1
|
Zhao B, Zhang Q, Yang H, Yu S, Fu R, Shi S, Wang Y, Zhou W, Cui Y, Guo Q, Zhang X. Peptide KN-17-Loaded Supramolecular Hydrogel Induces the Regeneration of the Pulp-Dentin Complex. ACS Biomater Sci Eng 2024; 10:2523-2533. [PMID: 38445444 DOI: 10.1021/acsbiomaterials.3c01376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Regenerating the pulp-dentin complex remains a decisive factor during apexification for immature permanent teeth. Peptide KN-17, which was modified based on the structure of cecropin B, could effectively interfere with bacterial growth and induce the migration of human bone marrow stromal cells (hBMSCs). This study aimed to investigate the effect of KN-17 on the tissue regeneration. To our surprise, KN-17 can significantly stimulate angiogenesis in vitro and in vivo, which may provide a guarantee for apical closure. Herein, a novel peptide/KN-17 coassembled hydrogel is developed via a heating-cooling process. Npx-FFEY/KN-17 supramolecular hydrogel can induce vessel development, stimulate odontogenic differentiation of human dental pulp stem cells (hDPSCs), and exert an antibacterial effect on Enterococcus faecalis (E. faecalis). Furthermore, coronal pulp excised rat molars are supplied with KN-17 or KN-17-loaded hydrogel and transplanted subcutaneously in BALB/c-nu mice. After 4 weeks, the hydrogel Npx-FFEY/KN-17 stimulates the formation of multiple odontoblast-like cells and dentin-like structures. Our findings demonstrate that the KN-17-loaded hydrogel can promote the regeneration of the pulp-dentin complex for continued root development.
Collapse
Affiliation(s)
- Borui Zhao
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Qian Zhang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Houzhi Yang
- Tianjin Medical University, Tianjin 300070, China
| | - Shuipeng Yu
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Rui Fu
- Tianjin Medical University, Tianjin 300070, China
| | - Shurui Shi
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Yuanyuan Wang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| | - Wei Zhou
- Immunology, Microenvironment & Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Yange Cui
- Program in Gene Expression and Regulation, The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Qingxiang Guo
- College of Chemical Engineering, Inner Mongolia University of Technology, Hohhot 010051, China
| | - Xi Zhang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
2
|
Guo X, Li J, Wu Y, Xu L. Recent advancements in hydrogels as novel tissue engineering scaffolds for dental pulp regeneration. Int J Biol Macromol 2024; 264:130708. [PMID: 38460622 DOI: 10.1016/j.ijbiomac.2024.130708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/22/2024] [Accepted: 03/05/2024] [Indexed: 03/11/2024]
Abstract
Although conventional root canal treatment offers an effective therapeutic solution, it negatively affects the viability of the affected tooth. In recent years, pulp regeneration technology has emerged as a novel method for treating irreversible pulpitis due to its ability to maintain tooth vitality. The successful implementation of this technique depends on scaffolds and transplantation of exogenous stem cells or recruitment of endogenous stem cells. Accordingly, the three-dimensional structure and viscoelastic characteristics of hydrogel scaffolds, which parallel those of the extracellular matrix, have generated considerable interest. Furthermore, hydrogels support the controlled release of regenerative drugs and to load a wide variety of bioactive molecules. By integrating antibacterial agents into the hydrogel matrix and stimulating an immune response, root canal disinfection can be significantly improved and the rate of pulp regeneration can be accelerated. This review aims to provide an overview of the clinical applications of hydrogels that have been reported in the last 5 years, and offer a comprehensive summary of the different approaches that have been utilized for the optimization of hydrogel scaffolds for pulp regeneration. Advancements and challenges in pulp regeneration using hydrogels treating aged teeth are discussed.
Collapse
Affiliation(s)
- Xiaofei Guo
- Xiangya Shool of Stomatology, Central South University, Changsha, Hunan, China
| | - Jiaxuan Li
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410083, China
| | - Yong Wu
- Department of Nephrology, The Second Xiangya Hospital, Key Laboratory of Kidney Disease and Blood Purification, Central South University, Changsha, Hunan, China
| | - Laijun Xu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China; School of Stomatology, Changsha Medical University, Changsha, Hunan 410219, China.
| |
Collapse
|
3
|
Roy A, Hao L, Francisco J, Guan J, Mareedu S, Zhai P, Dodd-O J, Heffernan C, Del Re D, Lee EJA, Kumar VA. Injectable Peptide Hydrogels Loaded with Murine Embryonic Stem Cells Relieve Ischemia In Vivo after Myocardial Infarction. Biomacromolecules 2024; 25:1319-1329. [PMID: 38291600 DOI: 10.1021/acs.biomac.3c01345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Myocardial infarction (MI) is a major cause of morbidity and mortality worldwide, especially in aging and metabolically unhealthy populations. A major target of regenerative tissue engineering is the restoration of viable cardiomyocytes to preserve cardiac function and circumvent the progression to heart failure post-MI. Amelioration of ischemia is a crucial component of such restorative strategies. Angiogenic β-sheet peptides can self-assemble into thixotropic nanofibrous hydrogels. These syringe aspiratable cytocompatible gels were loaded with stem cells and showed excellent cytocompatibility and minimal impact on the storage and loss moduli of hydrogels. Gels with and without cells were delivered into the myocardium of a mouse MI model (LAD ligation). Cardiac function and tissue remodeling were evaluated up to 4 weeks in vivo. Injectable peptide hydrogels synergized with loaded murine embryonic stem cells to demonstrate enhanced survival after intracardiac delivery during the acute phase post-MI, especially at 7 days. This approach shows promise for post-MI treatment and potentially functional cardiac tissue regeneration and warrants large-scale animal testing prior to clinical translation.
Collapse
Affiliation(s)
- Abhishek Roy
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Lei Hao
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Jamie Francisco
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Newark, New Jersey 07103, United States
| | - Jin Guan
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Newark, New Jersey 07103, United States
| | - Satvik Mareedu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Newark, New Jersey 07103, United States
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Newark, New Jersey 07103, United States
| | - Joseph Dodd-O
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Corey Heffernan
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Dominic Del Re
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Newark, New Jersey 07103, United States
| | - Eun Jung A Lee
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Vivek A Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
- Department of Endodontics, Rutgers School of Dental Medicine, Newark, New Jersey 07103, United States
| |
Collapse
|
4
|
Dodd-O J, Roy A, Siddiqui Z, Jafari R, Coppola F, Ramasamy S, Kolloli A, Kumar D, Kaundal S, Zhao B, Kumar R, Robang AS, Li J, Azizogli AR, Pai V, Acevedo-Jake A, Heffernan C, Lucas A, McShan AC, Paravastu AK, Prasad BVV, Subbian S, Král P, Kumar V. Antiviral fibrils of self-assembled peptides with tunable compositions. Nat Commun 2024; 15:1142. [PMID: 38326301 PMCID: PMC10850501 DOI: 10.1038/s41467-024-45193-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 01/17/2024] [Indexed: 02/09/2024] Open
Abstract
The lasting threat of viral pandemics necessitates the development of tailorable first-response antivirals with specific but adaptive architectures for treatment of novel viral infections. Here, such an antiviral platform has been developed based on a mixture of hetero-peptides self-assembled into functionalized β-sheets capable of specific multivalent binding to viral protein complexes. One domain of each hetero-peptide is designed to specifically bind to certain viral proteins, while another domain self-assembles into fibrils with epitope binding characteristics determined by the types of peptides and their molar fractions. The self-assembled fibrils maintain enhanced binding to viral protein complexes and retain high resilience to viral mutations. This method is experimentally and computationally tested using short peptides that specifically bind to Spike proteins of SARS-CoV-2. This platform is efficacious, inexpensive, and stable with excellent tolerability.
Collapse
Affiliation(s)
- Joseph Dodd-O
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Abhishek Roy
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Zain Siddiqui
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Roya Jafari
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Francesco Coppola
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Santhamani Ramasamy
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Afsal Kolloli
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Dilip Kumar
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Soni Kaundal
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Boyang Zhao
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ranjeet Kumar
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Alicia S Robang
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Jeffrey Li
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Abdul-Rahman Azizogli
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Varun Pai
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Amanda Acevedo-Jake
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Corey Heffernan
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
- SAPHTx Inc, Newark, NJ, 07104, USA
| | - Alexandra Lucas
- Center for Personalized Diagnostics and Center for Immunotherapy Vaccines and Virotherapy, Biodesign Institute, Arizona State University, 727 E, Tempe, AZ, USA
| | - Andrew C McShan
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Anant K Paravastu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - B V Venkataram Prasad
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Petr Král
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, 60607, USA.
- Department of Physics, University of Illinois at Chicago, Chicago, IL, 60607, USA.
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60607, USA.
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA.
| | - Vivek Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
- SAPHTx Inc, Newark, NJ, 07104, USA.
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
- Department of Endodontics, Rutgers School of Dental Medicine, Newark, NJ, 07103, USA.
| |
Collapse
|
5
|
Mahmoudi N, Mohamed E, Dehnavi SS, Aguilar LMC, Harvey AR, Parish CL, Williams RJ, Nisbet DR. Calming the Nerves via the Immune Instructive Physiochemical Properties of Self-Assembling Peptide Hydrogels. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303707. [PMID: 38030559 PMCID: PMC10837390 DOI: 10.1002/advs.202303707] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 10/22/2023] [Indexed: 12/01/2023]
Abstract
Current therapies for the devastating damage caused by traumatic brain injuries (TBI) are limited. This is in part due to poor drug efficacy to modulate neuroinflammation, angiogenesis and/or promoting neuroprotection and is the combined result of challenges in getting drugs across the blood brain barrier, in a targeted approach. The negative impact of the injured extracellular matrix (ECM) has been identified as a factor in restricting post-injury plasticity of residual neurons and is shown to reduce the functional integration of grafted cells. Therefore, new strategies are needed to manipulate the extracellular environment at the subacute phase to enhance brain regeneration. In this review, potential strategies are to be discussed for the treatment of TBI by using self-assembling peptide (SAP) hydrogels, fabricated via the rational design of supramolecular peptide scaffolds, as an artificial ECM which under the appropriate conditions yields a supramolecular hydrogel. Sequence selection of the peptides allows the tuning of these hydrogels' physical and biochemical properties such as charge, hydrophobicity, cell adhesiveness, stiffness, factor presentation, degradation profile and responsiveness to (external) stimuli. This review aims to facilitate the development of more intelligent biomaterials in the future to satisfy the parameters, requirements, and opportunities for the effective treatment of TBI.
Collapse
Affiliation(s)
- Negar Mahmoudi
- Laboratory of Advanced Biomaterialsthe John Curtin School of Medical ResearchAustralian National UniversityCanberraACT2601Australia
- ANU College of Engineering & Computer ScienceAustralian National UniversityCanberraACT2601Australia
- The Graeme Clark InstituteThe University of MelbourneMelbourneVIC3010Australia
- Department of Biomedical EngineeringFaculty of Engineering and Information TechnologyThe University of MelbourneMelbourneVIC3010Australia
| | - Elmira Mohamed
- Laboratory of Advanced Biomaterialsthe John Curtin School of Medical ResearchAustralian National UniversityCanberraACT2601Australia
| | - Shiva Soltani Dehnavi
- Laboratory of Advanced Biomaterialsthe John Curtin School of Medical ResearchAustralian National UniversityCanberraACT2601Australia
- ANU College of Engineering & Computer ScienceAustralian National UniversityCanberraACT2601Australia
| | - Lilith M. Caballero Aguilar
- Laboratory of Advanced Biomaterialsthe John Curtin School of Medical ResearchAustralian National UniversityCanberraACT2601Australia
- The Graeme Clark InstituteThe University of MelbourneMelbourneVIC3010Australia
- Department of Biomedical EngineeringFaculty of Engineering and Information TechnologyThe University of MelbourneMelbourneVIC3010Australia
| | - Alan R. Harvey
- School of Human SciencesThe University of Western Australiaand Perron Institute for Neurological and Translational SciencePerthWA6009Australia
| | - Clare L. Parish
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleMelbourneVIC3010Australia
| | | | - David R. Nisbet
- Laboratory of Advanced Biomaterialsthe John Curtin School of Medical ResearchAustralian National UniversityCanberraACT2601Australia
- The Graeme Clark InstituteThe University of MelbourneMelbourneVIC3010Australia
- Department of Biomedical EngineeringFaculty of Engineering and Information TechnologyThe University of MelbourneMelbourneVIC3010Australia
- Melbourne Medical SchoolFaculty of MedicineDentistry and Health ScienceThe University of MelbourneMelbourneVIC3010Australia
| |
Collapse
|
6
|
Roy A, Dodd-O JB, Robang AS, He D, West O, Siddiqui Z, Aguas ED, Goldberg H, Griffith A, Heffernan C, Hu Y, Paravastu AK, Kumar VA. Self-Assembling Peptides with Insulin-Like Growth Factor Mimicry. ACS APPLIED MATERIALS & INTERFACES 2024; 16:364-375. [PMID: 38145951 DOI: 10.1021/acsami.3c15660] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Growth factor (GF) mimicry involves recapitulating the signaling of larger molecules or cells. Although GF mimicry holds considerable promise in tissue engineering and drug design applications, difficulties in targeting the signaling molecule to the site of delivery and dissociation of mimicking peptides from their target receptors continue to limit its clinical application. To address these challenges, we utilized a self-assembling peptide (SAP) platform to generate synthetic insulin-like growth factor (IGF)-signaling, self-assembling GFs. Our peptide hydrogels are biocompatible and bind target IGF receptors in a dose-dependent fashion, activate proangiogenic signaling, and facilitate formation of angiogenic microtubules in vitro. Furthermore, infiltrated hydrogels are stable for weeks to months. We conclude that the enhanced targeting and long-term stability of our SAP/GF mimicry implants may improve the efficacy and safety of future GF mimic therapeutics.
Collapse
Affiliation(s)
- Abhishek Roy
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Joseph B Dodd-O
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Alicia S Robang
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Dongjing He
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Owen West
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Zain Siddiqui
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Erika Davidoff Aguas
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08544, United States
| | - Hannah Goldberg
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Alexandra Griffith
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Corey Heffernan
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Yuhang Hu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Anant K Paravastu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Vivek A Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
- Department of Biological Sciences, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
- Department of Endodontics, Rutgers School of Dental Medicine, Newark, New Jersey 07103, United States
| |
Collapse
|
7
|
Picotti S, Forte L, Serrentino J. A pre-market interventional, single-arm clinical investigation of a new topical lotion based on hyaluronic acid and peptides, EGYFIL TM, for the treatment of pain and stiffness in soft tissues. BMC Musculoskelet Disord 2023; 24:777. [PMID: 37784053 PMCID: PMC10544473 DOI: 10.1186/s12891-023-06903-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 09/21/2023] [Indexed: 10/04/2023] Open
Abstract
BACKGROUND Muscle pain and stiffness are strictly interconnected. Injuries frequently occur during sport activities, causing muscle pain, with or without stiffness, and require effective as well as fast-acting treatments. Topical products can be ideal for the treatment of such physical alterations as they are convenient and simple to use. In this study, it was investigated the application of a novel topical formulation, EGYFIL™, for the treatment of pain and stiffness due to muscle contracture, trauma, and/or overtraining. The lotion is composed of hyaluronic acid, a well-known ingredient for the pain alleviation, mixed with skin conditioning SH-Polypeptide-6 and SH-Oligopeptide-1, embedded in it. METHODS Twenty-six patients with pain and/or stiffness were enrolled. After a screening visit (Time 0, t0), patients were treated for the first time with the IP. The treatment consisted of topical application of the pain lotion. Level of pain and stiffness were measured with Numerical Rating Scale (NRS). Patients' pain and/or stiffness were evaluated at t0 (prior to using the product), after three hours (t1), and after three days (t2) of treatment. Participants were free to apply and re-apply the product ad libitum over the course of the study period (3 days). Potential adverse events (AE) and tolerance were evaluated during each visit. RESULTS There was a 22% decrease in pain in the first three hours (p < 0.001), followed by an additional 20% decrease after three days (p=0.0873). Overall, there was a 42% decrease in pain over the three days of the study (p =0.001). Furthermore, a 24% reduction in stiffness in the first three hours (p=0.025) and a 38% decrease in stiffness over three days (p < 0.001) were observed. Reduction in pain and stiffness were neither age, nor sex dependent. No adverse effects were reported during the study. CONCLUSION EGYFIL™ is safe and seems to reduce pain and stiffness in patients during the 3 days of treatment, already after 3 h from the first application. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT05711953. This trial was registered on 03/02/2023.
Collapse
Affiliation(s)
| | - Luca Forte
- Contrad Swiss SA, Via Ferruccio Pelli 2, Lugano, 6900, Switzerland.
| | - Jo Serrentino
- International Institute of Clinical Ecology (IICE), Quebec, Canada
| |
Collapse
|
8
|
Colombini A, Doro G, Ragni E, Forte L, de Girolamo L, Zerbinati F. Treatment with CR500® improves algofunctional scores in patients with knee osteoarthritis: a post-market confirmatory interventional, single arm clinical investigation. BMC Musculoskelet Disord 2023; 24:647. [PMID: 37573322 PMCID: PMC10422714 DOI: 10.1186/s12891-023-06754-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 07/25/2023] [Indexed: 08/14/2023] Open
Abstract
BACKGROUND Knee osteoarthritis (OA) is a progressive and degenerative condition. Several pharmacological and non-pharmacological treatments are able to improve the OA symptoms and the structural characteristics of the affected joints. Among these, infiltrative therapy with hyaluronic acid (HA) is the most used and consolidated procedure for the pain management. The addition of skin conditioning peptides to HA promotes the cartilage remodeling processes and a better permeation of the HA-based gel containing a peptide mixture, CR500®. Furthermore, the topic route of administration is convenient over the routinely used intra-articular injective procedures. In this study, the effectiveness of CR500® was evaluated in terms of improvement of the algo-functional symptoms related to unilateral knee OA. METHODS 38 mild and moderate OA patients were enrolled at a screening visit (V-1), treated at baseline visit (V1), and then continued the topical application of CR500® twice a week for 4 weeks, and followed-up for 3 visits (V2-V4) from week 2 to 4. Lequesne Knee Index (LKI) and Knee injury and Osteoarthritis Outcome Score (KOOS) were collected. Synovial fluid was collected and used for the quantification of neoepitope of type II collagen (C2C), C-terminal telopeptide of type II collagen (CTX-II), type II collagen propeptide (CPII), tumor necrosis factor alpha (TNFα) and HA. The expression of CD11c and CD206 was evaluated on cell pellets. RESULTS Three patients were excluded, thus 35 patients were included in the analysis. The treatment with CR500® was safe and well tolerated, with 7.9% patients had mild adverse events, not related to the device. The LKI total score showed a significant decrease from V1 to V4. KOOS score also showed a significant improvement of patient condition at V2, V3 and V4 in comparison with V1 for all subscales, except for KOOS sport subscale which improved only from V3. At V1 a negative correlation among KOOS pain subscale values and C2C, CPII and TNFα levels was observed, as well as a positive correlation between KOOS pain subscale and CD11c/CD206 ratio. CONCLUSION CR500® is safe and appear to be effective in improving pain and function in OA patients during the 4 weeks of treatment. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT05661162. This trial was registered on 22/12/2022.
Collapse
Affiliation(s)
- Alessandra Colombini
- Laboratorio di Biotecnologie Applicate all'Ortopedia, IRCCS Istituto Ortopedico Galeazzi, Via R. Galeazzi 4, Milan, 20161, Italy
| | - Gianluca Doro
- Orthopedics and Traumatology Department, Humanitas Mater Domini, Varese, Italy
| | - Enrico Ragni
- Laboratorio di Biotecnologie Applicate all'Ortopedia, IRCCS Istituto Ortopedico Galeazzi, Via R. Galeazzi 4, Milan, 20161, Italy
| | | | - Laura de Girolamo
- Laboratorio di Biotecnologie Applicate all'Ortopedia, IRCCS Istituto Ortopedico Galeazzi, Via R. Galeazzi 4, Milan, 20161, Italy.
| | - Fabio Zerbinati
- Orthopedics and Traumatology Department, Humanitas Mater Domini, Varese, Italy
| |
Collapse
|
9
|
Chai R, Yang X, Zhang AS. Different endodontic treatments induced root development of two nonvital immature teeth in the same patient: A case report. World J Clin Cases 2023; 11:2567-2575. [PMID: 37123304 PMCID: PMC10130992 DOI: 10.12998/wjcc.v11.i11.2567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/01/2023] [Accepted: 03/22/2023] [Indexed: 04/06/2023] Open
Abstract
BACKGROUND Pulp revascularization is a novel way to treat immature teeth with periapical disease, and the technique has become increasingly well established in recent years. By puncturing the periapical tissue, bleeding is induced, and a blood clot is formed in the root canal. The blood clot acts as a natural bioscaffold onto which mesenchymal stem cells from periapical tissue can be seeded and restore pulp vascularity, thus promoting root development as well as apical closure. Although the effect of pulp revascularization is ideal, there are certain requirements for the apical condition of the teeth. The apical barrier technique and apexification are still indispensable for teeth that cannot achieve ideal blood clot formation. In addition, a meta-analysis of several clinical studies concluded that pulp revascularization has no significant advantages over other treatments.
CASE SUMMARY A 10-year-old girl complained of pain in the right upper and lower posterior teeth for 2 d. Clinical and radiological examinations revealed that both the right maxillary and mandibular second premolars were immature with periapical radiolucency. The right maxillary second premolar was treated by pulp revascularization, while the right mandibular second premolar was treated by conventional apical barrier surgery after revascularization failed. The purpose of this report is to compare the different root maturation processes induced by the pulp revascularization and apical barrier techniques in the same patient in homonymous teeth from different jaws. Twelve months of follow-up showed that the apical foramen of both teeth presented a clear tendency to close; however, the tooth treated with pulp revascularization showed a significant increase in root length as well as root canal wall thickness.
CONCLUSION For the treatment of nonvital immature teeth, pulp revascularization showed a superior therapeutic effect in comparison with the apical barrier technique.
Collapse
Affiliation(s)
- Rong Chai
- Department of Stomatology, Xi'an International Medical Center Hospital Affiliated to Northwest University, Xi'an 710061, Shaanxi Province, China
| | - Xiu Yang
- Department of Stomatology, Xi'an International Medical Center Hospital Affiliated to Northwest University, Xi'an 710061, Shaanxi Province, China
| | - An-Sheng Zhang
- Department of Stomatology, Xi'an International Medical Center Hospital Affiliated to Northwest University, Xi'an 710061, Shaanxi Province, China
| |
Collapse
|
10
|
Brown M, Zhu S, Taylor L, Tabrizian M, Li-Jessen NY. Unraveling the Relevance of Tissue-Specific Decellularized Extracellular Matrix Hydrogels for Vocal Fold Regenerative Biomaterials: A Comprehensive Proteomic and In Vitro Study. ADVANCED NANOBIOMED RESEARCH 2023; 3:2200095. [PMID: 37547672 PMCID: PMC10398787 DOI: 10.1002/anbr.202200095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Decellularized extracellular matrix (dECM) is a promising material for tissue engineering applications. Tissue-specific dECM is often seen as a favorable material that recapitulates a native-like microenvironment for cellular remodeling. However, the minute quantity of dECM derivable from small organs like the vocal fold (VF) hampers manufacturing scalability. Small intestinal submucosa (SIS), a commercial product with proven regenerative capacity, may be a viable option for VF applications. This study aims to compare dECM hydrogels derived from SIS or VF tissue with respect to protein content and functionality using mass spectrometry-based proteomics and in vitro studies. Proteomic analysis reveals that VF and SIS dECM share 75% of core matrisome proteins. Although VF dECM proteins have greater overlap with native VF, SIS dECM shows less cross-sample variability. Following decellularization, significant reductions of soluble collagen (61%), elastin (81%), and hyaluronan (44%) are noted in VF dECM. SIS dECM contains comparable elastin and hyaluronan but 67% greater soluble collagen than VF dECM. Cells deposit more neo-collagen on SIS than VF-dECM hydrogels, whereas neo-elastin (~50 μg/scaffold) and neo-hyaluronan (~ 6 μg/scaffold) are comparable between the two hydrogels. Overall, SIS dECM possesses reasonably similar proteomic profile and regenerative capacity to VF dECM. SIS dECM is considered a promising alternative for dECM-derived biomaterials for VF regeneration.
Collapse
Affiliation(s)
- Mika Brown
- Department of Biomedical Engineering, McGill University 3655 Promenade Sir-William-Osler, Room 1003, Montreal, QC H3A 1A3, Canada
| | - Shirley Zhu
- Department of Microbiology and Immunology 2001 McGill College Ave, 8th Floor, Montreal, Quebec, H3A 1G1, Canada
| | - Lorne Taylor
- The Proteomics Platform, McGill University Health Center 1001 Decarie Boulevard Montreal Suite E01.5056 Montreal, Quebec, H4A 3J1, Canada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, McGill University 3655 Promenade Sir-William-Osler, Room 1003, Montreal, QC H3A 1A3, Canada
- Department of Bioengineering, McGill University 740 Avenue Dr. Penfield, Room 4300, Montreal, QC H3A 0G1, Canada
- Faculty of Dentistry, McGill University 740 Avenue Dr. Penfield, Room 4300, Montreal, QC H3A 0G1, Canada
| | - Nicole Y.K. Li-Jessen
- Department of Biomedical Engineering, McGill University 3655 Promenade Sir-William-Osler, Room 1003, Montreal, QC H3A 1A3, Canada
- School of Communication Sciences and Disorders, McGill University 2001 McGill College Ave, 8th Floor, Montreal, Quebec, H3A 1G1, Canada
- Department of Otolaryngology - Head and Neck Surgery, McGill University 2001 McGill College Ave, 8th Floor, Montreal, Quebec, H3A 1G1, Canada
- Research Institute of McGill University Health Center, McGill University 2001 McGill College Ave, 8th Floor, Montreal, Quebec, H3A 1G1, Canada
| |
Collapse
|
11
|
He L, Lan S, Cheng Q, Luo Z, Lin X. Self-Assembling Peptide SCIBIOIII Hydrogel for Three-Dimensional Cell Culture That Promotes Wound Healing in Diabetic Mice. Gels 2023; 9:gels9040265. [PMID: 37102877 PMCID: PMC10137493 DOI: 10.3390/gels9040265] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 04/28/2023] Open
Abstract
An important clinical challenge is improving the healing rate of diabetic chronic wounds, and developing new approaches that can promote chronic wound healing is essential. A new biomaterial that has demonstrated great potential for tissue regeneration and repair is self-assembling peptides (SAPs); however, they have been less studied for the treatment of diabetic wounds. Here, we explored the role of an SAP, SCIBIOIII, with a special nanofibrous structure mimicking the natural extracellular matrix for chronic diabetic wound repair. The results showed that the SCIBIOIII hydrogel in vitro has good biocompatibility and can create a three-dimensional (3D) culture microenvironment for the continuous growth of skin cells in a spherical state. The SCIBIOIII hydrogel in diabetic mice (in vivo) significantly improved wound closure, collagen deposition, and tissue remodeling and enhanced chronic wound angiogenesis. Thus, the SCIBIOIII hydrogel is a promising advanced biomaterial for 3D cell culture and diabetic wound tissue repair.
Collapse
Affiliation(s)
- Lu He
- College of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Shijian Lan
- Molecular Medicine and Cancer Research Center, College of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China
| | - Qingfeng Cheng
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhongli Luo
- Molecular Medicine and Cancer Research Center, College of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China
| | - Xuemei Lin
- College of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
12
|
Zhang Y, Li S, Zhou X, Dong L, Meng Q, Yu J. Preparation of a Cellulosic Photosensitive Hydrogel for Tubular Tissue Engineering. ACS APPLIED BIO MATERIALS 2023; 6:848-856. [PMID: 36723405 DOI: 10.1021/acsabm.2c01003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Since the concept of tissue engineering was proposed, biocompatible hydrogel materials have attracted the attention of researchers. With the help of three-dimensional (3D) printing technology, precise shaping of hydrogels can be realized. In this paper, we synthesized a cellulosic photosensitive acrylamide (AM)/N,N-methylenebisacrylamide (MBA) hydrogel. With the high-efficiency water-soluble photoinitiator TPO@Tw developed by our research group, the efficient photocuring cross-linking process of the hydrogel can be realized under 405 nm visible light. In consideration of the viscosity, curing mass, curing depth, and break distance of the hydrogel, we screened out hydroxypropyl cellulose (HPC) as the preferred tackifier of the material. The addition of HPC greatly improved the mechanical properties of the hydrogel. The compressive modulus of the optimal sample AM-HPC-5 increased by 709.2% and the tensile strength increased by 76.7% compared with the blank control group. By adding a PEGDA shell to the surface of the material, the water retention capacity of the hydrogel was effectively improved. The water loss rate was greatly reduced. The 3D wooden-pile structure model was printed by a DIW 3D printer. Further, through coaxial extrusion, the microtubule structure that may be applied in tissue engineering was obtained. Cell experiment results showed high biocompatibility of the hydrogel. NIH 3T3 cells could adhere and grow on the surface of microtubules.
Collapse
Affiliation(s)
- Yiming Zhang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shuai Li
- Department of Urology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Xiaowen Zhou
- Department of Urology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - Lize Dong
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qinghua Meng
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianjun Yu
- Department of Urology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, China
| |
Collapse
|
13
|
Shinde SD, Kulkarni N, Sahu B. Synthesis and Investigation of Backbone Modified Squaramide Dipeptide Self-Assembly. ACS APPLIED BIO MATERIALS 2023; 6:507-518. [PMID: 36716238 DOI: 10.1021/acsabm.2c00803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Dipeptides are minimalistic peptide building blocks that form well ordered structures through molecular self-assembly. The driving forces involved are cooperative noncovalent interactions such as π-π stacking, hydrogen bonding, and ionic as well as hydrophobic interactions. One of the most intriguing self-assembled motifs that has been extensively explored as a low molecular weight hydrogel for drug delivery, tissue engineering, imaging and techtonics, etc. is Phe-Phe (FF). The backbone of the dipeptide is very crucial for extending secondary structures in self-assembly, and any subtle change in the backbone drastically affect the molecular recognitions. The squaramide (SQ) motif has the unique advantage of hydrogen bonding which can promote the self-assembly process. In this work we have integrated the SQ unit into the dipeptide FF backbone to achieve molecular self-assembly. The resulting carbamate protected backbone modified dipeptide (BocFSAF-OH, 10) has exhibited molecular self-assembly with a fibrilar network. It formed a stable hydrogel (with CAC of 0.024 ± 0.0098 wt %) via the solvent switch method and was found to possess excellent enzymatic stability. The dipeptide and the resulting hydrogel were found to be cytocompatible. When integrated with a polysaccharide based biopolymer, e.g. sodium alginate, the resulting matrix exhibited strong hydrogel character. Therefore, the dipeptide hydrogel of 10 may find its applications in a variety of fields including drug delivery and tissue engineering.
Collapse
Affiliation(s)
- Suchita Dattatray Shinde
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat 380054, India
| | - Neeraj Kulkarni
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat 380054, India
| | - Bichismita Sahu
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat 380054, India
| |
Collapse
|
14
|
Chen Z, Wang L, Guo C, Qiu M, Cheng L, Chen K, Qi J, Deng L, He C, Li X, Yan Y. Vascularized polypeptide hydrogel modulates macrophage polarization for wound healing. Acta Biomater 2023; 155:218-234. [PMID: 36396041 DOI: 10.1016/j.actbio.2022.11.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 10/18/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022]
Abstract
Wound repair involves a sophisticated process that includes angiogenesis, immunoregulation and collagen deposition. However, weak revascularization performance and the lack of biochemical cues to trigger immunomodulatory function currently limit biomaterial applications for skin regeneration and tissue engineering. Herein, we fabricate a new bioactive polypeptide hydrogel (QK-SF) constituted by silk fibroin (SF) and a vascular endothelial growth factor mimetic peptide KLTWQELYQLKYKGI (QK) for tissue regeneration by simultaneously promoting vascularization and macrophage polarization. Our results showed that this QK-SF hydrogel can be prepared via an easy manufacturing process, and exhibited good gel stability and low cytotoxicity to cultured human umbilical vein endothelial cells (HUVECs) via both live/dead and cell counting kit-8 assays. Importantly, this QK-SF hydrogel triggered macrophage polarization from M1 into M2, as exemplified by the enhanced expression of the M2 marker and decreased expression of the M1 marker in RAW264.7 cells. Furthermore, the QK-SF hydrogel showed high capacity for inducing endothelial growth, migration and angiogenesis, which were proved by increased expression of angiogenesis-related genes in HUVECs. Consistent with in vitro findings, in vivo data show that the QK-SF hydrogel promoted M2 polarization, keratinocyte differentiation, and collagen deposition in the mouse skin wound model in immunohistochemistry assay. Furthermore, this QK-SF hydrogel can reduce inflammation, induce angiogenesis and promote wound healing as exemplified by the increased vessel formation and decreased wound area in the mouse skin wound model. Altogether, these results indicate that the bioactive QK-SF hydrogel plays dual functional roles in promoting angiogenesis and immunoregulation for tissue regeneration. STATEMENT OF SIGNIFICANCE: The QK-SF hydrogel plays dual functional roles in promoting angiogenesis and immunoregulation for tissue repair and wound healing. The QK-SF hydrogel can be prepared via an easy manufacturing process, and exhibited good gel stability and low cytotoxicity to cultured HUVECs. The QK-SF hydrogel triggered macrophage polarization from M1 into M2. The QK-SF hydrogel showed high capacity for inducing endothelial growth, migration and angiogenesis. The QK-SF hydrogel promoted M2 polarization, keratinocyte differentiation, and collagen deposition.
Collapse
Affiliation(s)
- Zhijie Chen
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Lianlian Wang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, PR China
| | - Changjun Guo
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Minglong Qiu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Liang Cheng
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Kaizhe Chen
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Jin Qi
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Lianfu Deng
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Chuan He
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China.
| | - Xinming Li
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, PR China.
| | - Yufei Yan
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China; Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China.
| |
Collapse
|
15
|
Cells and material-based strategies for regenerative endodontics. Bioact Mater 2022; 14:234-249. [PMID: 35310358 PMCID: PMC8897646 DOI: 10.1016/j.bioactmat.2021.11.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 10/29/2021] [Accepted: 11/09/2021] [Indexed: 12/21/2022] Open
Abstract
<p class = "Abstract" style = "margin: 0 cm; line-height: 32px; font-size: 12 pt; font-family: "Times New Roman", serif; color: rgb(0, 0, 0); "><span lang = "EN-US">The carious process leads to inflammation of pulp tissue. Current care options include root canal treatment or apexification. These procedures, however, result in the loss of tooth vitality, sensitivity, and healing. Pulp capping and dental pulp regeneration are continually evolving techniques to regenerate pulp tissue, avoiding necrosis and loss of vitality. Many studies have successfully employed stem/progenitor cell populations, revascularization approaches, scaffolds or material-based strategies for pulp regeneration. Here we outline advantages and disadvantages of different methods and techniques which are currently being used in the field of regenerative endodontics. We also summarize recent findings on efficacious peptide-based materials which target the dental niche.<o:p></o:p></span></p> Pulp infection necessitates removal of necrotic, inflamed and infected tissue. Materials used clinically are inert (such as gutta percha, mineral trioxide aggregate). Recent developments in materials (angiogenic hydrogels, stem cell composites) have tuneable bioactivity. Dental pulp regeneration may now be possible through the use of bioactive systems, that guide regeneration.
Collapse
|
16
|
Kim K, Siddiqui Z, Acevedo-Jake AM, Roy A, Choudhury M, Grasman J, Kumar V. Angiogenic Hydrogels to Accelerate Early Wound Healing. Macromol Biosci 2022; 22:e2200067. [PMID: 35579914 DOI: 10.1002/mabi.202200067] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/19/2022] [Indexed: 11/09/2022]
Abstract
The metabolic disorder diabetes mellitus affects an increasing proportion of the population, a number projected to double by 2060. Non-life-threatening comorbidities contribute to an interrupted healing process which is first delayed, then prolonged, and associated with increased susceptibility to infection and sustained and unresolved inflammation. This leads to chronic non-healing wounds and eventually potential amputation of extremities. Here we examine the use of a bioactive angiogenic peptide-based hydrogel, SLan, to improve early wound healing in diabetic rats, and compare its performance to clinically utilized biosynthetic peptide-based materials such as Puramatrix. Streptozotocin-treated diabetic rats underwent 8 mm biopsy wounding in their dorsum to remove the epithelium, adipose tissues and muscle layer of the skin, and served as a model for diabetic wound healing. Wounds were treated with either Low (1w%) SLan, High (4w%) SLan, PBS, Puramatrix or K2 (an unfunctionalized non-bioactive control sequentially similar to SLan), covered with Tegaderm and monitored on days 0, 3, 7, 10, 14, 17, 21, 28; animals were sacrificed for histomorphic analyses and immunostaining. An LC/MS method developed to detect SLan in plasma allows pharmacokinetic analysis showing no trafficking of peptides from the wound site into the circulation. Low and High SLan groups show similar final outcomes of wound contraction as control groups (Puramatrix, PBS and K2). SLan-treated rats, however, show marked improvement in healing in earlier time points, including increased deposition of new mature blood vessels. Additionally, rats in the Low SLan treatment groups showed significantly improved wound contraction over other groups and significantly improved healing in early time points. Altogether our results suggest this material can be used to "jumpstart" the diabetic wound healing process. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- KaKyung Kim
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Zain Siddiqui
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Amanda M Acevedo-Jake
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Abhishek Roy
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Marwa Choudhury
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Jonathan Grasman
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Vivek Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.,Department of Biology, New Jersey Institute of Technology, Newark, NJ, 07102, USA.,Department of Chemical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.,Department of Restorative Dentistry, Rutgers School of Dental Medicine, Newark, NJ, 07102, USA
| |
Collapse
|
17
|
Kim BR, Yoon JW, Choi H, Kim D, Kang S, Kim JH. Application of periostin peptide-decorated self-assembled protein cage nanoparticles for therapeutic angiogenesis. BMB Rep 2022. [PMID: 34814976 PMCID: PMC9058470 DOI: 10.5483/bmbrep.2022.55.4.137] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Peptides are gaining substantial attention as therapeutics for human diseases. However, they have limitations such as low bioavailability and poor pharmacokinetics. Periostin, a matricellular protein, can stimulate the repair of ischemic tissues by promoting angiogenesis. We have previously reported that a novel angiogenic peptide (amino acids 142-151) is responsible for the pro-angiogenic activity of periostin. To improve the in vivo delivery efficiency of periostin peptide (PP), we used proteins self-assembled into a hollow cage-like structure as a drug delivery nanoplatform in the present study. The periostin peptide was genetically inserted into lumazine synthase (isolated from Aquifex aeolicus) consisting of 60 identical subunits with an icosahedral capsid architecture. The periostin peptide-bearing lumazine synthase protein cage nanoparticle with 60 periostin peptides multivalently displayed was expressed in Escherichia coli and purified to homogeneity. Next, we examined angiogenic activities of this periostin peptide-bearing lumazine synthase protein cage nanoparticle. AaLS-periostin peptide (AaLS-PP), but not AaLS, promoted migration, proliferation, and tube formation of human endothelial colony-forming cells in vitro. Intramuscular injection of PP and AaLS-PP increased blood perfusion and attenuated severe limb loss in the ischemic hindlimb. However, AaLS did not increase blood perfusion or alleviate tissue necrosis. Moreover, in vivo administration of AaLS-PP, but not AaLS, stimulated angiogenesis in the ischemic hindlimb. These results suggest that AaLS is a highly useful nanoplatform for delivering pro-angiogenic peptides such as PP.
Collapse
Affiliation(s)
- Ba Reun Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas 75390-9071, TX, USA
| | - Jung Won Yoon
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Hyukjun Choi
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Dasol Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Sebyung Kang
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea
| |
Collapse
|
18
|
Kim BR, Yoon JW, Choi H, Kim D, Kang S, Kim JH. Application of periostin peptide-decorated self-assembled protein cage nanoparticles for therapeutic angiogenesis. BMB Rep 2022; 55:175-180. [PMID: 34814976 PMCID: PMC9058470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/14/2021] [Accepted: 11/10/2021] [Indexed: 09/17/2023] Open
Abstract
Peptides are gaining substantial attention as therapeutics for human diseases. However, they have limitations such as low bioavailability and poor pharmacokinetics. Periostin, a matricellular protein, can stimulate the repair of ischemic tissues by promoting angiogenesis. We have previously reported that a novel angiogenic peptide (amino acids 142-151) is responsible for the pro-angiogenic activity of periostin. To improve the in vivo delivery efficiency of periostin peptide (PP), we used proteins self-assembled into a hollow cage-like structure as a drug delivery nanoplatform in the present study. The periostin peptide was genetically inserted into lumazine synthase (isolated from Aquifex aeolicus) consisting of 60 identical subunits with an icosahedral capsid architecture. The periostin peptide-bearing lumazine synthase protein cage nanoparticle with 60 periostin peptides multivalently displayed was expressed in Escherichia coli and purified to homogeneity. Next, we examined angiogenic activities of this periostin peptide-bearing lumazine synthase protein cage nanoparticle. AaLS-periostin peptide (AaLS-PP), but not AaLS, promoted migration, proliferation, and tube formation of human endothelial colony-forming cells in vitro. Intramuscular injection of PP and AaLS-PP increased blood perfusion and attenuated severe limb loss in the ischemic hindlimb. However, AaLS did not increase blood perfusion or alleviate tissue necrosis. Moreover, in vivo administration of AaLS-PP, but not AaLS, stimulated angiogenesis in the ischemic hindlimb. These results suggest that AaLS is a highly useful nanoplatform for delivering pro-angiogenic peptides such as PP. [BMB Reports 2022; 55(4): 175-180].
Collapse
Affiliation(s)
- Ba Reun Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea, Yangsan 50612, Korea
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas 75390-9071, TX, USA, Yangsan 50612, Korea
| | - Jung Won Yoon
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea, Yangsan 50612, Korea
| | - Hyukjun Choi
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Dasol Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea, Yangsan 50612, Korea
| | - Sebyung Kang
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea, Yangsan 50612, Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea
| |
Collapse
|
19
|
Sadeghi A, Fatemi MJ, Zandi M, Bagheri T, Ghadimi T, Tamimi M, Pezeshki-Modaress M. Multilayered 3-D nanofibrous scaffold with chondroitin sulfate sustained release as dermal substitute. Int J Biol Macromol 2022; 206:718-729. [PMID: 35304196 DOI: 10.1016/j.ijbiomac.2022.03.061] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 03/04/2022] [Accepted: 03/11/2022] [Indexed: 12/31/2022]
Abstract
Electrospun nanofibers for skin tissue engineering applications face two main challenges. The low thickness of electrospun mats is the main reason for their weak load-bearing performance at clinical applications and limited cell penetration due to their small pore sizes. We have developed multi-layered nanofibrous 3D (M3DN) scaffolds comprising gelatin, polyvinyl alcohol, and chondroitin sulfate (CS) by an electrospinning method and attaching three electrospun layers via ethanol to cause interface fibers to come in contact with each other. Prepared M3DN scaffolds revealed a sustained CS release profile. The improved mechanical performance, stable release of CS, and penetration capability of the cells and blood vessels through the spaces between layers in the prepared multi-layered nanofibrous scaffolds demonstrate their potential applications in response to the increasing demand for replacement of damaged dermis. The results of animal studies on the dorsal skin of Rat with full-thickness wounds have shown that the reconstruction of full-thickness skin lesions is significantly higher for M3DN scaffolds than a control group (treated with sterile gauze). The amount of epithelization, collagen arrangement, and inflammatory cells (acute and chronic) has been investigated, and their associated results demonstrated that M3DN scaffolds have great potential for full-thickness wound restoration.
Collapse
Affiliation(s)
- Amin Sadeghi
- Soft Tissue Engineering Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Javad Fatemi
- Burn Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Plastic and Reconstructive Surgery, Hazrat Fatemeh Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mojgan Zandi
- Department of Biomaterials, Iran Polymer and Petrochemical Institute, Tehran, Iran
| | - Tooran Bagheri
- Burn Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Tayyeb Ghadimi
- Burn Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Plastic and Reconstructive Surgery, Hazrat Fatemeh Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Tamimi
- Hard Tissue Engineering Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mohamad Pezeshki-Modaress
- Burn Research Center, Iran University of Medical Sciences, Tehran, Iran; Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Acevedo-Jake A, Shi S, Siddiqui Z, Sanyal S, Schur R, Kaja S, Yuan A, Kumar VA. Preclinical Efficacy of Pro- and Anti-Angiogenic Peptide Hydrogels to Treat Age-Related Macular Degeneration. Bioengineering (Basel) 2021; 8:190. [PMID: 34940343 PMCID: PMC8698576 DOI: 10.3390/bioengineering8120190] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 11/20/2021] [Accepted: 11/21/2021] [Indexed: 12/15/2022] Open
Abstract
Pro-angiogenic and anti-angiogenic peptide hydrogels were evaluated against the standard of care wet age-related macular degeneration (AMD) therapy, Aflibercept (Eylea®). AMD was modeled in rats (laser-induced choroidal neovascularization (CNV) model), where the contralateral eye served as the control. After administration of therapeutics, vasculature was monitored for 14 days to evaluate leakiness. Rats were treated with either a low or high concentration of anti-angiogenic peptide hydrogel (0.02 wt% 8 rats, 0.2 wt% 6 rats), or a pro-angiogenic peptide hydrogel (1.0 wt% 7 rats). As controls, six rats were treated with commercially available Aflibercept and six with sucrose solution (vehicle control). Post lasering, efficacy was determined over 14 days via fluorescein angiography (FA) and spectral-domain optical coherence tomography (SD-OCT). Before and after treatment, the average areas of vascular leak per lesion were evaluated as well as the overall vessel leakiness. Unexpectedly, treatment with pro-angiogenic peptide hydrogel showed significant, immediate improvement in reducing vascular leak; in the short term, the pro-angiogenic peptide performed better than anti-angiogenic peptide hydrogel and was comparable to Aflibercept. After 14 days, both the pro-angiogenic and anti-angiogenic peptide hydrogels show a trend of improvement, comparable to Aflibercept. Based on our results, both anti-angiogenic and pro-angiogenic peptide hydrogels may prove good therapeutics in the future to treat wet AMD over a longer-term treatment period.
Collapse
Affiliation(s)
- Amanda Acevedo-Jake
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA; (A.A.-J.); (Z.S.)
| | - Siyu Shi
- Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA;
| | - Zain Siddiqui
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA; (A.A.-J.); (Z.S.)
| | - Sreya Sanyal
- Department of Biology, New Jersey Institute of Technology, Newark, NJ 07102, USA;
| | - Rebecca Schur
- Cole Eye Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA; (R.S.); (A.Y.)
| | - Simon Kaja
- Research & Development Division, Experimentica Ltd., 70211 Kuopio, Finland;
- Department of Ophthalmology, Loyola University Chicago, Maywood, IL 60153, USA
| | - Alex Yuan
- Cole Eye Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA; (R.S.); (A.Y.)
| | - Vivek A. Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA; (A.A.-J.); (Z.S.)
- Department of Biology, New Jersey Institute of Technology, Newark, NJ 07102, USA;
- Department of Chemical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
- Department of Restorative Dentistry, Rutgers School of Dental Medicine, Newark, NJ 07102, USA
| |
Collapse
|
21
|
Siddiqui Z, Sarkar B, Kim KK, Kumar A, Paul R, Mahajan A, Grasman JM, Yang J, Kumar VA. Self-assembling Peptide Hydrogels Facilitate Vascularization in Two-Component Scaffolds. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2021; 422:130145. [PMID: 34054331 PMCID: PMC8158327 DOI: 10.1016/j.cej.2021.130145] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
One of the major constraints against using polymeric scaffolds as tissue-regenerative matrices is a lack of adequate implant vascularization. Self-assembling peptide hydrogels can sequester small molecules and biological macromolecules, and they can support infiltrating cells in vivo. Here we demonstrate the ability of self-assembling peptide hydrogels to facilitate angiogenic sprouting into polymeric scaffolds after subcutaneous implantation. We constructed two-component scaffolds that incorporated microporous polymeric scaffolds and viscoelastic nanoporous peptide hydrogels. Nanofibrous hydrogels modified the biocompatibility and vascular integration of polymeric scaffolds with microscopic pores (pore diameters: 100-250 μm). In spite of similar amphiphilic sequences, charges, secondary structures, and supramolecular nanostructures, two soft hydrogels studied herein had different abilities to aid implant vascularization, but had similar levels of cellular infiltration. The functional difference of the peptide hydrogels was predicted by the difference in the bioactive moieties inserted into the primary sequences of the peptide monomers. Our study highlights the utility of soft supramolecular hydrogels to facilitate host-implant integration and control implant vascularization in biodegradable polyester scaffolds in vivo. Our study provides useful tools in designing multi-component regenerative scaffolds that recapitulate vascularized architectures of native tissues.
Collapse
Affiliation(s)
- Zain Siddiqui
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Biplab Sarkar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Ka Kyung Kim
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Arjun Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Reshma Paul
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Aryan Mahajan
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Jonathan M. Grasman
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Jian Yang
- Department of Biomedical Engineering, Huck Institutes of The Life Sciences, Materials Research Institute, Pennsylvania State University, University Park, PA, USA
| | - Vivek A. Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
- Department of Chemical & Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
- Department of Restorative Dentistry, Rutgers School of Dental Medicine, Newark, NJ, USA
| |
Collapse
|
22
|
Wang L, Chen Z, Yan Y, He C, Li X. Fabrication of injectable hydrogels from silk fibroin and angiogenic peptides for vascular growth and tissue regeneration. CHEMICAL ENGINEERING JOURNAL 2021; 418:129308. [DOI: 10.1016/j.cej.2021.129308] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
|
23
|
Siddiqui Z, Sarkar B, Kim KK, Kadincesme N, Paul R, Kumar A, Kobayashi Y, Roy A, Choudhury M, Yang J, Shimizu E, Kumar VA. Angiogenic hydrogels for dental pulp revascularization. Acta Biomater 2021; 126:109-118. [PMID: 33689817 PMCID: PMC8096688 DOI: 10.1016/j.actbio.2021.03.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 12/15/2022]
Abstract
Angiogenesis is critical for tissue healing and regeneration. Promoting angiogenesis in materials implanted within dental pulp after pulpectomy is a major clinical challenge in endodontics. We demonstrate the ability of acellular self-assembling peptide hydrogels to create extracellular matrix mimetic architectures that guide in vivo development of neovasculature and tissue deposition. The hydrogels possess facile injectability, as well as sequence-level functionalizability. We explore the therapeutic utility of an angiogenic hydrogel to regenerate vascularized pulp-like soft tissue in a large animal (canine) orthotopic model. The regenerated soft tissue recapitulates key features of native pulp, such as blood vessels, neural filaments, and an odontoblast-like layer next to dentinal tubules. Our study establishes angiogenic peptide hydrogels as potent scaffolds for promoting soft tissue regeneration in vivo. STATEMENT OF SIGNIFICANCE: A major challenge to endodontic tissue engineering is the lack of in situ angiogenesis within intracanal implants, especially after complete removal of the dental pulp. The lack of a robust vasculature in implants limit integration of matrices with the host tissue and regeneration of soft tissue. We demonstrate the development of an acellular material that promotes tissue revascularization in vivo without added growth factors, in a preclinical canine model of pulp-like soft-tissue regeneration. Such acellular biomaterials would facilitate pulp revascularization approaches in large animal models, and translation into human clinical trials.
Collapse
Affiliation(s)
- Zain Siddiqui
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Biplab Sarkar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Ka-Kyung Kim
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Nurten Kadincesme
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Reshma Paul
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Arjun Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Yoshifumi Kobayashi
- Department of Oral Biology, Department of Endodontics, Rutgers School of Dental Medicine, Newark, NJ, USA
| | - Abhishek Roy
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Marwa Choudhury
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Jian Yang
- Department of Biomedical Engineering, Huck Institutes of The Life Sciences, Materials Research Institute, Pennsylvania State University, University Park, PA, USA
| | - Emi Shimizu
- Department of Oral Biology, Department of Endodontics, Rutgers School of Dental Medicine, Newark, NJ, USA
| | - Vivek A Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA; Department of Oral Biology, Department of Endodontics, Rutgers School of Dental Medicine, Newark, NJ, USA; Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA; Department of Restorative Dentistry, Rutgers School of Dental Medicine, Newark, NJ, USA.
| |
Collapse
|
24
|
Zhu M, He H, Meng Q, Zhu Y, Ye X, Xu N, Yu J. Osteopontin sequence modified mesoporous calcium silicate scaffolds to promote angiogenesis in bone tissue regeneration. J Mater Chem B 2021; 8:5849-5861. [PMID: 32530014 DOI: 10.1039/d0tb00527d] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Sufficient blood supply remains the key issue to be addressed for an optimal performance of implanted bone tissue engineering scaffolds. Host vessel invasion is limited to a depth of only several hundred micrometers from the scaffold/host interface. In this study, an osteopontin sequenced polypeptide SVVYGLR was grafted into/onto mesoporous calcium silicate (MCS) and then 3D-printed into scaffolds. The peptide motifs can be accessed on the scaffold surfaces and released as well. In vitro studies of human umbilical vein endothelial cells (HUVECs) indicated enhanced cell adhesion and vascular-like structure formation on MCS-SVVYGLR scaffolds. At the same time, human bone marrow stromal cells (hBMSCs) showed enhanced osteogenic differentiation capability and higher expression levels of angiogenic genes and proteins as well. The results of in vivo radial defect repair tests of rabbits showed that more tubular vessels formed throughout the whole MCS-SVVYGLR scaffolds, and therefore, a more homogeneous new bone formation pattern was obtained on MCS-SVVYGLR scaffolds instead of a peripheral bone growth pattern on pure MCS scaffolds by Micro-CT and tissue staining techniques over 3 months. Relative gene and protein expressions in PI3K/AKT and ERK1/2 pathways suggested that the SVVYGLR motif on the MCS scaffold surface could initiate the PI3K/AKT signaling pathway and up-regulate ERK1/2 expression, which positively stimulated VEGF expression, to improve angiogenesis.
Collapse
Affiliation(s)
- Min Zhu
- School of Materials Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P. R. China
| | | | | | | | | | | | | |
Collapse
|
25
|
Sarkar B, Ma X, Agas A, Siddiqui Z, Iglesias-Montoro P, Nguyen PK, Kim KK, Haorah J, Kumar VA. In vivo Neuroprotective Effect of a Self-assembled Peptide Hydrogel. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2021; 408:127295. [PMID: 37842134 PMCID: PMC10571100 DOI: 10.1016/j.cej.2020.127295] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Traumatic brain injury (TBI) is associated with poor intrinsic healing responses and long-term cognitive decline. A major pathological outcome of TBI is acute glutamate-mediated excitotoxicity (GME) experienced by neurons. Short peptides based on the neuroprotective extracellular glycoprotein ependymin have shown the ability to slow down the effect of GME - however, such short peptides tend to diffuse away from target sites after in vivo delivery. We have designed a self-assembling peptide containing an ependymin mimic that can form nanofibrous matrices. The peptide was evaluated in situ to assess neuroprotective utility after an acute fluidpercussion injury. This biomimetic matrix can conform to the intracranial damaged site after delivery, due its shear-responsive rheological properties. We demonstrated the potential efficacy of the peptide for supporting neuronal survival in vitro and in vivo. Our study demonstrates the potential of these implantable acellular hydrogels for managing the acute (up to 7 days) pathophysiological sequelae after traumatic brain injury. Further work is needed to evaluate less invasive administrative routes and long-term functional and behavioral improvements after injury.
Collapse
Affiliation(s)
- Biplab Sarkar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Xiaotang Ma
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Agnieszka Agas
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Zain Siddiqui
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | | | - Peter K. Nguyen
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Ka Kyung Kim
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - James Haorah
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Vivek A. Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
- Department of Chemical & Materials Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
- Department of Restorative Dentistry, Rutgers School of Dental Medicine, Newark, NJ 07103, USA
| |
Collapse
|
26
|
Lau CYJ, Mastrobattista E. Programming supramolecular peptide materials by modulating the intermediate steps in the complex assembly pathway: Implications for biomedical applications. Curr Opin Colloid Interface Sci 2021. [DOI: 10.1016/j.cocis.2020.101396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
27
|
Harbour V, Casillas C, Siddiqui Z, Sarkar B, Sanyal S, Nguyen P, Kim KK, Roy A, Iglesias-Montoro P, Patel S, Podlaski F, Tolias P, Windsor W, Kumar V. Regulation of Lipoprotein Homeostasis by Self-Assembling Peptides. ACS APPLIED BIO MATERIALS 2020; 3:8978-8988. [PMID: 35019574 PMCID: PMC10790182 DOI: 10.1021/acsabm.0c01229] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
High levels of serum low-density lipoprotein (LDL) cholesterol contribute to atherosclerosis, a key risk factor of cardiovascular diseases. PCSK9 is a circulatory enzyme that downregulates expression of hepatic LDL receptors, concomitantly increasing serum LDL-C. This work investigates a small, self-assembling peptide, EPep2-8, as a peptide inhibitor of PCSK9. EPep2-8 is a multidomain peptide comprising a self-assembling domain, E2, conjugated to a bioactive domain, Pep2-8, previously shown to inhibit PCSK9. The E2 domain facilitates self-assembly of EPep2-8 into long, nanofibrous polymers with an underlying supramolecular β-sheet secondary structure. Intermolecular interactions between nanofibers drive EPep2-8 to form a thixotropic and cytocompatible hydrogel in aqueous and charge-neutral solutions. These properties enable EPep2-8 to be delivered as an in situ depot for regulation of lipoprotein homeostasis. In surface plasmon resonance studies, EPep2-8 bound specifically to PCSK9 with an apparent, noncovalent, and irreversible dissociation, significantly improving the binding affinity of Pep2-8 alone (KD = 667 ± 48 nM). Increased binding affinity of EPep2-8 is primarily due to the superstoichiometric interaction of the peptide with PCSK9. Promisingly, EPep2-8 retains bioactivity in vitro, engendering dose-dependent uptake of LDL-C in hepatocytes. This mechanism of self-assembly on a target site may be a simple method to improve the affinity of peptide inhibitors.
Collapse
Affiliation(s)
- Victoria Harbour
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Candice Casillas
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Zain Siddiqui
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Biplab Sarkar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Sreya Sanyal
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Peter Nguyen
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Ka Kyung Kim
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Abhishek Roy
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Patricia Iglesias-Montoro
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Saloni Patel
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Frank Podlaski
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Peter Tolias
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - William Windsor
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Vivek Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
- Department of Restorative Dentistry, Rutgers School of Dental Medicine, Newark, New Jersey 07103, United States
| |
Collapse
|
28
|
Han C, Zhang Z, Sun J, Li K, Li Y, Ren C, Meng Q, Yang J. Self-Assembling Peptide-Based Hydrogels in Angiogenesis. Int J Nanomedicine 2020; 15:10257-10269. [PMID: 33364757 PMCID: PMC7751603 DOI: 10.2147/ijn.s277046] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/22/2020] [Indexed: 12/22/2022] Open
Abstract
Ischemic diseases, especially in the heart and the brain, have become a serious threat to human health. Growth factor and cell therapy are emerging as promising therapeutic strategies; however, their retention and sustainable functions in the injured tissue are limited. Self-assembling peptide (SAP)-based hydrogels, mimicking the extracellular matrix, are therefore introduced to encapsulate and controllably release cells, cell-derived exosomes or growth factors, thus promoting angiogenesis and tissue recovery after ischemia. We will summarize the classification, composition and structure of SAPs, and the influencing factors for SAP gelation. Moreover, we will describe the functionalized SAPs, and the combinatorial therapy of cells, exosomes or growth factors with functionalized SAPs for angiogenic process as well as its advantage in immunogenicity and injectability. Finally, an outlook on future directions and challenges is provided.
Collapse
Affiliation(s)
- Chaoshan Han
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Zhiwei Zhang
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
| | - Jiacheng Sun
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ke Li
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
| | - Yangxin Li
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou 215006, People's Republic of China
| | - Chuanlu Ren
- Department of Clinical Laboratory, The 904th Hospital of the People's Liberation Army, Wuxi 214044, People's Republic of China
| | - Qingyou Meng
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou 215006, People's Republic of China
| | - Junjie Yang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
29
|
Crowe KM, Siddiqui Z, Harbour V, Kim K, Syed S, Paul R, Roy A, Naik R, Mitchell K, Mahajan A, Sarkar B, Kumar VA. Evaluation of Injectable Naloxone-Releasing Hydrogels. ACS APPLIED BIO MATERIALS 2020; 3:7858-7864. [PMID: 35019526 DOI: 10.1021/acsabm.0c01016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The opioid epidemic in the United States is a serious public health crisis affecting over 1.7 million Americans. In the last two decades, almost 450 000 people have died from an opioid overdose, with nearly 20% of these deaths occurring in 2017 and 2018 alone. During an overdose, overstimulation of the μ-opioid receptor leads to severe and potentially fatal respiratory depression. Naloxone is a competitive μ-opioid-receptor antagonist that is widely used to displace opioids and rescue from an overdose. Here, we describe the development of a slow-release, subcutaneous naloxone formulation for potential management of opioid overdose, chronic pain, and opioid-induced constipation. Naloxone is loaded into self-assembling peptide hydrogels for controlled drug release. The mechanical, chemical, and structural properties of the nanofibrous hydrogel enable subcutaneous administration and slow, diffusion-based release kinetics of naloxone over 30 days in vitro. The naloxone hydrogel scaffold showed cytocompatibility and did not alter the β-sheet secondary structure or thixotropic properties characteristic of self-assembling peptide hydrogels. Our results show that this biocompatible and injectable self-assembling peptide hydrogel may be useful as a vehicle for tunable, sustained release of therapeutic naloxone. This therapy may be particularly suited for preventing renarcotization in patients who refuse additional medical assistance following an overdose.
Collapse
Affiliation(s)
- Kaytlyn M Crowe
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Zain Siddiqui
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Victoria Harbour
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - KaKyung Kim
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Shareef Syed
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Reshma Paul
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Abhishek Roy
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Ruhi Naik
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Kayla Mitchell
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Aryan Mahajan
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Biplab Sarkar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Vivek A Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States.,Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey 07102, United States.,Department of Restorative Dentistry, Rutgers School of Dental Medicine, Newark, New Jersey 07102, United States
| |
Collapse
|
30
|
Sarkar B, Siddiqui Z, Kim KK, Nguyen PK, Reyes X, McGill TJ, Kumar VA. Implantable anti-angiogenic scaffolds for treatment of neovascular ocular pathologies. Drug Deliv Transl Res 2020; 10:1191-1202. [PMID: 32232681 PMCID: PMC7483832 DOI: 10.1007/s13346-020-00753-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The retinal physiology can accrue oxidative damage and inflammatory insults due to age and metabolic irregularities. Two notable diseases that involve retinal and choroidal neovascularization are proliferative diabetic retinopathy and wet age-related macular degeneration. Currently, these diseases are mainly treated with anti-VEGF drugs (VEGF = vascular endothelial growth factor), generally on a monthly dosage scheme. We discuss recent developments for the treatment of these diseases, including bioactive tissue-engineered materials, which may reduce frequency of dosage and propose a path forward for improving patient outcomes. Graphical abstract Development of materials for long-term intravitreal delivery for management of posterior segment diseases.
Collapse
Affiliation(s)
- Biplab Sarkar
- Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St. LSEB 316, Newark, NJ, 07102, USA
| | - Zain Siddiqui
- Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St. LSEB 316, Newark, NJ, 07102, USA
| | - Ka Kyung Kim
- Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St. LSEB 316, Newark, NJ, 07102, USA
| | - Peter K Nguyen
- Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St. LSEB 316, Newark, NJ, 07102, USA
| | - Xavier Reyes
- Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St. LSEB 316, Newark, NJ, 07102, USA
| | - Trevor J McGill
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Vivek A Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St. LSEB 316, Newark, NJ, 07102, USA.
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
- Department of Restorative Dentistry, Rutgers School of Dental Medicine, Newark, NJ, USA.
| |
Collapse
|
31
|
Gupta S, Singh I, Sharma AK, Kumar P. Ultrashort Peptide Self-Assembly: Front-Runners to Transport Drug and Gene Cargos. Front Bioeng Biotechnol 2020; 8:504. [PMID: 32548101 PMCID: PMC7273840 DOI: 10.3389/fbioe.2020.00504] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/29/2020] [Indexed: 12/18/2022] Open
Abstract
The translational therapies to promote interaction between cell and signal come with stringent eligibility criteria. The chemically defined, hierarchically organized, and simpler yet blessed with robust intermolecular association, the peptides, are privileged to make the cut-off for sensing the cell-signal for biologics delivery and tissue engineering. The signature service and insoluble network formation of the peptide self-assemblies as hydrogels have drawn a spell of research activity among the scientists all around the globe in the past decades. The therapeutic peptide market players are anticipating promising growth opportunities due to the ample technological advancements in this field. The presence of the other organic moieties, enzyme substrates and well-established protecting groups like Fmoc and Boc etc., bring the best of both worlds. Since the large sequences of peptides severely limit the purification and their isolation, this article reviews the account of last 5 years' efforts on novel approaches for formulation and development of single molecule amino acids, ultra-short peptide self-assemblies (di- and tri- peptides only) and their derivatives as drug/gene carriers and tissue-engineering systems.
Collapse
Affiliation(s)
- Seema Gupta
- Chemistry Department, Acharya Narendra Dev College, University of Delhi, New Delhi, India
| | - Indu Singh
- Chemistry Department, Acharya Narendra Dev College, University of Delhi, New Delhi, India
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Ashwani K. Sharma
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Pradeep Kumar
- Nucleic Acids Research Laboratory, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| |
Collapse
|
32
|
Maaßen A, Gebauer JM, Theres Abraham E, Grimm I, Neudörfl J, Kühne R, Neundorf I, Baumann U, Schmalz H. Triple‐Helix‐Stabilizing Effects in Collagen Model Peptides Containing PPII‐Helix‐Preorganized Diproline Modules. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201914101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Andreas Maaßen
- University of Cologne Department of Chemistry Greinstraße 4 50939 Cologne Germany
| | - Jan M. Gebauer
- University of Cologne Department of Chemistry Zülpicher Straße 47a 50674 Cologne Germany
| | - Elena Theres Abraham
- University of Cologne Department of Chemistry Zülpicher Straße 47a 50674 Cologne Germany
| | - Isabelle Grimm
- University of Cologne Department of Chemistry Greinstraße 4 50939 Cologne Germany
| | - Jörg‐Martin Neudörfl
- University of Cologne Department of Chemistry Greinstraße 4 50939 Cologne Germany
| | - Ronald Kühne
- Leibniz-Institut für Molekulare Pharmakologie (FMP) Campus Berlin-Buch Robert-Rössle-Straße 10 13125 Berlin Germany
| | - Ines Neundorf
- University of Cologne Department of Chemistry Zülpicher Straße 47a 50674 Cologne Germany
| | - Ulrich Baumann
- University of Cologne Department of Chemistry Zülpicher Straße 47a 50674 Cologne Germany
| | - Hans‐Günther Schmalz
- University of Cologne Department of Chemistry Greinstraße 4 50939 Cologne Germany
| |
Collapse
|
33
|
Maaßen A, Gebauer JM, Theres Abraham E, Grimm I, Neudörfl J, Kühne R, Neundorf I, Baumann U, Schmalz H. Triple-Helix-Stabilizing Effects in Collagen Model Peptides Containing PPII-Helix-Preorganized Diproline Modules. Angew Chem Int Ed Engl 2020; 59:5747-5755. [PMID: 31944532 PMCID: PMC7154665 DOI: 10.1002/anie.201914101] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Indexed: 02/02/2023]
Abstract
Collagen model peptides (CMPs) serve as tools for understanding stability and function of the collagen triple helix and have a potential for biomedical applications. In the past, interstrand cross-linking or conformational preconditioning of proline units through stereoelectronic effects have been utilized in the design of stabilized CMPs. To further study the effects determining collagen triple helix stability we investigated a series of CMPs containing synthetic diproline-mimicking modules (ProMs), which were preorganized in a PPII-helix-type conformation by a functionalizable intrastrand C2 bridge. Results of CD-based denaturation studies were correlated with calculated (DFT) conformational preferences of the ProM units, revealing that the relative helix stability is mainly governed by an interplay of main-chain preorganization, ring-flip preference, adaptability, and steric effects. Triple helix integrity was proven by crystal structure analysis and binding to HSP47.
Collapse
Affiliation(s)
- Andreas Maaßen
- University of CologneDepartment of ChemistryGreinstraße 450939CologneGermany
| | - Jan M. Gebauer
- University of CologneDepartment of ChemistryZülpicher Straße 47a50674CologneGermany
| | - Elena Theres Abraham
- University of CologneDepartment of ChemistryZülpicher Straße 47a50674CologneGermany
| | - Isabelle Grimm
- University of CologneDepartment of ChemistryGreinstraße 450939CologneGermany
| | | | - Ronald Kühne
- Leibniz-Institut für Molekulare Pharmakologie (FMP)Campus Berlin-BuchRobert-Rössle-Straße 1013125BerlinGermany
| | - Ines Neundorf
- University of CologneDepartment of ChemistryZülpicher Straße 47a50674CologneGermany
| | - Ulrich Baumann
- University of CologneDepartment of ChemistryZülpicher Straße 47a50674CologneGermany
| | | |
Collapse
|
34
|
Ma X, Agas A, Siddiqui Z, Kim K, Iglesias-Montoro P, Kalluru J, Kumar V, Haorah J. Angiogenic peptide hydrogels for treatment of traumatic brain injury. Bioact Mater 2020; 5:124-132. [PMID: 32128463 PMCID: PMC7042674 DOI: 10.1016/j.bioactmat.2020.01.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 01/07/2023] Open
Abstract
Traumatic brain injury (TBI) impacts over 3.17 million Americans. Management of hemorrhage and coagulation caused by vascular disruption after TBI is critical for the recovery of patients. Cerebrovascular pathologies play an important role in the underlying mechanisms of TBI. The objective of this study is to evaluate a novel regenerative medicine for the injured tissue after brain injury. We utilized a recently described synthetic growth factor with angiogenic potential to facilitate vascular growth in situ at the injury site. Previous work has shown how this injectable self-assembling peptide-based hydrogel (SAPH) creates a regenerative microenvironment for neovascularization at the injury site. Supramolecular assembly allows for thixotropy; the injectable drug delivery system provides sustained in vivo efficacy. In this study, a moderate blunt injury model was used to cause physical vascular damage and hemorrhage. The angiogenic SAPH was then applied directly on the injured rat brain. At day 7 post-TBI, significantly more blood vessels were observed than the sham and injury control group, as well as activation of VEGF-receptor 2, demonstrating the robust angiogenic response elicited by the angiogenic SAPH. Vascular markers von-Willebrand factor (vWF) and α-smooth muscle actin (α-SMA) showed a concomitant increase with blood vessel density in response to the angiogenic SAPH. Moreover, blood brain barrier integrity and blood coagulation were also examined as the parameters to indicate wound recovery post TBI. Neuronal rescue examination by NeuN and myelin basic protein staining showed that the angiogenic SAPH may provide and neuroprotective benefit in the long-term recovery.
Collapse
Affiliation(s)
- Xiaotang Ma
- Department of Biomedical Engineering, Center for Injury Bio-Mechanics, Materials and Medicine, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Agnieszka Agas
- Department of Biomedical Engineering, Center for Injury Bio-Mechanics, Materials and Medicine, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Zain Siddiqui
- Department of Biomedical Engineering, Center for Injury Bio-Mechanics, Materials and Medicine, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - KaKyung Kim
- Department of Biomedical Engineering, Center for Injury Bio-Mechanics, Materials and Medicine, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Patricia Iglesias-Montoro
- Department of Biomedical Engineering, Center for Injury Bio-Mechanics, Materials and Medicine, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Jagathi Kalluru
- Department of Biomedical Engineering, Center for Injury Bio-Mechanics, Materials and Medicine, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Vivek Kumar
- Department of Biomedical Engineering, Center for Injury Bio-Mechanics, Materials and Medicine, New Jersey Institute of Technology, Newark, NJ, 07102, USA
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
- Department of Restorative Dentistry, Rutgers School of Dental Medicine, Newark, NJ, USA
| | - James Haorah
- Department of Biomedical Engineering, Center for Injury Bio-Mechanics, Materials and Medicine, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| |
Collapse
|
35
|
Kim KK, Siddiqui Z, Patel M, Sarkar B, Kumar VA. A self-assembled peptide hydrogel for cytokine sequestration. J Mater Chem B 2020; 8:945-950. [PMID: 31919489 DOI: 10.1039/c9tb02250c] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cytokine-directed monocyte infiltration is involved in multiple pathological processes. Immuno-isolating matrices that can sequester cell-released chemokines in a microenvironment may prolong the viability and functionality of implanted materials. We describe a self-assembling peptide-based hydrogel that can capture the cytokine CCL2 released in the extracellular space by immune cells and stromal cells. The shear-responsive matrix can absorb and retain this signaling molecule needed for the chemotaxis of the infiltrating monocytes and their differentiation into phagocytic macrophages. Such cytokine-sequestering biomaterials may be useful as adjunctive materials with the delivery of exogenous implants or cell suspensions for tissue regeneration, without the administration of systemic immunosuppressants. Our work highlights the versatility of nanofibrous peptide hydrogels for modulating the biological response in tissue niches.
Collapse
Affiliation(s)
- Ka-Kyung Kim
- Department of Biomedical Engineering, New Jersey Institute of Technology, NJ, USA.
| | - Zain Siddiqui
- Department of Biomedical Engineering, New Jersey Institute of Technology, NJ, USA.
| | - Manali Patel
- Department of Biomedical Engineering, New Jersey Institute of Technology, NJ, USA.
| | - Biplab Sarkar
- Department of Biomedical Engineering, New Jersey Institute of Technology, NJ, USA.
| | - Vivek A Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, NJ, USA. and Department of Chemical and Materials Engineering, New Jersey Institute of Technology, NJ, USA and Department of Restorative Dentistry, Rutgers School of Dental Medicine, NJ, USA and KumarLab, 138 Warren St. Room 316, Newark, NJ 07102, USA
| |
Collapse
|
36
|
Tough, hybrid chondroitin sulfate nanofibers as a promising scaffold for skin tissue engineering. Int J Biol Macromol 2019; 132:63-75. [DOI: 10.1016/j.ijbiomac.2019.03.208] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 03/19/2019] [Accepted: 03/26/2019] [Indexed: 12/19/2022]
|
37
|
Petrak K, Vissapragada R, Shi S, Siddiqui Z, Kim KK, Sarkar B, Kumar VA. Challenges in Translating from Bench to Bed-Side: Pro-Angiogenic Peptides for Ischemia Treatment. Molecules 2019; 24:E1219. [PMID: 30925755 PMCID: PMC6479440 DOI: 10.3390/molecules24071219] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 03/21/2019] [Accepted: 03/25/2019] [Indexed: 02/07/2023] Open
Abstract
We describe progress and obstacles in the development of novel peptide-hydrogel therapeutics for unmet medical needs in ischemia treatment, focusing on the development and translation of therapies specifically in peripheral artery disease (PAD). Ischemia is a potentially life-threatening complication in PAD, which affects a significant percentage of the elderly population. While studies on inducing angiogenesis to treat PAD were started two decades ago, early results from animal models as well as clinical trials have not yet been translated into clinical practice. We examine some of the challenges encountered during such translation. We further note the need for sustained angiogenic effect involving whole growth factor, gene therapy and synthetic growth factor strategies. Finally, we discuss the need for tissue depots for de novo formation of microvasculature. These scaffolds can act as templates for neovasculature development to improve circulation and healing at the preferred anatomical location.
Collapse
Affiliation(s)
| | - Ravi Vissapragada
- Department of Gastrointestinal Surgery, Flinders Medical Centre, 5042 Bedford Park, South Australia, Australia.
| | - Siyu Shi
- Department of Medicine Stanford School of Medicine, Stanford, CA 94305, USA.
| | - Zain Siddiqui
- Department of Biomedical Engineering, Newark, NJ 07102, USA.
| | - Ka Kyung Kim
- Department of Biomedical Engineering, Newark, NJ 07102, USA.
| | - Biplab Sarkar
- Department of Biomedical Engineering, Newark, NJ 07102, USA.
| | - Vivek A Kumar
- Department of Biomedical Engineering, Newark, NJ 07102, USA.
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA.
- Rutgers School of Dental Medicine, Newark, NJ 07103, USA.
| |
Collapse
|