the fields of medicine and energy. Here, we discuss how chemical functionalization of these carbon nanostructures is a crucial tool to master their properties and deliver innovation.
Collapse
Affiliation(s)
- Silvia Marchesan
- Center of Excellence for Nanostructured Materials, INSTM, Unit of Trieste, Dipartimento di Scienze Chimiche e Farmaceutiche, University of Trieste , Piazzale Europa 1, 34127 Trieste, Italy
| | - Michele Melchionna
- Center of Excellence for Nanostructured Materials, INSTM, Unit of Trieste, Dipartimento di Scienze Chimiche e Farmaceutiche, University of Trieste , Piazzale Europa 1, 34127 Trieste, Italy
| | - Maurizio Prato
- Center of Excellence for Nanostructured Materials, INSTM, Unit of Trieste, Dipartimento di Scienze Chimiche e Farmaceutiche, University of Trieste , Piazzale Europa 1, 34127 Trieste, Italy
| |
Collapse
|
33
|
Abstract
Strategies to enhance, suppress, or qualitatively shape the immune response are of importance for diverse biomedical applications, such as the development of new vaccines, treatments for autoimmune diseases and allergies, strategies for regenerative medicine, and immunotherapies for cancer. However, the intricate cellular and molecular signals regulating the immune system are major hurdles to predictably manipulating the immune response and developing safe and effective therapies. To meet this challenge, biomaterials are being developed that control how, where, and when immune cells are stimulated in vivo, and that can finely control their differentiation in vitro. We review recent advances in the field of biomaterials for immunomodulation, focusing particularly on designing biomaterials to provide controlled immunostimulation, targeting drugs and vaccines to lymphoid organs, and serving as scaffolds to organize immune cells and emulate lymphoid tissues. These ongoing efforts highlight the many ways in which biomaterials can be brought to bear to engineer the immune system.
Collapse
Affiliation(s)
- Nathan A Hotaling
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine
- Parker H. Petit Institute for Bioengineering and Biosciences, and
| | - Li Tang
- Department of Materials Science and Engineering
- Department of Biological Engineering, and
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139;
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139
| | - Darrell J Irvine
- Department of Materials Science and Engineering
- Department of Biological Engineering, and
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139;
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139
- Howard Hughes Medical Institute, Chevy Chase, Maryland 20815
| | - Julia E Babensee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine
- Parker H. Petit Institute for Bioengineering and Biosciences, and
- Center for Immunoengineering, Georgia Institute of Technology, Atlanta, Georgia 30332;
| |
Collapse
|
34
|
Amoozgar Z, Goldberg MS. Targeting myeloid cells using nanoparticles to improve cancer immunotherapy. Adv Drug Deliv Rev 2015; 91:38-51. [PMID: 25280471 DOI: 10.1016/j.addr.2014.09.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 09/15/2014] [Accepted: 09/16/2014] [Indexed: 12/23/2022]
Abstract
While nanoparticles have traditionally been used to deliver cytotoxic drugs directly to tumors to induce cancer cell death, emerging data suggest that nanoparticles are likely to generate a larger impact on oncology through the delivery of agents that can stimulate antitumor immunity. Tumor-targeted nanocarriers have generally been used to localize chemotherapeutics to tumors and thus decrease off-target toxicity while enhancing efficacy. Challengingly, tumor heterogeneity and evolution render tumor-intrinsic approaches likely to succumb to relapse. The immune system offers exquisite specificity, cytocidal potency, and long-term activity that leverage an adaptive memory response. For this reason, the ability to manipulate immune cell specificity and function would be desirable, and nanoparticles represent an exciting means by which to perform such manipulation. Dendritic cells and tumor-associated macrophages are cells of the myeloid lineage that function as natural phagocytes, so they naturally take up nanoparticles. Dendritic cells direct the specificity and potency of cellular immune responses that can be targeted for cancer vaccines. Herein, we discuss the specific criteria needed for efficient vaccine design, including but not limited to the route of administration, size, morphology, surface charge, targeting ligands, and nanoparticle composition. In contrast, tumor-associated macrophages are critical mediators of immunosuppression whose trans-migratory abilities can be exploited to localize therapeutics to the tumor core and which can be directly targeted for elimination or for repolarization to a tumor suppressive phenotype. It is likely that a combination of targeting dendritic cells to stimulate antitumor immunity and tumor-associated macrophages to reduce immune suppression will impart significant benefits and result in durable antitumor responses.
Collapse
|
35
|
Meng J, Li X, Wang C, Guo H, Liu J, Xu H. Carbon nanotubes activate macrophages into a M1/M2 mixed status: recruiting naïve macrophages and supporting angiogenesis. ACS APPLIED MATERIALS & INTERFACES 2015; 7:3180-3188. [PMID: 25591447 DOI: 10.1021/am507649n] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The potential of carbon nanotubes (CNTs) in medical applications has been attracting constant research interest as well as raising concerns related to toxicity. The immune system serves as the first line of defense against invasion. In this work, interactions of oxidized multiwalled carbon nanotubes (MWCNT) with macrophages were investigated to unravel the activation profile of macrophages, using cytokine array, ELISA assay, transwell assay, confocal microscopy, and reactive oxygen species examination. Results show that MWCNT initiate phagocytosis of macrophages and upregulate CD14, CD11b, TLR-4/MD2, and CD206, which does not alter the MHCII expression of the macrophages. The macrophages engulfing MWCNT (MWCNT-RAW) secrete a large amount of MIP-1α and MIP-2 to recruit naïve macrophages and produce angiogenesis-related cytokines MMP-9 and VEGF, while inducing much lower levels of proinflammatory cytokines than those activated by LPS. In conclusion, MWCNT activate macrophages into a M1/M2 mixed status, which allows the cells to recruit naïve macrophages and support angiogenesis.
Collapse
Affiliation(s)
- Jie Meng
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, P. R. China
| | | | | | | | | | | |
Collapse
|
36
|
Ballerini C, Baldi G, Aldinucci A, Maggi P. Nanomaterial applications in multiple sclerosis inflamed brain. J Neuroimmune Pharmacol 2015; 10:1-13. [PMID: 25616566 DOI: 10.1007/s11481-015-9588-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 01/14/2015] [Indexed: 12/22/2022]
Abstract
In the last years scientific progress in nanomaterials, where size and shape make the difference, has increased their utilization in medicine with the development of a promising new translational science: nanomedicine. Due to their surface and core biophysical properties, nanomaterials hold the promise for medical applications in central nervous system (CNS) diseases: inflammatory, degenerative and tumors. The present review is focused on nanomaterials at the neuro-immune interface, evaluating two aspects: the possible CNS inflammatory response induced by nanomaterials and the developments of nanomaterials to improve treatment and diagnosis of neuroinflammatory diseases, with a focus on multiple sclerosis (MS). Indeed, nanomedicine allows projecting new ways of drug delivery and novel techniques for CNS imaging. Despite the wide field of application in neurological diseases of nanomaterials, our topic here is to review the more recent development of nanomaterials that cross blood brain barrier (BBB) and reach specific target during CNS inflammatory diseases, a crucial strategy for CNS early diagnosis and drug delivery, indeed the main challenges of nanomedicine.
Collapse
Affiliation(s)
- Clara Ballerini
- Department of Neurofarba, University of Florence, Viale Pieraccini, 6, 50137, Florence, Italy,
| | | | | | | |
Collapse
|
37
|
Kyle S, Saha S. Nanotechnology for the detection and therapy of stroke. Adv Healthc Mater 2014; 3:1703-20. [PMID: 24692428 DOI: 10.1002/adhm.201400009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Indexed: 01/06/2023]
Abstract
Over the years, nanotechnology has greatly developed, moving from careful design strategies and synthesis of novel nanostructures to producing them for specific medical and biological applications. The use of nanotechnology in diagnostics, drug delivery, and tissue engineering holds great promise for the treatment of stroke in the future. Nanoparticles are employed to monitor grafted cells upon implantation, or to enhance the imagery of the tissue, which is coupled with a noninvasive imaging modality such as magnetic resonance imaging, computed axial tomography or positron emission tomography scan. Contrast imaging agents used can range from iron oxide, perfluorocarbon, cerium oxide or platinum nanoparticles to quantum dots. The use of nanomaterial scaffolds for neuroregeneration is another area of nanomedicine, which involves the creation of an extracellular matrix mimic that not only serves as a structural support but promotes neuronal growth, inhibits glial differentiation, and controls hemostasis. Promisingly, carbon nanotubes can act as scaffolds for stem cell therapy and functionalizing these scaffolds may enhance their therapeutic potential for treatment of stroke. This Progress Report highlights the recent developments in nanotechnology for the detection and therapy of stroke. Recent advances in the use of nanomaterials as tissue engineering scaffolds for neuroregeneration will also be discussed.
Collapse
Affiliation(s)
- Stuart Kyle
- School of Medicine; University of Leeds; Leeds LS2 9JT UK
| | - Sikha Saha
- Division of Cardiovascular and Diabetes Research; Leeds Institute of Genetics; Health and Therapeutics; University of Leeds; Leeds LS2 9JT UK
| |
Collapse
|
38
|
Bonechi E, Aldinucci A, Mazzanti B, di Gioia M, Repice AM, Manuelli C, Saccardi R, Massacesi L, Ballerini C. Increased CXCL10 expression in MS MSCs and monocytes is unaffected by AHSCT. Ann Clin Transl Neurol 2014; 1:650-8. [PMID: 25493279 PMCID: PMC4241792 DOI: 10.1002/acn3.92] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 06/25/2014] [Accepted: 07/15/2014] [Indexed: 01/04/2023] Open
Abstract
Objective To confirm CXCL10 over production in bone marrow mesenchymal stem cells (MSCs) and circulating monocytes isolated from multiple sclerosis patients (MS) and identify predate cell molecular signature; to extend this analysis after autologous hematopoietic stem cell transplantation (AHSCT) to test if therapy has modifying effects on MSCs and circulating monocytes. Methods MSCs and monocytes were isolated from 19 MS patients who undergone AHSCT before and seven of them at least 3 years after transplant. CXCL10 production was detected after LPS/IFN-γ stimulation. TLR4 signaling pathways were investigated by means of transcription factors phosphorylation/activation level. RT-PCR of activated transcription factors was performed to quantify their expression. All experiments were conducted in parallel with 24 matched healthy donors (HD). Results CXCL10 expression was significantly increased in both peripheral circulating monocytes and BM MSCs compared to HD. We showed that CXCL10 production is determined by an altered signaling pathway downstream TLR4, with the involvement of STAT-1, NF-κB, p38, JNK, and CREB. All upregulated transcription factors are more phosphorylated in MS patient sample. These features are not modified after AHSCT. Interpretation We demonstrated that in MS two different cell lineages are characterized by significantly increased production of CXCL10, due to altered signaling pathways of innate immune reaction mediated by TLR4, probably associated with disease phenotype. This characteristic is not modified by AHSCT, suggesting that when T and B lymphocytes are reset, other possible components of MS pathology, such as CXCL10 over production, do not determine therapy outcome.
Collapse
Affiliation(s)
- Elena Bonechi
- Dept. NEUROFARBA, University of Florence Florence, Italy
| | | | | | | | | | | | | | - Luca Massacesi
- Dept. NEUROFARBA, University of Florence Florence, Italy
| | | |
Collapse
|
39
|
Yang Y, Shi S, Ding Q, Chen J, Peng J, Xu Y. Multiwalled carbon nanotube-modified poly(d,l-lactide-co-glycolide) scaffolds for dendritic cell load. J Biomed Mater Res A 2014; 103:1045-52. [DOI: 10.1002/jbm.a.35255] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 05/15/2014] [Accepted: 06/04/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Yanzhu Yang
- School of Pharmacy, Shanghai Jiao Tong University; Shanghai 200240 People's Republic of China
| | - Sanyuan Shi
- School of Biomedical Engineering/Med-X Research Institute, Shanghai Jiao Tong University; Shanghai 200030 People's Republic of China
| | - Qian Ding
- School of Pharmacy, Shanghai Jiao Tong University; Shanghai 200240 People's Republic of China
| | - Jian Chen
- School of Pharmacy, Shanghai Jiao Tong University; Shanghai 200240 People's Republic of China
| | - Jinliang Peng
- School of Pharmacy, Shanghai Jiao Tong University; Shanghai 200240 People's Republic of China
- School of Biomedical Engineering/Med-X Research Institute, Shanghai Jiao Tong University; Shanghai 200030 People's Republic of China
| | - Yuhong Xu
- School of Pharmacy, Shanghai Jiao Tong University; Shanghai 200240 People's Republic of China
- School of Biomedical Engineering/Med-X Research Institute, Shanghai Jiao Tong University; Shanghai 200030 People's Republic of China
| |
Collapse
|
40
|
Immunotherapy applications of carbon nanotubes: from design to safe applications. Trends Biotechnol 2014; 32:198-209. [DOI: 10.1016/j.tibtech.2014.02.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 01/20/2014] [Accepted: 02/07/2014] [Indexed: 02/05/2023]
|
41
|
Tian Y, Wang H, Liu Y, Mao L, Chen W, Zhu Z, Liu W, Zheng W, Zhao Y, Kong D, Yang Z, Zhang W, Shao Y, Jiang X. A peptide-based nanofibrous hydrogel as a promising DNA nanovector for optimizing the efficacy of HIV vaccine. NANO LETTERS 2014; 14:1439-45. [PMID: 24564254 DOI: 10.1021/nl404560v] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
This report shows that a nanovector composed of peptide-based nanofibrous hydrogel can condense DNA to result in strong immune responses against HIV. This nanovector can strongly activate both humoral and cellular immune responses to a balanced level rarely reported in previous studies, which is crucial for HIV prevention and therapy. In addition, this nanovector shows good biosafety in vitro and in vivo. Detailed characterizations show that the nanofibrous structure of the hydrogel is critical for the dramatically improved immune responses compared to existing materials. This peptide-based nanofibrous hydrogel shows great potential for efficacious HIV DNA vaccines and can be potentially used for delivering other vaccines and drugs.
Collapse
Affiliation(s)
- Yue Tian
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology , No., 11 Zhongguancun Beiyitiao, Beijing 100190, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
McCarthy DP, Hunter ZN, Chackerian B, Shea LD, Miller SD. Targeted immunomodulation using antigen-conjugated nanoparticles. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2014; 6:298-315. [PMID: 24616452 DOI: 10.1002/wnan.1263] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 01/23/2014] [Accepted: 02/01/2014] [Indexed: 12/20/2022]
Abstract
The growing prevalence of nanotechnology in the fields of biology, medicine, and the pharmaceutical industry is confounded by the relatively small amount of data on the impact of these materials on the immune system. In addition to concerns surrounding the potential toxicity of nanoparticle (NP)-based delivery systems, there is also a demand for a better understanding of the mechanisms governing interactions of NPs with the immune system. Nanoparticles can be tailored to suppress, enhance, or subvert recognition by the immune system. This 'targeted immunomodulation' can be achieved by delivery of unmodified particles, or by modifying particles to deliver drugs, proteins/peptides, or genes to a specific site. In order to elicit the desired, beneficial immune response, considerations should be made at every step of the design process: the NP platform itself, ligands, and other modifiers, the delivery route, and the immune cells that will encounter the conjugated NPs can all impact host immune responses.
Collapse
Affiliation(s)
- Derrick P McCarthy
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | | | | | | |
Collapse
|
43
|
The Significance and Insignificance of Carbon Nanotube-Induced Inflammation. FIBERS 2014. [DOI: 10.3390/fib2010045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
44
|
Fileti E, Colherinhas G, Malaspina T. Predicting the properties of a new class of host–guest complexes: C60 fullerene and CB[9] cucurbituril. Phys Chem Chem Phys 2014; 16:22823-9. [DOI: 10.1039/c4cp03299c] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
DFT, semi-empirical and classical molecular dynamics methods were used to describe the structure and stability of the inclusion complex formed by the fullerene C60 and the cucurbituril CB[9].
Collapse
Affiliation(s)
- Eudes Fileti
- Instituto de Ciência e Tecnologia
- Universidade Federal de São Paulo
- São José dos Campos, Brazil
| | | | - Thaciana Malaspina
- Instituto de Ciência e Tecnologia
- Universidade Federal de São Paulo
- São José dos Campos, Brazil
| |
Collapse
|
45
|
Battigelli A, Ménard-Moyon C, Bianco A. Carbon nanomaterials as new tools for immunotherapeutic applications. J Mater Chem B 2014; 2:6144-6156. [DOI: 10.1039/c4tb00563e] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The possibility to exploit carbon-based nanostructures such as carbon nanotubes and graphene as immunotherapeutic agents has interesting future prospects. In particular, their applications for anticancer treatment, imaging and vaccine development, together with their immunomodulator properties are highlighted.
Collapse
Affiliation(s)
- Alessia Battigelli
- CNRS
- Institut de Biologie Moléculaire et Cellulaire
- Laboratoire d'Immunopathologie et Chimie Thérapeutique
- 67000 Strasbourg, France
| | - Cécilia Ménard-Moyon
- CNRS
- Institut de Biologie Moléculaire et Cellulaire
- Laboratoire d'Immunopathologie et Chimie Thérapeutique
- 67000 Strasbourg, France
| | - Alberto Bianco
- CNRS
- Institut de Biologie Moléculaire et Cellulaire
- Laboratoire d'Immunopathologie et Chimie Thérapeutique
- 67000 Strasbourg, France
| |
Collapse
|