1
|
Soleimanifar F, Aghapur N, Rezaei-Kiasari Z, Mahboudi H, Kaabi M, Mansour RN, Kehtari M, Abazari M, Enderami SE, Hassannia H. The generation of islet-like insulin-producing cells from Wharton's jelly-derived mesenchymal stem cells on the PES/fish gelatin scaffold. Regen Ther 2024; 26:251-259. [PMID: 38974324 PMCID: PMC11225687 DOI: 10.1016/j.reth.2024.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/12/2024] [Accepted: 05/30/2024] [Indexed: 07/09/2024] Open
Abstract
Diabetes Mellitus (DM) disrupts the body's capability to control blood glucose statuses. Type 1 diabetes mellitus (T1DM) arises from inadequate insulin production and is treated with insulin replacement therapy. Stem cell therapy is a hopeful treatment for T1DM that involves using adult stem cells to generate insulin-producing cells (IPCs). Mesenchymal stem cells (MSCs) are particularly advantageous for generating IPCs. The islet cells require interactions with the extracellular matrix for survival, which is lacking in conventional 2D culture systems. Natural or synthetic polymers create a supportive 3D microenvironment in tissue engineering. We aim to construct superior differentiation conditions employing polyethersulfone (PES)/Fish gelatin scaffolds to differentiate Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) to IPCs. In this study, the PES/fish gelatin scaffold (3D) was manufactured by electrospinning, and then its biocompatibility and non-toxicity were investigated by MTT assay. After that, scaffold-supportive effects on WJ-MSCs differentiation to IPCs were studied at the gene and protein levels. After exposure to the differentiation media, 2D and 3D (PES/Fish gelatin) cultured cells were slowly aggregated and developed spherical-shaped clusters. The viability of cells was found to be comparable in both 2D and 3D cultures. The gene expression analysis showed that efficiency of differentiation was more elevated in 3D culture. Additionally, ELISA results indicated that C-peptide and insulin release were more significant in 3D than in 2D culture. In conclusion, the PES/fish gelatin scaffold is highly promising for pancreatic tissue engineering because it supports the viability, growth, and differentiation of WJ-MSCs into IPCs.
Collapse
Affiliation(s)
- Fatemeh Soleimanifar
- Department of Medical Biotechnology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Zeinab Rezaei-Kiasari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hosein Mahboudi
- Department of Medical Biotechnology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Reyhaneh Nassiri Mansour
- Department of Regenerative Medicine, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mousa Kehtari
- Department of Animal Biology, School of Biology, Faculty of Science, University of Tehran, Tehran, Iran
| | - Mohammadfoad Abazari
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hadi Hassannia
- Amol Faculty of Paramedical Sciences, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
2
|
Zhang S, Yang W, Gong W, Lu Y, Yu DG, Liu P. Recent progress of electrospun nanofibers as burning dressings. RSC Adv 2024; 14:14374-14391. [PMID: 38694552 PMCID: PMC11061782 DOI: 10.1039/d4ra01514b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/25/2024] [Indexed: 05/04/2024] Open
Abstract
Burns are a global public health problem, which brings great challenges to public health and the economy. Severe burns often lead to systemic infection, shock, multiple organ failure, and even death. With the increasing demand for the therapeutic effect of burn wounds, traditional dressings have been unable to meet people's needs due to their single function and many side effects. In this context, electrospinning shows a great prospect on the way to open up advanced wound dressings that promote wound repairing and prevent infection. With its large specific surface area, high porosity, and similar to natural extracellular matrix (ECM), electrospun nanofibers can load drugs and accelerate wound healing. It provides a promising solution for the treatment and management of burn wounds. This review article introduces the concept of burn and the types of electrospun nanofibers, then summarizes the polymers used in electrospun nanofiber dressings. Finally, the drugs (plant extracts, small molecule drugs and nanoparticles) loaded with electrospun burn dressings are summarized. Some promising aspects for developing commercial electrospun burn dressings are proposed.
Collapse
Affiliation(s)
- Shengwei Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology Shanghai 200093 China
| | - Wei Yang
- The Base of Achievement Transformation, Shidong Hospital Affiliated to University of Shanghai for Science and Technology Shanghai 200443 China
| | - Wenjian Gong
- School of Materials and Chemistry, University of Shanghai for Science and Technology Shanghai 200093 China
| | - Yuhang Lu
- School of Materials and Chemistry, University of Shanghai for Science and Technology Shanghai 200093 China
| | - Deng-Guang Yu
- School of Materials and Chemistry, University of Shanghai for Science and Technology Shanghai 200093 China
| | - Ping Liu
- The Base of Achievement Transformation, Shidong Hospital Affiliated to University of Shanghai for Science and Technology Shanghai 200443 China
| |
Collapse
|
3
|
Alhattab DM, Isaioglou I, Alshehri S, Khan ZN, Susapto HH, Li Y, Marghani Y, Alghuneim AA, Díaz-Rúa R, Abdelrahman S, Al-Bihani S, Ahmed F, Felimban RI, Alkhatabi H, Alserihi R, Abedalthagafi M, AlFadel A, Awidi A, Chaudhary AG, Merzaban J, Hauser CAE. Fabrication of a three-dimensional bone marrow niche-like acute myeloid Leukemia disease model by an automated and controlled process using a robotic multicellular bioprinting system. Biomater Res 2023; 27:111. [PMID: 37932837 PMCID: PMC10626721 DOI: 10.1186/s40824-023-00457-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/29/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a hematological malignancy that remains a therapeutic challenge due to the high incidence of disease relapse. To better understand resistance mechanisms and identify novel therapies, robust preclinical models mimicking the bone marrow (BM) microenvironment are needed. This study aimed to achieve an automated fabrication process of a three-dimensional (3D) AML disease model that recapitulates the 3D spatial structure of the BM microenvironment and applies to drug screening and investigational studies. METHODS To build this model, we investigated a unique class of tetramer peptides with an innate ability to self-assemble into stable hydrogel. An automated robotic bioprinting process was established to fabricate a 3D BM (niche-like) multicellular AML disease model comprised of leukemia cells and the BM's stromal and endothelial cellular fractions. In addition, monoculture and dual-culture models were also fabricated. Leukemia cell compatibility, functionalities (in vitro and in vivo), and drug assessment studies using our model were performed. In addition, RNAseq and gene expression analysis using TaqMan arrays were also performed on 3D cultured stromal cells and primary leukemia cells. RESULTS The selected peptide hydrogel formed a highly porous network of nanofibers with mechanical properties similar to the BM extracellular matrix. The robotic bioprinter and the novel quadruple coaxial nozzle enabled the automated fabrication of a 3D BM niche-like AML disease model with controlled deposition of multiple cell types into the model. This model supported the viability and growth of primary leukemic, endothelial, and stromal cells and recapitulated cell-cell and cell-ECM interactions. In addition, AML cells in our model possessed quiescent characteristics with improved chemoresistance attributes, resembling more the native conditions as indicated by our in vivo results. Moreover, the whole transcriptome data demonstrated the effect of 3D culture on enhancing BM niche cell characteristics. We identified molecular pathways upregulated in AML cells in our 3D model that might contribute to AML drug resistance and disease relapse. CONCLUSIONS Our results demonstrate the importance of developing 3D biomimicry models that closely recapitulate the in vivo conditions to gain deeper insights into drug resistance mechanisms and novel therapy development. These models can also improve personalized medicine by testing patient-specific treatments.
Collapse
Affiliation(s)
- Dana M Alhattab
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- KAUST Smart Health Initiative (KSHI), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Ioannis Isaioglou
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Salwa Alshehri
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Zainab N Khan
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Red Sea Research Center (RSRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Hepi H Susapto
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Yanyan Li
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Yara Marghani
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Arwa A Alghuneim
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Rubén Díaz-Rúa
- Core Laboratories, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Sherin Abdelrahman
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Shuroug Al-Bihani
- Core Laboratories, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Farid Ahmed
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Raed I Felimban
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Heba Alkhatabi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Hematology Research Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Raed Alserihi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Malak Abedalthagafi
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, USA
| | - AlShaibani AlFadel
- Division of Hematology, Stem Cell Transplantation & Cellular Therapy, Oncology Center, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Abdalla Awidi
- Cell Therapy Center, The University of Jordan, Amman, Jordan
- Medical School, The University of Jordan, Amman, Jordan
- Jordan University Hospital, Amman, Jordan
| | - Adeel Gulzar Chaudhary
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Jasmeen Merzaban
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Charlotte A E Hauser
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
- KAUST Smart Health Initiative (KSHI), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
- Red Sea Research Center (RSRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
| |
Collapse
|
4
|
Martinez B, Peplow PV. Biomaterial and tissue-engineering strategies for the treatment of brain neurodegeneration. Neural Regen Res 2022; 17:2108-2116. [PMID: 35259816 PMCID: PMC9083174 DOI: 10.4103/1673-5374.336132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The incidence of neurodegenerative diseases is increasing due to changing age demographics and the incidence of sports-related traumatic brain injury is tending to increase over time. Currently approved medicines for neurodegenerative diseases only temporarily reduce the symptoms but cannot cure or delay disease progression. Cell transplantation strategies offer an alternative approach to facilitating central nervous system repair, but efficacy is limited by low in vivo survival rates of cells that are injected in suspension. Transplanting cells that are attached to or encapsulated within a suitable biomaterial construct has the advantage of enhancing cell survival in vivo. A variety of biomaterials have been used to make constructs in different types that included nanoparticles, nanotubes, microspheres, microscale fibrous scaffolds, as well as scaffolds made of gels and in the form of micro-columns. Among these, Tween 80-methoxy poly(ethylene glycol)-poly(lactic-co-glycolic acid) nanoparticles loaded with rhynchophylline had higher transport across a blood-brain barrier model and decreased cell death in an in vitro model of Alzheimer’s disease than rhynchophylline or untreated nanoparticles with rhynchophylline. In an in vitro model of Parkinson’s disease, trans-activating transcriptor bioconjugated with zwitterionic polymer poly(2-methacryoyloxyethyl phosphorylcholine) and protein-based nanoparticles loaded with non-Fe hemin had a similar protective ability as free non-Fe hemin. A positive effect on neuron survival in several in vivo models of Parkinson’s disease was associated with the use of biomaterial constructs such as trans-activating transcriptor bioconjugated with zwitterionic polymer poly(2-methacryoyloxyethyl phosphorylcholine) and protein-based nanoparticles loaded with non-Fe hemin, carbon nanotubes with olfactory bulb stem cells, poly(lactic-co-glycolic acid) microspheres with attached DI-MIAMI cells, ventral midbrain neurons mixed with short fibers of poly-(L-lactic acid) scaffolds and reacted with xyloglucan with/without glial-derived neurotrophic factor, ventral midbrain neurons mixed with Fmoc-DIKVAV hydrogel with/without glial-derived neurotrophic factor. Further studies with in vivo models of Alzheimer’s disease and Parkinson’s disease are warranted especially using transplantation of cells in agarose micro-columns with an inner lumen filled with an appropriate extracellular matrix material.
Collapse
Affiliation(s)
- Bridget Martinez
- Department of Medicine, St. Georges University School of Medicine, Grenada
| | - Philip V Peplow
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
5
|
Abstract
In this highlight, we describe the construction of supramolecular single/double/triple-helical assemblies from small di/tri/tetrapeptides and their applications.
Collapse
Affiliation(s)
- Rajat Subhra Giri
- Department of Chemistry, Laboratory of Peptide and Amyloid Research, Indian Institute of Technology Guwahati, Assam-781039, India
| | - Bhubaneswar Mandal
- Department of Chemistry, Laboratory of Peptide and Amyloid Research, Indian Institute of Technology Guwahati, Assam-781039, India
| |
Collapse
|
6
|
Yang S, Zhu J, Lu C, Chai Y, Cao Z, Lu J, Zhang Z, Zhao H, Huang YY, Yao S, Kong X, Zhang P, Wang X. Aligned fibrin/functionalized self-assembling peptide interpenetrating nanofiber hydrogel presenting multi-cues promotes peripheral nerve functional recovery. Bioact Mater 2021; 8:529-544. [PMID: 34541418 PMCID: PMC8435993 DOI: 10.1016/j.bioactmat.2021.05.056] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/07/2021] [Accepted: 05/28/2021] [Indexed: 12/19/2022] Open
Abstract
Nerve guidance conduits with hollow lumen fail to regenerate critical-sized peripheral nerve defects (15 mm in rats and 25 mm in humans), which can be improved by a beneficial intraluminal microenvironment. However, individual cues provided by intraluminal filling materials are inadequate to eliminate the functional gap between regenerated nerves and normal nerves. Herein, an aligned fibrin/functionalized self-assembling peptide (AFG/fSAP) interpenetrating nanofiber hydrogel that exerting synergistic topographical and biochemical cues for peripheral nerve regeneration is constructed via electrospinning and molecular self-assembly. The hydrogel possesses an aligned structure, high water content, appropriate mechanical properties and suitable biodegradation capabilities for nerve repair, which enhances the alignment and neurotrophin secretion of primary Schwann cells (SCs) in vitro, and successfully bridges a 15-mm sciatic nerve gap in rats in vivo. The rats transplanted with the AFG/fSAP hydrogel exhibit satisfactory morphological and functional recovery in myelinated nerve fibers and innervated muscles. The motor function recovery facilitated by the AFG/fSAP hydrogel is comparable with that of autografts. Moreover, the AFG/fSAP hydrogel upregulates the regeneration-associated gene expression and activates the PI3K/Akt and MAPK signaling pathways in the regenerated nerve. Altogether, the AFG/fSAP hydrogel represents a promising approach for peripheral nerve repair through an integration of structural guidance and biochemical stimulation. A novel aligned interpenetrating nanofiber hydrogel is first constructed for peripheral nerve regeneration. The aligned hydrogel presents synergistic topographical and biochemical cues for peripheral nerve regeneration. Nerve conduits filled with the aligned hydrogel can repair the long-distance sciatic nerve defects in 12 weeks. The function recovery facilitated by the aligned hydrogel is comparable with that of autografts. The aligned hydrogel upregulates regeneration-related genes and activates the PI3K/Akt and MAPK signaling pathways.
Collapse
Affiliation(s)
- Shuhui Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Jinjin Zhu
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China.,Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang, Hangzhou, 310016, PR China
| | - Changfeng Lu
- Key Laboratory of Trauma and Neural Regeneration, Peking University, Ministry of Education, Department of Trauma and Orthopedics, Peking University People's Hospital, Beijing, 100044, PR China
| | - Yi Chai
- School of Clinical Medicine, Tsinghua University, Beijing, 100084, PR China
| | - Zheng Cao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Jiaju Lu
- School of Materials Science and Engineering, Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - Zhe Zhang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - He Zhao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Yin-Yuan Huang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| | - Shenglian Yao
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, PR China
| | - Xiangdong Kong
- School of Materials Science and Engineering, Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, PR China
| | - Peixun Zhang
- Key Laboratory of Trauma and Neural Regeneration, Peking University, Ministry of Education, Department of Trauma and Orthopedics, Peking University People's Hospital, Beijing, 100044, PR China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, PR China
| |
Collapse
|
7
|
Zhang S. Self-assembling peptides: From a discovery in a yeast protein to diverse uses and beyond. Protein Sci 2020; 29:2281-2303. [PMID: 32939884 PMCID: PMC7586918 DOI: 10.1002/pro.3951] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 12/11/2022]
Abstract
Well-defined nanofiber scaffold hydrogels made of self-assembling peptides have found their way into various 3D tissue culture and clinical products. I reflect initial puzzlement of the unexpected discovery, gradual understanding of how these peptides undergo self-assembly, to eventually translating designer biological scaffolds into commercial products. Peptides are ubiquitous in nature and useful in many fields. They are found as hormones, pheromones, antibacterial, and antifungal agents in innate immunity systems, toxins, as well anti-inset pesticides. However, the concept of peptides as materials was not recognized until 1990 when a self-assembling peptide as a repeating segment in a yeast protein was serendipitously discovered. The peptide materials have bona fide materials properties and are made from simple amino acids with well-ordered nanostructures under physiological conditions. Some current applications include: (a) Real 3D tissue cell cultures of diverse tissue cells and various stem cells; (b) reparative and regenerative medicine as well as tissue engineering; (c) 3D tissue printing; (d) sustained releases of small molecules, growth factors and monoclonal antibodies; and (e) accelerated wound healing of skin and diabetic ulcers as well as instant hemostasis in surgery. Self-assembling peptide nanobiotechnology will likely continue to expand in many directions in the coming years. I will also briefly introduce my current research using a simple QTY code for membrane protein design. I am greatly honored and humbled to be invited to contribute an Award Winner Recollection of the 2020 Emil Thomas Kaiser Award from the Protein Society.
Collapse
Affiliation(s)
- Shuguang Zhang
- Laboratory of Molecular ArchitectureMedia Lab, Massachusetts Institute of Technology77 Massachusetts Avenue E15‐391CambridgeMassachusetts02139‐4306USA
| |
Collapse
|
8
|
Yang S, Wang C, Zhu J, Lu C, Li H, Chen F, Lu J, Zhang Z, Yan X, Zhao H, Sun X, Zhao L, Liang J, Wang Y, Peng J, Wang X. Self-assembling peptide hydrogels functionalized with LN- and BDNF- mimicking epitopes synergistically enhance peripheral nerve regeneration. Theranostics 2020; 10:8227-8249. [PMID: 32724468 PMCID: PMC7381722 DOI: 10.7150/thno.44276] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 05/31/2020] [Indexed: 12/16/2022] Open
Abstract
The regenerative capacity of the peripheral nervous system is closely related to the role that Schwann cells (SCs) play in construction of the basement membrane containing multiple extracellular matrix proteins and secretion of neurotrophic factors, including laminin (LN) and brain-derived neurotrophic factor (BDNF). Here, we developed a self-assembling peptide (SAP) nanofiber hydrogel based on self-assembling backbone Ac-(RADA)4-NH2 (RAD) dual-functionalized with laminin-derived motif IKVAV (IKV) and a BDNF-mimetic peptide epitope RGIDKRHWNSQ (RGI) for peripheral nerve regeneration, with the hydrogel providing a three-dimensional (3D) microenvironment for SCs and neurites. Methods: Circular dichroism (CD), atomic force microscopy (AFM), and scanning electron microscopy (SEM) were used to characterize the secondary structures, microscopic structures, and morphologies of self-assembling nanofiber hydrogels. Then the SC adhesion, myelination and neurotrophin secretion were evaluated on the hydrogels. Finally, the SAP hydrogels were injected into hollow chitosan tubes to bridge a 10-mm-long sciatic nerve defect in rats, and in vivo gene expression at 1 week, axonal regeneration, target muscular re-innervation, and functional recovery at 12 weeks were assessed. Results: The bioactive peptide motifs were covalently linked to the C-terminal of the self-assembling peptide and the functionalized peptides could form well-defined nanofibrous hydrogels capable of providing a 3D microenvironment similar to native extracellular matrix. SCs displayed improved cell adhesion on hydrogels with both IKV and RGI, accompanied by increased cell spreading and elongation relative to other groups. RSCs cultured on hydrogels with IKV and RGI showed enhanced gene expression of NGF, BDNF, CNTF, PMP22 and NRP2, and decreased gene expression of NCAM compared with those cultured on other three groups after a 7-day incubation. Additionally, the secretion of NGF, BDNF, and CNTF of RSCs was significantly improved on dual-functionalized peptide hydrogels after 3 days. At 1 week after implantation, the expressions of neurotrophin and myelin-related genes in the nerve grafts in SAP and Autograft groups were higher than that in Hollow group, and the expression of S100 in groups containing both IKV and RGI was significantly higher than that in groups containing either IKV or RGI hydrogels, suggesting enhanced SC proliferation. The morphometric parameters of the regenerated nerves, their electrophysiological performance, the innervated muscle weight and remodeling of muscle fibers, and motor function showed that RAD/IKV/RGI and RAD/IKV-GG-RGI hydrogels could markedly improve axonal regeneration with enhanced re-myelination and motor functional recovery through the synergetic effect of IKV and RGI functional motifs. Conclusions: We found that the dual-functionalized SAP hydrogels promoted RSC adhesion, myelination, and neurotrophin secretion in vitro and successfully bridged a 10-mm gap representing a sciatic nerve defect in rats in vivo. The results demonstrated the synergistic effect of IKVAV and RGI on axonal regrowth and function recovery after peripheral nerve injury.
Collapse
Affiliation(s)
- Shuhui Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Chong Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226007, China
| | - Jinjin Zhu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang, Hangzhou 310016, China
| | - Changfeng Lu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226007, China
- Department of Orthopaedics and Trauma, Peking University People's Hospital, Beijing 100191, China
| | - Haitao Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226007, China
| | - Fuyu Chen
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226007, China
| | - Jiaju Lu
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Zhe Zhang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xiaoqing Yan
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
- School of Clinical Medicine, Tsinghua University, Beijing 100084, China
| | - He Zhao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xiaodan Sun
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Lingyun Zhao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Jing Liang
- Department of Pediatrics, Tianjin Hospital, Tianjin University, No. 406 Jiefang Nan Road, Tianjin 300211, China
| | - Yu Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226007, China
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226007, China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| |
Collapse
|
9
|
Giri RS, Pal S, Roy S, Dolai G, Manne SR, Paul S, Mandal B. Nanostructures from protected L/L and D/L amino acid containing dipeptides. Pept Sci (Hoboken) 2020. [DOI: 10.1002/pep2.24176] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Rajat Subhra Giri
- Department of Chemistry, Laboratory of Peptide and Amyloid Research Indian Institute of Technology Guwahati Guwahati India
| | - Saikat Pal
- Department of Chemistry Indian Institute of Technology Guwahati Guwahati India
| | - Sayanta Roy
- Department of Chemistry, Laboratory of Peptide and Amyloid Research Indian Institute of Technology Guwahati Guwahati India
| | - Gobinda Dolai
- Department of Chemistry, Laboratory of Peptide and Amyloid Research Indian Institute of Technology Guwahati Guwahati India
| | - Srinivasa Rao Manne
- Department of Chemistry, Laboratory of Peptide and Amyloid Research Indian Institute of Technology Guwahati Guwahati India
| | - Sandip Paul
- Department of Chemistry Indian Institute of Technology Guwahati Guwahati India
| | - Bhubaneswar Mandal
- Department of Chemistry, Laboratory of Peptide and Amyloid Research Indian Institute of Technology Guwahati Guwahati India
| |
Collapse
|
10
|
A novel silk/PES hybrid nanofibrous scaffold promotes the in vitro proliferation and differentiation of adipose‐derived mesenchymal stem cells into insulin producing cells. POLYM ADVAN TECHNOL 2020. [DOI: 10.1002/pat.4912] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
11
|
Guo WW, Zhang ZT, Wei Q, Zhou Y, Lin MT, Chen JJ, Wang TT, Guo NN, Zhong XC, Lu YY, Yang QY, Han M, Gao J. Intracellular Restructured Reduced Glutathione-Responsive Peptide Nanofibers for Synergetic Tumor Chemotherapy. Biomacromolecules 2020; 21:444-453. [PMID: 31851512 DOI: 10.1021/acs.biomac.9b01202] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Self-assembled peptide nanofibers have been widely studied in cancer nanotherapeutics with their excellent biocompatibility and low toxicity of degradation products, showing the significant potential in inhibiting tumor progression. However, poor solubility prevents direct intravenous administration of nanofibers. Although water-soluble peptide precursors have been formed via the method of phosphorylation for intravenous administration, their opportunities for broad in vivo application are limited by the weak capacity of encapsulating drugs. Herein, we designed a novel restructured reduced glutathione (GSH)-responsive drug delivery system encapsulating doxorubicin for systemic administration, which achieved the intracellular restructuration from three-dimensional micelles into one-dimensional nanofibers. After a long blood circulation, micelles endocytosed by tumor cells could degrade in response to high GSH levels, achieving more release and accumulation of doxorubicin at desired sites. Further, the synergistic chemotherapy effects of self-assembled nanofibers were confirmed in both in vitro and in vivo experiments.
Collapse
|
12
|
Bhartiya A, Madi K, Disney CM, Courtois L, Jupe A, Zhang F, Bodey AJ, Lee P, Rau C, Robinson IK, Yusuf M. Phase-contrast 3D tomography of HeLa cells grown in PLLA polymer electrospun scaffolds using synchrotron X-rays. JOURNAL OF SYNCHROTRON RADIATION 2020; 27:158-163. [PMID: 31868748 DOI: 10.1107/s1600577519015583] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 11/18/2019] [Indexed: 06/10/2023]
Abstract
Advanced imaging is useful for understanding the three-dimensional (3D) growth of cells. X-ray tomography serves as a powerful noninvasive, nondestructive technique that can fulfill these purposes by providing information about cell growth within 3D platforms. There are a limited number of studies taking advantage of synchrotron X-rays, which provides a large field of view and suitable resolution to image cells within specific biomaterials. In this study, X-ray synchrotron radiation microtomography at Diamond Light Source and advanced image processing were used to investigate cellular infiltration of HeLa cells within poly L-lactide (PLLA) scaffolds. This study demonstrates that synchrotron X-rays using phase contrast is a useful method to understand the 3D growth of cells in PLLA electrospun scaffolds. Two different fiber diameter (2 and 4 µm) scaffolds with different pore sizes, grown over 2, 5 and 8 days in vitro, were examined for infiltration and cell connectivity. After performing visualization by segmentation of the cells from the fibers, the results clearly show deeper cell growth and higher cellular interconnectivity in the 4 µm fiber diameter scaffold. This indicates the potential for using such 3D technology to study cell-scaffold interactions for future medical use.
Collapse
Affiliation(s)
- A Bhartiya
- London Centre for Nanotechnology, University College London, London WC1H 0AH, UK
| | - K Madi
- 3DMagination Ltd, Atlas Building, Fermi Avenue, Harwell, Didcot OX11 0QX, UK
| | - C M Disney
- School of Mechanical, Aerospace and Civil Engineering, University of Manchester, Manchester, UK
| | - L Courtois
- 3DMagination Ltd, Atlas Building, Fermi Avenue, Harwell, Didcot OX11 0QX, UK
| | - A Jupe
- Department of Applied Computing, The University of Buckingham, UK
| | - F Zhang
- London Centre for Nanotechnology, University College London, London WC1H 0AH, UK
| | - A J Bodey
- Diamond Light Source, Oxfordshire OX11 0DE, UK
| | - P Lee
- Mechanical Engineering, University College London, London WC1E 7JE, UK
| | - C Rau
- Diamond Light Source, Oxfordshire OX11 0DE, UK
| | - I K Robinson
- London Centre for Nanotechnology, University College London, London WC1H 0AH, UK
| | - M Yusuf
- London Centre for Nanotechnology, University College London, London WC1H 0AH, UK
| |
Collapse
|
13
|
Barati G, Rahmani A, Nadri S. In vitro differentiation of conjunctiva mesenchymal stem cells into insulin producing cells on natural and synthetic electrospun scaffolds. Biologicals 2019; 62:33-38. [PMID: 31635936 DOI: 10.1016/j.biologicals.2019.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 10/04/2019] [Accepted: 10/14/2019] [Indexed: 12/29/2022] Open
Abstract
Polymers are used in tissue engineering as a scaffold. In this study the differentiation capability of conjunctiva mesenchymal stem cells (CJMSCs) on natural and synthetic nanofibrous electrospun scaffolds into insulin producing cells (IPCs) were studied. Natural Silk fibroin and synthetic PLLA polymers were used to fabricate electrospun scaffolds. These scaffolds are characterized by SEM and CJMSCs were differentiated into IPCs on these scaffolds. The differentiation efficiency was measured by analysis the expression of specific pancreatic markers by RT-qPCR and insulin release capacity via ELISA. Microscopy analysis showed the fabrication of uniform nanofibers and the formation of the islet-like clusters at the end of differentiation period. Significant differences in expression of Pdx-1 and glucagon were observed in PLLA scaffold compared to Silk scaffold (Fold: 1.625 and 1.434, respectively; P-value ≤ 0.0001 for both). Furthermore, insulin secretion at high glucose concentration was significantly higher in cells differentiated on PLLA scaffold than those cultured on Silk scaffold (P-value: 0.012). The scaffolds can enhance the differentiation of IPCs from CJMSCs. In this way, PLLA synthetic scaffold was more efficient than Silk natural scaffold. We conclude that the nanofibrous scaffolds reported herein could be used as a potential supportive matrix for islet tissue engineering.
Collapse
Affiliation(s)
- Ghasem Barati
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Rahmani
- Department of Medical Nanotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Samad Nadri
- Department of Medical Nanotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran; Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran; Zanjan Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
14
|
Klein A, Sagi-Assif O, Meshel T, Telerman A, Izraely S, Ben-Menachem S, Bayry J, Marzese DM, Ohe S, Hoon DSB, Erez N, Witz IP. CCR4 is a determinant of melanoma brain metastasis. Oncotarget 2018; 8:31079-31091. [PMID: 28415693 PMCID: PMC5458190 DOI: 10.18632/oncotarget.16076] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/02/2017] [Indexed: 01/21/2023] Open
Abstract
We previously identified the chemokine receptor CCR4 as part of the molecular signature of melanoma brain metastasis. The aim of this study was to determine the functional significance of CCR4 in melanoma brain metastasis. We show that CCR4 is more highly expressed by brain metastasizing melanoma cells than by local cutaneous cells from the same melanoma. Moreover, we found that the expression of CCR4 is significantly higher in paired clinical specimens of melanoma metastases than in samples of primary tumors from the same patients. Notably, the expression of the CCR4 ligands, Ccl22 and Ccl17 is upregulated at the earliest stages of brain metastasis, and precedes the infiltration of melanoma cells to the brain. In-vitro, CCL17 induced migration and transendothelial migration of melanoma cells. Functionally, human melanoma cells over-expressing CCR4 were more tumorigenic and produced a higher load of spontaneous brain micrometastasis than control cells. Blocking CCR4 with a small molecule CCR4 antagonist in-vivo, reduced the tumorigenicity and micrometastasis formation of melanoma cells. Taken together, these findings implicate CCR4 as a driver of melanoma brain metastasis.
Collapse
Affiliation(s)
- Anat Klein
- Department of Cell Research and Immunology, George S. Wise, Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| | - Orit Sagi-Assif
- Department of Cell Research and Immunology, George S. Wise, Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| | - Tsipi Meshel
- Department of Cell Research and Immunology, George S. Wise, Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| | - Alona Telerman
- Department of Cell Research and Immunology, George S. Wise, Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| | - Sivan Izraely
- Department of Cell Research and Immunology, George S. Wise, Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| | - Shlomit Ben-Menachem
- Department of Cell Research and Immunology, George S. Wise, Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| | - Jagadeesh Bayry
- Inserm Unité 1138, Center de Recherche des Cordeliers, Université Pierre et Marie Curie, Université, Paris Descartes, Paris, France
| | - Diego M Marzese
- Department of Molecular Oncology, John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Shuichi Ohe
- Department of Molecular Oncology, John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Dave S B Hoon
- Department of Molecular Oncology, John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Neta Erez
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Isaac P Witz
- Department of Cell Research and Immunology, George S. Wise, Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|
15
|
Pan M, Wang X, Chen Y, Cao S, Wen J, Wu G, Li Y, Li L, Qian C, Qin Z, Li Z, Tan D, Fan Z, Wu W, Guo J. Tissue engineering with peripheral blood-derived mesenchymal stem cells promotes the regeneration of injured peripheral nerves. Exp Neurol 2017; 292:92-101. [DOI: 10.1016/j.expneurol.2017.03.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 02/08/2017] [Accepted: 03/06/2017] [Indexed: 12/11/2022]
|
16
|
Jamal A, Nyrkova I, Mesini P, Militzer S, Reiter G. Solvent-controlled reversible switching between adsorbed self-assembled nanoribbons and nanotubes. NANOSCALE 2017; 9:3293-3303. [PMID: 28225113 DOI: 10.1039/c6nr08211d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
We have demonstrated that solutions of 3,5-bis-(5-hexylcarbamoylpentyloxy)-benzoic acid decyl ester (BHPB-10) can form metastable nanostructures on solid substrates and in the bulk. BHPB-10 is an achiral molecule involving several distinct, strongly interacting groups (SIGs), one aromatic-ester ring and two amide groups per molecule. Specific solvents affect the interactions between particular SIGs, thus promoting various nano-structures: lamellae, nanoribbons, helical ribbons, or nanotubes. In cyclohexane, a solvent allowing for both inter-amide hydrogen bonds and mutual attraction of rings, the formation of nanotubes with a diameter of 28 ± 5 nm was observed in the bulk and on surfaces. By contrast, in cyclohexanone, which suppresses inter-amide hydrogen bonds, flat nanoribbons with a specific width of 12 ± 4 nm were formed on solid substrates after drying. By annealing in cyclohexane vapor, we followed the process of switching surface structures from nanoribbons to nanotubes and observed helical ribbons as the precursor of nanotubes. We also turned nanotubes back into nanoribbons by adding cyclohexanone, thus demonstrating reversible switching along the route: tubes → lamellae → flat ribbons → helical ribbons → tubes. We propose models explaining the observed nanostructures and their transformations, including the origin of spontaneous chirality of the helical ribbons. Our findings on the self-assembly in the achiral BHPB-10 solutions provide insight into the influence of complementary inter-molecular specific SIG-based interactions and demonstrate an effective route for tailoring the shape and size of nanostructures derived from the same building unit.
Collapse
Affiliation(s)
- Asad Jamal
- Institute of Physics, University of Freiburg, Herman-Herder-Strasse 3, 79104 Freiburg, Germany. and Freiburg Materials Research Center (FMF), University of Freiburg, Stefan-Meier-Strasse 21, 79104 Freiburg, Germany
| | - Irina Nyrkova
- Institut Charles Sadron, 23 rue du Loess BP 84047, F-67034 Strasbourg Cedex 2, France
| | - Philippe Mesini
- Institut Charles Sadron, 23 rue du Loess BP 84047, F-67034 Strasbourg Cedex 2, France
| | - Swann Militzer
- Institut Charles Sadron, 23 rue du Loess BP 84047, F-67034 Strasbourg Cedex 2, France
| | - Günter Reiter
- Institute of Physics, University of Freiburg, Herman-Herder-Strasse 3, 79104 Freiburg, Germany. and Freiburg Materials Research Center (FMF), University of Freiburg, Stefan-Meier-Strasse 21, 79104 Freiburg, Germany
| |
Collapse
|
17
|
Liu Y, Zhang L, Wei W. Effect of noncovalent interaction on the self-assembly of a designed peptide and its potential use as a carrier for controlled bFGF release. Int J Nanomedicine 2017; 12:659-670. [PMID: 28176898 PMCID: PMC5261598 DOI: 10.2147/ijn.s124523] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Peptide self-assembly is one of the promising bottom-up approaches for creating synthetic supermolecular architectures. Noncovalent interactions such as hydrophobic packing, electrostatic interaction, and polypeptide chain entropy (ΔSC) are the most relevant factors that affect the folding and self-assembly of peptides and the stability of supermolecular structures. The GVGV tetrapeptide is an abundant repeat in elastin, an extracellular matrix protein. In this study, four GVGV-containing peptides were designed with the aim of understanding the effects of these weak interactions on peptide self-assembly. Transmission electron microscopy, circular dichroism spectroscopy, dynamic light scattering measurements, and rheometry assays were used to study the structural features of the peptides. Because self-assembling peptides with different amino acid sequences may significantly affect protein release, basic fibroblast growth factor (bFGF) was used as a model molecule and encapsulated within the P2 (RLDLGVGVRLDLGVGV) hydrogel to study the release kinetics. The results showed that the balance among hydrophobic effects, electrostatic interactions, and chain entropy determined the molecular state and self-assembly of the peptide. Moreover, encapsulation of bFGF within the P2 hydrogel allowed its sustained release without causing changes in the secondary structure. The release profiles could be tuned by adjusting the P2 hydrogel concentration. Cell Counting Kit-8 and Western blot assays demonstrated that the encapsulated and released bFGFs were biologically active and capable of promoting the proliferation of murine fibroblast NIH-3T3 cells, most likely due to the activation of downstream signaling pathways.
Collapse
Affiliation(s)
- Yanfei Liu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, People’s Republic of China
| | - Ling Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, People’s Republic of China
| | - Wei Wei
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, People’s Republic of China
| |
Collapse
|
18
|
Zweckberger K, Ahuja CS, Liu Y, Wang J, Fehlings MG. Self-assembling peptides optimize the post-traumatic milieu and synergistically enhance the effects of neural stem cell therapy after cervical spinal cord injury. Acta Biomater 2016; 42:77-89. [PMID: 27296842 DOI: 10.1016/j.actbio.2016.06.016] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 06/01/2016] [Accepted: 06/09/2016] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The hostile environment after spinal cord injury (SCI) can compromise effects of regenerative therapies. We hypothesized that optimizing the post-traumatic environment with QL6 self-assembling peptides (SAPs) before neural precursor cell (NPC) transplantation would improve cell survival, differentiation and functional recovery. METHODS A total of 90 Wistar rats received a clip-compression SCI at C7. Within each of two study arms, animals were randomized into 5 groups (NPC, SAP, NPC+SAP, vehicle, and sham). SAPs and NPCs were injected into the spinal cord 1day and 14days post-injury, respectively. Animals received growth factors over 7days and were immunosuppressed. Rats were sacrificed at 4weeks and sections of the cervical spinal cord prepared for immunohistochemistry (first study arm). Neurological function was assessed weekly for 8weeks using a battery of behavioral tests. Nine weeks post-SCI, the corticospinal tract was assessed using fiber-tracking (second arm). RESULTS SAP-treated animals had significantly more surviving NPCs which showed increased differentiation to neurons and oligodendrocytes compared to controls. SAPs alone or in combination with NPCs resulted in smaller intramedullary cysts and larger volume of preserved tissue compared to other groups. The combined treatment group showed reduced astrogliosis and chondroitin sulfate proteoglycan deposition. Synaptic connectivity was increased in the NPC and combined treatment groups. Corticospinal tract preservation and behavioral outcomes improved with combinatorial treatment. CONCLUSION Injecting SAPs after SCI enhances subsequent NPC survival, integration and differentiation and improves functional recovery. STATEMENT OF SIGNIFICANCE The hostile environment after spinal cord injury (SCI) can compromise effects of regenerative therapies. We hypothesized that improving this environment with self-assembling peptides (SAPs) before neural precursor cell (NPC) transplantation would support their beneficial effects. SAPs assemble once injected, providing a supportive scaffold for repair and regeneration. We investigated this in a rat model of spinal cord injury. More NPCs survived in SAP-treated animals and these showed increased differentiation compared to controls. SAPS alone or in combination with NPCs resulted in smaller cysts and larger volume of preserved tissue with the combined treatment also reducing scarring and improving behavioral outcomes. Overall, injection of SAPs was shown to improve the efficacy of NPC treatment, a promising finding for those with SCIs.
Collapse
|
19
|
Abstract
Anti-microbial peptides (AMPs) were originally thought to exert protecting actions against bacterial infection by disintegrating bacterial membranes. Upon identification of internal bacterial targets, the view changed and moved toward inhibition of prokaryote-specific biochemical processes. However, the level of none of these activities can explain the robust efficacy of some of these peptides in animal models of systemic and cutaneous infections. A rapidly growing panel of reports suggests that AMPs, now called host-defense peptides (HDPs), act through activating the immune system of the host. This includes recruitment and activation of macrophages and mast cells, inducing chemokine production and altering NF-κB signaling processes. As a result, both pro- and anti-inflammatory responses are elevated together with activation of innate and adaptive immunity mechanisms, wound healing, and apoptosis. HDPs sterilize the systemic circulation and local injury sites significantly more efficiently than pure single-endpoint in vitro microbiological or biochemical data would suggest and actively aid recovering from tissue damage after or even without bacterial infections. However, the multiple and, often opposing, immunomodulatory functions of HDPs require exceptional care in therapeutic considerations.
Collapse
Affiliation(s)
- Laszlo Otvos
- 1 Olpe LLC, Audubon, PA, USA
- 2 Institute of Medical Microbiology , Semmelweis University , Budapest, Hungary
| |
Collapse
|
20
|
Kim JE, Jung KM, Kim SH, Jung Y. Combined Treatment with Systemic and Local Delivery of Substance P Coupled with Self-Assembled Peptides for a Hind Limb Ischemia Model. Tissue Eng Part A 2016; 22:545-55. [DOI: 10.1089/ten.tea.2015.0412] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Ji Eun Kim
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
- NBIT, KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Ki Moon Jung
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Soo Hyun Kim
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
- NBIT, KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
- Department of Biomedical Engineering, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Youngmee Jung
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Department of Biomedical Engineering, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
21
|
Osteogenesis of peripheral blood mesenchymal stem cells in self assembling peptide nanofiber for healing critical size calvarial bony defect. Sci Rep 2015; 5:16681. [PMID: 26568114 PMCID: PMC4645224 DOI: 10.1038/srep16681] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 10/16/2015] [Indexed: 02/08/2023] Open
Abstract
Peripheral blood mesenchymal stem cells (PBMSCs) may be easily harvested from patients, permitting autologous grafts for bone tissue engineering in the future. However, the PBMSC’s capabilities of survival, osteogenesis and production of new bone matrix in the defect area are still unclear. Herein, PBMSCs were seeded into a nanofiber scaffold of self-assembling peptide (SAP) and cultured in osteogenic medium. The results indicated SAP can serve as a promising scaffold for PBMSCs survival and osteogenic differentiation in 3D conditions. Furthermore, the SAP seeded with the induced PBMSCs was splinted by two membranes of poly(lactic)-glycolic acid (PLGA) to fabricate a composited scaffold which was then used to repair a critical-size calvarial bone defect model in rat. Twelve weeks later the defect healing and mineralization were assessed by H&E staining and microcomputerized tomography (micro-CT). The osteogenesis and new bone formation of grafted cells in the scaffold were evaluated by immunohistochemistry. To our knowledge this is the first report with solid evidence demonstrating PBMSCs can survive in the bone defect area and directly contribute to new bone formation. Moreover, the present data also indicated the tissue engineering with PBMSCs/SAP/PLGA scaffold can serve as a novel prospective strategy for healing large size cranial defects.
Collapse
|
22
|
AP-1-Targeted Anti-Inflammatory Activities of the Nanostructured, Self-Assembling S5 Peptide. Mediators Inflamm 2015; 2015:451957. [PMID: 26074678 PMCID: PMC4446838 DOI: 10.1155/2015/451957] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 04/27/2015] [Accepted: 04/27/2015] [Indexed: 01/24/2023] Open
Abstract
Peptide-based therapeutics have received increasing attention in medical research. However, the local delivery of such therapeutics poses unique challenges. Self-assembling peptides that use decorated nanofibers are one approach by which these therapeutics may be delivered. We previously found that the self-assembling K5 peptide affects the anti-inflammatory response. The aim of the present study was to investigate another self-assembling peptide, S5. Unlike the K5 peptide which has a positive charge, the S5 peptide has a free hydroxyl (-OH) group. We first examined whether the S5 peptide regulates the inflammatory response in primary cells and found that the S5 peptide reduced the production of prostaglandin E2 (PGE2) and tumor necrosis factor (TNF)-α in lipopolysaccharide- (LPS-) treated bone marrow-derived macrophages. Moreover, the S5 peptide significantly downregulated cyclooxygenase- (COX-) 2, TNF-α, and interleukin- (IL-) 1β expression by blocking the nuclear translocation of c-Jun. Consistent with this finding, the S5 peptide diminished the activation of inflammatory signaling enzymes related to p38. The S5 peptide also inhibited the formation of the p38/c-Jun signaling complex in RAW264.7 cells. Similarly, p38 and MKK3/6 were inhibited by the S5 peptide in LPS-activated peritoneal macrophages. Taken together, these results strongly suggest that the S5 peptide could exert anti-inflammatory effects by inhibiting the c-Jun/p38 signaling pathway.
Collapse
|
23
|
Otvos L, Ostorhazi E. Therapeutic utility of antibacterial peptides in wound healing. Expert Rev Anti Infect Ther 2015; 13:871-81. [PMID: 25835521 DOI: 10.1586/14787210.2015.1033402] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cationic antimicrobial peptides were first thought to fight infection in animal models by disintegrating bacterial peptides and later by inhibiting bacteria-specific intracellular processes. However, ever increasing evidences indicate that cationic peptides accumulate around and modulate the immune system both systemically and in cutaneous and mucosal surfaces where injuries and infections occur. Native and designer antibacterial peptides as well as cationic peptides, never considered as antibiotics, promote wound healing at every step of cutaneous tissue regeneration. This article provides an introductory list of examples of how cationic peptides are involved in immunostimulation and epithelial tissue repair, eliminating wound infections and promoting wound healing in potential therapeutic utility in sight. Although a few antimicrobial peptides reached the Phase II clinical trial stage, toxicity concerns limit the potential administration routes. Resistance induction to both microbiology actions and the integrity of the innate immune system has to be carefully monitored.
Collapse
|
24
|
Wang X, Pan M, Wen J, Tang Y, Hamilton AD, Li Y, Qian C, Liu Z, Wu W, Guo J. A novel artificial nerve graft for repairing long-distance sciatic nerve defects: a self-assembling peptide nanofiber scaffold-containing poly(lactic-co-glycolic acid) conduit. Neural Regen Res 2015; 9:2132-41. [PMID: 25657734 PMCID: PMC4316446 DOI: 10.4103/1673-5374.147944] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2014] [Indexed: 01/20/2023] Open
Abstract
In this study, we developed a novel artificial nerve graft termed self-assembling peptide nanofiber scaffold (SAPNS)-containing poly(lactic-co-glycolic acid) (PLGA) conduit (SPC) and used it to bridge a 10-mm-long sciatic nerve defect in the rat. Retrograde tracing, behavioral testing and histomorphometric analyses showed that compared with the empty PLGA conduit implantation group, the SPC implantation group had a larger number of growing and extending axons, a markedly increased diameter of regenerated axons and a greater thickness of the myelin sheath in the conduit. Furthermore, there was an increase in the size of the neuromuscular junction and myofiber diameter in the target muscle. These findings suggest that the novel artificial SPC nerve graft can promote axonal regeneration and remyelination in the transected peripheral nerve and can be used for repairing peripheral nerve injury.
Collapse
Affiliation(s)
- Xianghai Wang
- Department of Histology and Embryology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Mengjie Pan
- Department of Histology and Embryology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jinkun Wen
- Department of Histology and Embryology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yinjuan Tang
- Department of Histology and Embryology, Xiangnan University, Chenzhou, Hunan Province, China
| | - Audra D Hamilton
- Department of Neurology, Vanderbilt University, Nashville, TN, USA
| | - Yuanyuan Li
- Department of Histology and Embryology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Changhui Qian
- Department of Histology and Embryology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhongying Liu
- Department of Histology and Embryology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Wutian Wu
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, Guangzhou, Guangdong Province, China ; GHM Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Jiasong Guo
- Department of Histology and Embryology, Southern Medical University, Guangzhou, Guangdong Province, China ; Key Laboratory of Tissue Construction and Detection of Guangdong Province, Guangzhou, Guangdong Province, China ; Institute of Bone Biology, Academy of Orthopedics, Guangzhou, Guangdong Province, China
| |
Collapse
|
25
|
Kim JE, Lee SM, Kim SH, Tatman P, Gee AO, Kim DH, Lee KE, Jung Y, Kim SJ. Effect of self-assembled peptide-mesenchymal stem cell complex on the progression of osteoarthritis in a rat model. Int J Nanomedicine 2014; 9 Suppl 1:141-57. [PMID: 24872709 PMCID: PMC4024982 DOI: 10.2147/ijn.s54114] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
PURPOSE To evaluate the efficacy of mesenchymal stem cells (MSCs) encapsulated in self-assembled peptide (SAP) hydrogels in a rat knee model for the prevention of osteoarthritis (OA) progression. MATERIALS AND METHODS Nanostructured KLD-12 SAPs were used as the injectable hydrogels. Thirty-three Sprague Dawley rats were used for the OA model. Ten rats were used for the evaluation of biotin-tagged SAP disappearance. Twenty-three rats were divided into four groups: MSC (n=6), SAP (n=6), SAP-MSC (n=6), and no treatment (n=5). MSCs, SAPs, and SAP-MSCs were injected into the knee joints 3 weeks postsurgery. Histologic examination, immunofluorescent staining, measurement of cytokine levels, and micro-computed tomography analysis were conducted 6 weeks after injections. Behavioral studies were done to establish baseline measurements before treatment, and repeated 3 and 6 weeks after treatment to measure the efficacy of SAP-MSCs. RESULTS Concentration of biotinylated SAP at week 1 was not significantly different from those at week 3 and week 6 (P=0.565). Bone mineral density was significantly lower in SAP-MSC groups than controls (P=0.002). Significant differences in terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick-end labeling staining between the control group and all other groups were observed. Caspase-8, tissue inhibitor of metalloproteinases 1, and matrix metalloproteinase 9 were diffusely stained in controls, whereas localized or minimal staining was observed in other groups. Modified Mankin scores were significantly lower in the SAP and SAP-MSC groups than in controls (P=0.001 and 0.013). Although not statistically significant, synovial inflammation scores were lower in the SAP (1.3±0.3) and SAP-MSC (1.3±0.2) groups than in controls (2.6±0.2). However, neither the cytokine level nor the behavioral score was significantly different between groups. CONCLUSION Injection of SAP-MSC hydrogels showed evidence of chondroprotection, as measured by the histologic grading and decreased expression of biochemical markers of inflammation and apoptosis. It also lowered subchondral bone mineral density, which can be increased by OA. This suggests that the SAP-MSC complex may have clinical potential to inhibit OA progression.
Collapse
Affiliation(s)
- Ji Eun Kim
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, South Korea
| | - Sang Mok Lee
- Department of Physical and Rehabilitation Medicine, Samsung Medical Center, Seoul, South Korea
| | - Soo Hyun Kim
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, South Korea
| | - Phil Tatman
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Albert O Gee
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA, USA
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA, USA ; Institute for Stem Cell and Regenerative Medicine and Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| | - Kyung Eun Lee
- Advanced Analysis Center, Korea Institute of Science and Technology, Seoul, South Korea
| | - Youngmee Jung
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, South Korea
| | - Sang Jun Kim
- Department of Physical and Rehabilitation Medicine, Samsung Medical Center, Seoul, South Korea
| |
Collapse
|
26
|
Cunha C, Panseri S, Gelain F. Engineering of a 3D nanostructured scaffold made of functionalized self-assembling peptides and encapsulated neural stem cells. Methods Mol Biol 2014; 1058:171-82. [PMID: 23526438 DOI: 10.1007/7651_2012_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Three-dimensional (3D) in vitro models of cell culture aim to fill the gap between the standard 2D cell studies and the in vivo environment. Especially for neural tissue regeneration approaches where there is little regenerative capacity, such models must rely on scaffolds that mimic the extracellular matrix in providing support; allowing the natural flow of oxygen, nutrients, and growth factors; and possibly favoring neural cell regrowth. Their combined use with stem cells has many potentialities for tissue engineering applications. Here, we describe a new 3D model of stem cell culture, using a nanostructured biomaterial, made of self-assembling peptides, where adult neural stem cells are completely embedded. This new 3D cell culture system takes advantage of the nano- and microfiber assembling process of these biomaterials under physiological conditions. The assembled scaffold forms an intricate and biologically active matrix able to display specifically designed functional motifs such as RGD, BMHP1, and BMHP2. Such model has the potential to be tailored to develop ad hoc designed peptides for specific cell lines.
Collapse
Affiliation(s)
- Carla Cunha
- NEWTherapies Group, INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | | | | |
Collapse
|
27
|
Wolun-Cholewa M, Langer K, Szymanowski K, Glodek A, Jankowska A, Warchol W, Langer J. An efficient 3D cell culture method on biomimetic nanostructured grids. PLoS One 2013; 8:e72936. [PMID: 24023793 PMCID: PMC3759432 DOI: 10.1371/journal.pone.0072936] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 07/15/2013] [Indexed: 11/18/2022] Open
Abstract
Current techniques of in vitro cell cultures are able to mimic the in vivo environment only to a limited extent, as they enable cells to grow only in two dimensions. Therefore cell culture approaches should rely on scaffolds that provide support comparable to the extracellular matrix. Here we demonstrate the advantages of novel nanostructured three-dimensional grids fabricated using electro-spinning technique, as scaffolds for cultures of neoplastic cells. The results of the study show that the fibers allow for a dynamic growth of HeLa cells, which form multi-layer structures of symmetrical and spherical character. This indicates that the applied scaffolds are nontoxic and allow proper flow of oxygen, nutrients, and growth factors. In addition, grids have been proven to be useful in in situ examination of cells ultrastructure.
Collapse
Affiliation(s)
- Maria Wolun-Cholewa
- Department of Cell Biology, Poznan University of Medical Science, Poznan, Poland
| | | | | | | | | | | | | |
Collapse
|
28
|
Liu Y, Ye H, Satkunendrarajah K, Yao GS, Bayon Y, Fehlings MG. A self-assembling peptide reduces glial scarring, attenuates post-traumatic inflammation and promotes neurological recovery following spinal cord injury. Acta Biomater 2013; 9:8075-88. [PMID: 23770224 DOI: 10.1016/j.actbio.2013.06.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 05/23/2013] [Accepted: 06/03/2013] [Indexed: 01/22/2023]
Abstract
The pathophysiology of spinal cord injury (SCI) involves post-traumatic inflammation and glial scarring which interfere with repair and recovery. Self-assembling peptides (SAPs) are molecules designed for tissue engineering. Here, we tested the performance of K2(QL)6K2 (QL6), a SAP that attenuates inflammation and glial scarring, and facilitates functional recovery. We injected QL6 into the spinal cord tissue of rats 24 h after clip compression SCI. QL6 led to a significant reduction in post-traumatic apoptosis, inflammation and astrogliosis. It also resulted in significant tissue preservation as determined by quantitative histomorphometry. Furthermore, QL6 promoted axonal preservation/regeneration, demonstrated by BDA anterograde and Fluorogold retrograde tracing. In vitro experiments found that a QL6 scaffold enhanced neuronal differentiation and suppressed astrocytic development. The electrophysiology confirmed that QL6 led to significant functional improvement of axons, including increased conduction velocity, reduced refractoriness and enhanced high-frequency conduction. These neuroanatomical and electrophysiological improvements were associated with significant neurobehavioral recovery as assessed by the Basso-Beattie-Bresnahan technique. As the first detailed examination of the pathophysiological properties of QL6 in SCI, this work reveals the therapeutic potential of SAPs, and may suggest an approach for the reconstruction of the injured spinal cord.
Collapse
Affiliation(s)
- Yang Liu
- Department of Genetics and Development, Toronto Western Research Institute and Spinal Program, Krembil Neuroscience Centre, University Health Network, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
29
|
Sangiambut S, Channon K, Thomson NM, Sato S, Tsuge T, Doi Y, Sivaniah E. A robust route to enzymatically functional, hierarchically self-assembled peptide frameworks. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2013; 25:2661-2665. [PMID: 23341342 DOI: 10.1002/adma.201204127] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 11/21/2012] [Indexed: 06/01/2023]
Abstract
The addition of enzyme biofunctionality to self-assembling peptide nanofibers is challenging since such additions can inhibit functionality or self-assembly. We introduce a method for peptide nanofiber enzyme functionalization, demonstrated by the attachment of a polymerization synthase to peptide nanofibers. The enzyme generates a biocompatible, biodegradable biopolyester coat on the fibers with applicablity in medical engineering. This approach provides a template for generation of functional bionanomaterials.
Collapse
Affiliation(s)
- S Sangiambut
- Biological and Soft Systems, Cavendish Laboratory, University of Cambridge, CB3 0HE, UK
| | | | | | | | | | | | | |
Collapse
|
30
|
Nanofiber scaffolds facilitate functional regeneration of peripheral nerve injury. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 9:305-15. [DOI: 10.1016/j.nano.2012.08.009] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 05/09/2012] [Accepted: 08/08/2012] [Indexed: 01/17/2023]
|
31
|
Caprini A, Silva D, Zanoni I, Cunha C, Volontè C, Vescovi A, Gelain F. A novel bioactive peptide: assessing its activity over murine neural stem cells and its potential for neural tissue engineering. N Biotechnol 2013; 30:552-62. [PMID: 23541699 DOI: 10.1016/j.nbt.2013.03.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Revised: 02/11/2013] [Accepted: 03/18/2013] [Indexed: 01/16/2023]
Abstract
The design of biomimetic scaffolds suitable for cell-based therapies is a fundamental step for the regeneration of the damaged nervous system; indeed growing interest is focusing on the discovery of peptide sequences to modulate the fate of transplanted cells and, in particular, the differentiation outcome of multipotent neural stem cells. By applying the Phage Display technique to murine neural stem cells we isolated a peptide, KLPGWSG, present in proteins involved in both stem cell maintenance and differentiation. We show that KLPGWSG binds molecules expressed on the cell surface of murine adult neural stem cells, thus may potentially be involved in stem cell fate determination. Indeed we demonstrated that this peptide in solution enhances per se cell differentiation toward the neuronal phenotype. Hence, we synthesized two LDLK-12-based self-assembling peptides functionalized with KLPGWSG peptide (KLP and Ac-KLP) and characterized them via atomic force microscopy, rheometry and circular dichroism, obtaining nanostructured hydrogels supporting murine neural stem cells differentiation in vitro. Interestingly, we demonstrated that, when scaffold stiffness is comparable to that of the brain in vivo, the Ac-KLP SAP-based scaffold enhances the neuronal differentiation of neural stem cells. These evidences place both KLPGWSG and the functionalized self-assembling peptide Ac-KLP as promising candidates for, respectively, biomimetic studies and stem cell therapies for nervous regeneration.
Collapse
Affiliation(s)
- Andrea Caprini
- Center for Nanomedicine and Tissue Engineering, A.O. Ospedale Niguarda Ca' Granda, Milan 20162, Italy
| | | | | | | | | | | | | |
Collapse
|
32
|
Liedmann A, Frech S, Morgan PJ, Rolfs A, Frech MJ. Differentiation of human neural progenitor cells in functionalized hydrogel matrices. Biores Open Access 2013; 1:16-24. [PMID: 23515105 PMCID: PMC3560381 DOI: 10.1089/biores.2012.0209] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Hydrogel-based three-dimensional (3D) scaffolds are widely used in the field of regenerative medicine, translational medicine, and tissue engineering. Recently, we reported the effect of scaffold formation on the differentiation and survival of human neural progenitor cells (hNPCs) using PuraMatrix™ (RADA-16) scaffolds. Here, we were interested in the impact of PuraMatrix modified by the addition of short peptide sequences, based on a bone marrow homing factor and laminin. The culture and differentiation of the hNPCs in the modified matrices resulted in an approximately fivefold increase in neuronal cells. The examination of apoptotic and necrotic cells, as well as the level of the anti-apoptotic protein Bcl-2, indicates benefits for cells hosted in the modified formulations. In addition, we found a trend to lower proportions of apoptotic or necrotic neuronal cells in the modified matrices. Interestingly, the neural progenitor cell pool was increased in all the tested matrices in comparison to the standard 2D culture system, while no difference was found between the modified matrices. We conclude that a combination of elevated neuronal differentiation and a protective effect of the modified matrices underlies the increased proportion of neuronal cells.
Collapse
Affiliation(s)
- Andrea Liedmann
- Albrecht-Kossel-Institute for Neuroregeneration, University of Rostock , Rostock, Germany
| | | | | | | | | |
Collapse
|
33
|
Huang H, Ding Y, Sun XS, Nguyen TA. Peptide hydrogelation and cell encapsulation for 3D culture of MCF-7 breast cancer cells. PLoS One 2013; 8:e59482. [PMID: 23527204 PMCID: PMC3603912 DOI: 10.1371/journal.pone.0059482] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 02/14/2013] [Indexed: 01/15/2023] Open
Abstract
Three-dimensional (3D) cell culture plays an invaluable role in tumor biology by providing in vivo like microenviroment and responses to therapeutic agents. Among many established 3D scaffolds, hydrogels demonstrate a distinct property as matrics for 3D cell culture. Most of the existing pre-gel solutions are limited under physiological conditions such as undesirable pH or temperature. Here, we report a peptide hydrogel that shows superior physiological properties as an in vitro matrix for 3D cell culture. The 3D matrix can be accomplished by mixing a self-assembling peptide directly with a cell culture medium without any pH or temperature adjustment. Results of dynamic rheological studies showed that this hydrogel can be delivered multiple times via pipetting without permanently destroying the hydrogel architecture, indicating the deformability and remodeling ability of the hydrogel. Human epithelial cancer cells, MCF-7, are encapsulated homogeneously in the hydrogel matrix during hydrogelation. Compared with two-dimensional (2D) monolayer culture, cells residing in the hydrogel matrix grow as tumor-like clusters in 3D formation. Relevant parameters related to cell morphology, survival, proliferation, and apoptosis were analyzed using MCF-7 cells in 3D hydrogels. Interestingly, treatment of cisplatin, an anti-cancer drug, can cause a significant decrease of cell viability of MCF-7 clusters in hydrogels. The responses to cisplatin were dose- and time-dependent, indicating the potential usage of hydrogels for drug testing. Results of confocal microscopy and Western blotting showed that cells isolated from hydrogels are suitable for downstream proteomic analysis. The results provided evidence that this peptide hydrogel is a promising 3D cell culture material for drug testing.
Collapse
Affiliation(s)
- Hongzhou Huang
- Department of Grain Science and Industry, Kansas State University, Manhattan, Kansas, United States of America
| | - Ying Ding
- Department of Biochemistry, Kansas State University, Manhattan, Kansas, United States of America
| | - Xiuzhi S. Sun
- Department of Grain Science and Industry, Kansas State University, Manhattan, Kansas, United States of America
- * E-mail: (XSS); (TAN)
| | - Thu A. Nguyen
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, Kansas, United States of America
- * E-mail: (XSS); (TAN)
| |
Collapse
|
34
|
Wang H, Yang Z. Short-peptide-based molecular hydrogels: novel gelation strategies and applications for tissue engineering and drug delivery. NANOSCALE 2012; 4:5259-67. [PMID: 22814874 DOI: 10.1039/c2nr31149f] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Molecular hydrogels hold big potential for tissue engineering and controlled drug delivery. Our lab focuses on short-peptide-based molecular hydrogels formed by biocompatible methods and their applications in tissue engineering (especially, 3D cell culture) and controlled drug delivery. This feature article firstly describes our recent progresses of the development of novel methods to form hydrogels, including the strategy of disulfide bond reduction and assistance with specific protein-peptide interactions. We then introduce the applications of our hydrogels in fields of controlled stem cell differentiation, cell culture, surface modifications of polyester materials by molecular self-assembly, and anti-degradation of recombinant complex proteins. A novel molecular hydrogel system of hydrophobic compounds that are only formed by hydrolysis processes was also included in this article. The hydrogels of hydrophobic compounds, especially those of hydrophobic therapeutic agents, may be developed into a carrier-free delivery system for long term delivery of therapeutic agents. With the efforts in this field, we believe that molecular hydrogels formed by short peptides and hydrophobic therapeutic agents can be practically applied for 3D cell culture and long term drug delivery in near future, respectively.
Collapse
Affiliation(s)
- Huaimin Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | | |
Collapse
|
35
|
Yang WS, Park YC, Kim JH, Kim HR, Yu T, Byeon SE, Unsworth LD, Lee J, Cho JY. Nanostructured, self-assembling peptide K5 blocks TNF-α and PGE₂ production by suppression of the AP-1/p38 pathway. Mediators Inflamm 2012; 2012:489810. [PMID: 22315508 PMCID: PMC3270444 DOI: 10.1155/2012/489810] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 11/17/2011] [Indexed: 02/06/2023] Open
Abstract
Nanostructured, self-assembling peptides hold promise for a variety of regenerative medical applications such as 3D cell culture systems, accelerated wound healing, and nerve repair. The aim of this study was to determine whether the self-assembling peptide K5 can be applied as a carrier of anti-inflammatory drugs. First, we examined whether the K5 self-assembling peptide itself can modulate various cellular inflammatory responses. We found that peptide K5 significantly suppressed the release of tumor-necrosis-factor- (TNF-) α and prostaglandin E₂ (PGE₂) from RAW264.7 cells and peritoneal macrophages stimulated by lipopolysaccharide (LPS). Similarly, there was inhibition of cyclooxygenase- (COX-) 2 mRNA expression assessed by real-time PCR, indicating that the inhibition is at the transcriptional level. In agreement with this finding, peptide K5 suppressed the translocation of the transcription factors activator protein (AP-1) and c-Jun and inhibited upstream inflammatory effectors including mitogen activated protein kinase (MAPK), p38, and mitogen-activated protein kinase kinase 3/6 (MKK 3/6). Whether this peptide exerts its effects via a transmembrane or cytoplasmic receptor is not clear. However, our data strongly suggest that the nanostructured, self-assembling peptide K5 may possess significant anti-inflammatory activity via suppression of the p38/AP-1 pathway.
Collapse
Affiliation(s)
- Woo Seok Yang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Yung Chul Park
- College of Forest & Environmental Sciences, Kangwon National University, Chuncheon 200-701, Republic of Korea
| | - Ji Hye Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Hye Ri Kim
- College of Forest & Environmental Sciences, Kangwon National University, Chuncheon 200-701, Republic of Korea
| | - Tao Yu
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Se Eun Byeon
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Larry D. Unsworth
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada T6G 2G6
| | - Jaehwi Lee
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Republic of Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| |
Collapse
|
36
|
Luo Z, Zhang S. Designer nanomaterials using chiral self-assembling peptide systems and their emerging benefit for society. Chem Soc Rev 2012; 41:4736-54. [DOI: 10.1039/c2cs15360b] [Citation(s) in RCA: 183] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
37
|
Sawada T, Mihara H. Dense surface functionalization using peptides that recognize differences in organized structures of self-assembling nanomaterials. MOLECULAR BIOSYSTEMS 2012; 8:1264-74. [DOI: 10.1039/c2mb05435c] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
38
|
|
39
|
Liu J, Zhao X. Design of self-assembling peptides and their biomedical applications. Nanomedicine (Lond) 2011; 6:1621-43. [PMID: 22077465 DOI: 10.2217/nnm.11.142] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Combining physics, engineering, chemistry and biology, we can now design, synthesize and fabricate biological nanomaterials at the molecular scale using self-assembling peptide systems. These peptides have been used for fabrication of nanomaterials, including nanofibers, nanotubes and vesicles, nanometer-thick surface coating and nanowires. Some of these peptides are used for stabilizing membrane proteins and drug delivery, and others provide a more permissive environment for 3D cell culture, tissue engineering and repair of tissues in regenerative medicine. Self-assembling peptides are also useful for fabricating a wide spectrum of exquisitely fine architectures, nanomaterials and nanodevices for nanomedicine and nanobiotechnology. These peptide systems lie at the interface between molecular biology, chemistry, materials science and engineering. The studies of designed self-assembling peptides and their applications will help us to understand nature’s enormous power and how to apply it to benefit other disciplines and society.
Collapse
Affiliation(s)
- Jingping Liu
- West China Hospital Laboratory of Nanomedicine & Institute for Nanobiomedical Technology & Membrane Biology, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Transplant Engineering & Immunology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaojun Zhao
- Center for Biomedical Engineering, NE47-379, Massachusetts Institute of Technology, Cambridge, MA 02139-4307, USA
| |
Collapse
|
40
|
Liu J, Zhang L, Yang Z, Zhao X. Controlled release of paclitaxel from a self-assembling peptide hydrogel formed in situ and antitumor study in vitro. Int J Nanomedicine 2011; 6:2143-53. [PMID: 22114478 PMCID: PMC3215155 DOI: 10.2147/ijn.s24038] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Background A nanoscale injectable in situ-forming hydrogel drug delivery system was developed in this study. The system was based on a self-assembling peptide RADA16 solution, which can spontaneously form a hydrogel rapidly under physiological conditions. We used the RADA16 hydrogel for the controlled release of paclitaxel (PTX), a hydrophobic antitumor drug. Methods The RADA16-PTX suspension was prepared simply by magnetic stirring, followed by atomic force microscopy, circular dichroism analysis, dynamic light scattering, rheological analysis, an in vitro release assay, and a cell viability test. Results The results indicated that RADA16 and PTX can interact with each other and that the amphiphilic peptide was able to stabilize hydrophobic drugs in aqueous solution. The particle size of PTX was markedly decreased in the RADA16 solution compared with its size in water. The RADA16-PTX suspension could form a hydrogel in culture medium, and the elasticity of the hydrogel showed a positive correlation with peptide concentration. In vitro release measurements indicated that hydrogels with a higher peptide concentration had a longer half-release time. The RADA16-PTX hydrogel could effectively inhibit the growth of the breast cancer cell line, MDA-MB-435S, in vitro, and hydrogels with higher peptide concentrations were more effective at inhibiting tumor cell proliferation. The RADA16-PTX hydrogel was effective at controlling the release of PTX and inhibiting tumor cell growth in vitro. Conclusion Self-assembling peptide hydrogels may work well as a system for drug delivery.
Collapse
Affiliation(s)
- Jingping Liu
- West China Hospital Laboratory of Nanomedicine and Institute for Nanobiomedical Technology and Membrane Biology, Sichuan University, Chengdu, China
| | | | | | | |
Collapse
|
41
|
Tarabout C, Roux S, Gobeaux F, Fay N, Pouget E, Meriadec C, Ligeti M, Thomas D, IJsselstijn M, Besselievre F, Buisson DA, Verbavatz JM, Petitjean M, Valéry C, Perrin L, Rousseau B, Artzner F, Paternostre M, Cintrat JC. Control of peptide nanotube diameter by chemical modifications of an aromatic residue involved in a single close contact. Proc Natl Acad Sci U S A 2011; 108:7679-84. [PMID: 21518895 PMCID: PMC3093526 DOI: 10.1073/pnas.1017343108] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Supramolecular self-assembly is an attractive pathway for bottom-up synthesis of novel nanomaterials. In particular, this approach allows the spontaneous formation of structures of well-defined shapes and monodisperse characteristic sizes. Because nanotechnology mainly relies on size-dependent physical phenomena, the control of monodispersity is required, but the possibility of tuning the size is also essential. For self-assembling systems, shape, size, and monodispersity are mainly settled by the chemical structure of the building block. Attempts to change the size notably by chemical modification usually end up with the loss of self-assembly. Here, we generated a library of 17 peptides forming nanotubes of monodisperse diameter ranging from 10 to 36 nm. A structural model taking into account close contacts explains how a modification of a few Å of a single aromatic residue induces a fourfold increase in nanotube diameter. The application of such a strategy is demonstrated by the formation of silica nanotubes of various diameters.
Collapse
Affiliation(s)
- Christophe Tarabout
- Unité Mixte de Recherche 6251, Centre National de la Recherche Scientifique and Université Rennes 1, F-35 Rennes, France
| | - Stéphane Roux
- Unité Mixte de Recherche 6251, Centre National de la Recherche Scientifique and Université Rennes 1, F-35 Rennes, France
- Institut de Biologie et de Technologies de Saclay/Service de Chimie Bioorganique et Marquage, Commissariat à l’Énergie Atomique et aux Energies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
| | - Frédéric Gobeaux
- Institut de Biologie et de Technologies de Saclay/Service de Bioénergétique, Biologie Structurale et Mécanismes, Commissariat à l’Énergie Atomique et aux Énergies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
- Unité de Recherche Associée 2096, Centre Nationale de la Recherche Scientifique, Commissariat à l’Énergie Atomique et aux Energies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
| | - Nicolas Fay
- Institut de Biologie et de Technologies de Saclay/Service de Bioénergétique, Biologie Structurale et Mécanismes, Commissariat à l’Énergie Atomique et aux Énergies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
- Unité de Recherche Associée 2096, Centre Nationale de la Recherche Scientifique, Commissariat à l’Énergie Atomique et aux Energies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
| | - Emilie Pouget
- Unité Mixte de Recherche 6251, Centre National de la Recherche Scientifique and Université Rennes 1, F-35 Rennes, France
| | - Cristelle Meriadec
- Unité Mixte de Recherche 6251, Centre National de la Recherche Scientifique and Université Rennes 1, F-35 Rennes, France
| | - Melinda Ligeti
- Institut de Biologie et de Technologies de Saclay/Service de Chimie Bioorganique et Marquage, Commissariat à l’Énergie Atomique et aux Energies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
| | - Daniel Thomas
- Unité Mixte de Recherche 6026, Centre National de la Recherche Scientifique and Université Rennes 1, F-35042 Rennes Cedex, France; and
| | - Maarten IJsselstijn
- Institut de Biologie et de Technologies de Saclay/Service de Chimie Bioorganique et Marquage, Commissariat à l’Énergie Atomique et aux Energies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
| | - François Besselievre
- Institut de Biologie et de Technologies de Saclay/Service de Chimie Bioorganique et Marquage, Commissariat à l’Énergie Atomique et aux Energies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
| | - David-Alexandre Buisson
- Institut de Biologie et de Technologies de Saclay/Service de Chimie Bioorganique et Marquage, Commissariat à l’Énergie Atomique et aux Energies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
| | - Jean-Marc Verbavatz
- Institut de Biologie et de Technologies de Saclay/Service de Bioénergétique, Biologie Structurale et Mécanismes, Commissariat à l’Énergie Atomique et aux Énergies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
- Unité de Recherche Associée 2096, Centre Nationale de la Recherche Scientifique, Commissariat à l’Énergie Atomique et aux Energies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
| | - Michel Petitjean
- Institut de Biologie et de Technologies de Saclay/Service de Bioénergétique, Biologie Structurale et Mécanismes, Commissariat à l’Énergie Atomique et aux Énergies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
- Unité de Recherche Associée 2096, Centre Nationale de la Recherche Scientifique, Commissariat à l’Énergie Atomique et aux Energies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
| | - Céline Valéry
- Ipsen-Pharma, san Feliu de Llobregat, Barcelona, Spain
| | - Lionel Perrin
- Institut de Biologie et de Technologies de Saclay/Service de Bioénergétique, Biologie Structurale et Mécanismes, Commissariat à l’Énergie Atomique et aux Énergies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
- Unité de Recherche Associée 2096, Centre Nationale de la Recherche Scientifique, Commissariat à l’Énergie Atomique et aux Energies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
| | - Bernard Rousseau
- Institut de Biologie et de Technologies de Saclay/Service de Chimie Bioorganique et Marquage, Commissariat à l’Énergie Atomique et aux Energies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
| | - Franck Artzner
- Unité Mixte de Recherche 6251, Centre National de la Recherche Scientifique and Université Rennes 1, F-35 Rennes, France
| | - Maité Paternostre
- Institut de Biologie et de Technologies de Saclay/Service de Bioénergétique, Biologie Structurale et Mécanismes, Commissariat à l’Énergie Atomique et aux Énergies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
- Unité de Recherche Associée 2096, Centre Nationale de la Recherche Scientifique, Commissariat à l’Énergie Atomique et aux Energies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
| | - Jean-Christophe Cintrat
- Institut de Biologie et de Technologies de Saclay/Service de Chimie Bioorganique et Marquage, Commissariat à l’Énergie Atomique et aux Energies Alternatives-Saclay, F-91191 Gif-sur-Yvette, France
| |
Collapse
|
42
|
Cunha C, Panseri S, Villa O, Silva D, Gelain F. 3D culture of adult mouse neural stem cells within functionalized self-assembling peptide scaffolds. Int J Nanomedicine 2011; 6:943-55. [PMID: 21720506 PMCID: PMC3124398 DOI: 10.2147/ijn.s17292] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Indexed: 12/20/2022] Open
Abstract
Three-dimensional (3D) in vitro models of cell culture aim to fill the gap between the standard two-dimensional cell studies and the in vivo environment. Especially for neural tissue regeneration approaches where there is little regenerative capacity, these models are important for mimicking the extracellular matrix in providing support, allowing the natural flow of oxygen, nutrients, and growth factors, and possibly favoring neural cell regrowth. We have previously demonstrated that a new self-assembling nanostructured biomaterial, based on matrigel, was able to support adult neural stem cell (NSC) culture. In this study, we developed a new 3D cell culture system that takes advantage of the nano- and microfiber assembling process, under physiologic conditions, of these biomaterials. The assembled scaffold forms an intricate and biologically active matrix that displays specifically designed functional motifs: RGD (Arg-Gly-Asp), BMHP1 (bone marrow homing peptide 1), and BMHP2, for the culture of adult NSCs. These scaffolds were prepared at different concentrations, and microscopic examination of the cell-embedded scaffolds showed that NSCs are viable and they proliferate and differentiate within the nanostructured environment of the scaffold. Such a model has the potential to be tailored to develop ad hoc designed peptides for specific cell lines.
Collapse
Affiliation(s)
- Carla Cunha
- Department of Biotechnology and Biosciences, University of Milano-Bicocca
- Center for Nanomedicine and Tissue Engineering, CNTE – A.O. Ospedale Niguarda Ca’ Granda, Milan
| | - Silvia Panseri
- Laboratory of Biomechanics and Technology Innovation, Rizzoli Orthopaedic Institute, Bologna
- Laboratory of Nano-Biomagnetism, Institute of Science and Technology for Ceramics, National Research Council, Faenza, Italy
| | - Omar Villa
- Department of Biotechnology and Biosciences, University of Milano-Bicocca
- Center for Nanomedicine and Tissue Engineering, CNTE – A.O. Ospedale Niguarda Ca’ Granda, Milan
| | - Diego Silva
- Department of Biotechnology and Biosciences, University of Milano-Bicocca
- Center for Nanomedicine and Tissue Engineering, CNTE – A.O. Ospedale Niguarda Ca’ Granda, Milan
| | - Fabrizio Gelain
- Department of Biotechnology and Biosciences, University of Milano-Bicocca
- Center for Nanomedicine and Tissue Engineering, CNTE – A.O. Ospedale Niguarda Ca’ Granda, Milan
| |
Collapse
|
43
|
Luo Z, Wang S, Zhang S. Fabrication of self-assembling d-form peptide nanofiber scaffold d-EAK16 for rapid hemostasis. Biomaterials 2011; 32:2013-20. [DOI: 10.1016/j.biomaterials.2010.11.049] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Accepted: 11/19/2010] [Indexed: 10/18/2022]
|
44
|
Amadi OC, Steinhauser ML, Nishi Y, Chung S, Kamm RD, McMahon AP, Lee RT. A low resistance microfluidic system for the creation of stable concentration gradients in a defined 3D microenvironment. Biomed Microdevices 2011; 12:1027-41. [PMID: 20661647 DOI: 10.1007/s10544-010-9457-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The advent of microfluidic technology allows control and interrogation of cell behavior by defining the local microenvironment with an assortment of biochemical and biophysical stimuli. Many approaches have been developed to create gradients of soluble factors, but the complexity of such systems or their inability to create defined and controllable chemical gradients has limited their widespread implementation. Here we describe a new microfluidic device which employs a parallel arrangement of wells and channels to create stable, linear concentration gradients in a gel region between a source and a sink well. Pressure gradients between the source and sink wells are dissipated through low resistance channels in parallel with the gel channel, thus minimizing the convection of solute in this region. We demonstrate the ability of the new device to quantitate chemotactic responses in a variety of cell types, yielding a complete profile of the migratory response and representing the total number of migrating cells and the distance each cell has migrated. Additionally we show the effect of concentration gradients of the morphogen Sonic hedgehog on the specification of differentiating neural progenitors in a 3-dimensional matrix.
Collapse
Affiliation(s)
- Ovid C Amadi
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Yang Z, Zhao X. A 3D model of ovarian cancer cell lines on peptide nanofiber scaffold to explore the cell-scaffold interaction and chemotherapeutic resistance of anticancer drugs. Int J Nanomedicine 2011; 6:303-10. [PMID: 21383855 PMCID: PMC3044183 DOI: 10.2147/ijn.s15279] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Indexed: 02/05/2023] Open
Abstract
RADA16-I peptide hydrogel, a type of nanofiber scaffold derived from self-assembling peptide RADA16-I, has been extensively applied to regenerative medicine and tissue repair in order to develop novel nanomedicine systems. In this study, using RADA16-I peptide hydrogel, a three-dimensional (3D) cell culture model was fabricated for in vitro culture of three ovarian cancer cell lines. Firstly, the peptide nanofiber scaffold was evaluated by transmission electron microscopy and atom force microscopy. Using phase contrast microscopy, the appearance of the representative ovarian cancer cells encapsulated in RADA16-I peptide hydrogel on days 1, 3, and 7 in 24-well Petri dishes was illustrated. The cancer cell-nanofiber scaffold construct was cultured for 5 days, and the ovarian cancer cells had actively proliferative potential. The precultured ovarian cancer cells exhibited nearly similar adhesion properties and invasion potentials in vitro between RADA16-I peptide nanofiber and type I collagen, which suggested that RADA16-I peptide hydrogel had some similar characteristics to type I collagen. The precultured ovarian cancer cells had two-fold to five-fold higher anticancer drug resistance than the conventional two-dimensional Petri dish culture. So the 3D cell model on peptide nanofiber scaffold is an optimal type of cell pattern for anticancer drug screening and tumor biology.
Collapse
Affiliation(s)
- Zehong Yang
- Nanomedicine Laboratory, West China Hospital and Institute for Nanobiomedical Technology and Membrane Biology, Sichuan University, Chengdu, People’s Republic of China
| | - Xiaojun Zhao
- Nanomedicine Laboratory, West China Hospital and Institute for Nanobiomedical Technology and Membrane Biology, Sichuan University, Chengdu, People’s Republic of China
- Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
46
|
Abstract
Cell culture in two dimensions has been routinely and diligently undertaken in thousands of laboratories worldwide for the past four decades. However, the culture of cells in two dimensions is arguably primitive and does not reproduce the anatomy or physiology of a tissue for informative or useful study. Creating a third dimension for cell culture is clearly more relevant, but requires a multidisciplinary approach and multidisciplinary expertise. When entering the third dimension, investigators need to consider the design of scaffolds for supporting the organisation of cells or the use of bioreactors for controlling nutrient and waste product exchange. As 3D culture systems become more mature and relevant to human and animal physiology, the ability to design and develop co-cultures becomes possible as does the ability to integrate stem cells. The primary objectives for developing 3D cell culture systems vary widely - and range from engineering tissues for clinical delivery through to the development of models for drug screening. The intention of this review is to provide a general overview of the common approaches and techniques for designing 3D culture models.
Collapse
Affiliation(s)
- John W Haycock
- Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield, UK.
| |
Collapse
|
47
|
Soto-Gutierrez A, Yagi H, Uygun BE, Navarro-Alvarez N, Uygun K, Kobayashi N, Yang YG, Yarmush ML. Cell delivery: from cell transplantation to organ engineering. Cell Transplant 2010; 19:655-65. [PMID: 20525441 DOI: 10.3727/096368910x508753] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cell populations derived from adult tissue and stem cells possess a great expectation for the treatment of several diseases. Great efforts have been made to generate cells with therapeutic impact from stem cells. However, it is clear that the development of systems to deliver such cells to induce efficient engraftment, growth, and function is a real necessity. Biologic and artificial scaffolds have received significant attention for their potential therapeutic application when use to form tissues in vitro and facilitate engraftment in vivo. Ultimately more sophisticated methods for decellularization of organs have been successfully used in tissue engineering and regenerative medicine applications. These decellularized tissues and organs appear to provide bioactive molecules and bioinductive properties to induce homing, differentiation, and proliferation of cells. The combination of decellularized organs and stem cells may dramatically improve the survival, engraftment, and fate control of transplanted stem cells and their ultimate clinical utility, opening the doors to a new era of organ engineering.
Collapse
Affiliation(s)
- Alejandro Soto-Gutierrez
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and the Shriners Hospitals for Children, Center for Engineering in Medicine, Boston, MA 76104, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Koopmans RJ, Aggeli A. Nanobiotechnology—quo vadis? Curr Opin Microbiol 2010; 13:327-34. [DOI: 10.1016/j.mib.2010.01.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Accepted: 01/18/2010] [Indexed: 01/12/2023]
|
49
|
Abstract
To deepen understanding and hasten the development of treatments, cancer needs to be modelled more accurately in vitro; applying tissue-engineering concepts and approaches in this field could bridge the gap between two-dimensional studies and in vivo animal models.
Collapse
Affiliation(s)
- Dietmar W Hutmacher
- Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove Queensland 4059, Australia.
| |
Collapse
|
50
|
Baker BM, Handorf AM, Ionescu LC, Li WJ, Mauck RL. New directions in nanofibrous scaffolds for soft tissue engineering and regeneration. Expert Rev Med Devices 2009; 6:515-32. [PMID: 19751124 DOI: 10.1586/erd.09.39] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
This review focuses on the role of nanostructure and nanoscale materials for tissue engineering applications. We detail a scaffold production method (electrospinning) for the production of nanofiber-based scaffolds that can approximate many critical features of the normal cellular microenvironment, and so foster and direct tissue formation. Further, we describe new and emerging methods to increase the applicability of these scaffolds for in vitro and in vivo application. This discussion includes a focus on methods to further functionalize scaffolds to promote cell infiltration, methods to tune scaffold mechanics to meet in vivo demands and methods to control the release of pharmaceuticals and other biologic agents to modulate the wound environment and foster tissue regeneration. This review provides a perspective on the state-of-the-art production, application and functionalization of these unique nanofibrous structures, and outlines future directions in this growing field.
Collapse
Affiliation(s)
- Brendon M Baker
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|