1
|
Smith AN, Gregor A, Baker L, Sharp DJ, Byrnes KR. Downregulation of Fidgetin-Like 2 Increases Microglial Function: The Relationship Between Microtubules, Morphology, and Activity. Mol Neurobiol 2025; 62:2726-2739. [PMID: 39160390 PMCID: PMC11790376 DOI: 10.1007/s12035-024-04404-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 07/27/2024] [Indexed: 08/21/2024]
Abstract
The microtubule cytoskeleton regulates microglial morphology, motility, and effector functions. The microtubule-severing enzyme, fidgetin-like 2 (FL2), negatively regulates cell motility and nerve regeneration, making it a promising therapeutic target for central nervous system injury. Microglia perform important functions in response to inflammation and injury, but how FL2 affects microglia is unclear. In this study, we investigated the role of FL2 in microglial morphology and injury responses in vitro. We first determined that the pro-inflammatory stimulus, lipopolysaccharide (LPS), induced a dose- and time-dependent reduction in FL2 expression associated with reduced microglial ramification. We then administered nanoparticle-encapuslated FL2 siRNA to knockdown FL2 and assess microglial functions compared to negative control siRNA and vehicle controls. Time-lapse live-cell microscopy showed that FL2 knockdown increased the velocity of microglial motility. After incubation with fluorescently labeled IgG-opsonized beads, FL2 knockdown increased phagocytosis. Microglia were exposed to low-dose LPS after nanoparticle treatment to model injury-induced cytokine secretion. FL2 knockdown enhanced LPS-induced cytokine secretion of IL-1α, IL-1β, and TNFα. These results identify FL2 as a regulator of microglial morphology and suggest that FL2 can be targeted to increase or accelerate microglial injury responses.
Collapse
Affiliation(s)
- Austin N Smith
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Alison Gregor
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | - David J Sharp
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kimberly R Byrnes
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
2
|
Qi Q, Su D, Zhuang S, Yao S, Heindl LM, Fan X, Lin M, Li J, Pang Y. Progress in Nanotechnology for Treating Ocular Surface Chemical Injuries: Reflecting on Advances in Ophthalmology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407340. [PMID: 39755928 PMCID: PMC11809354 DOI: 10.1002/advs.202407340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/26/2024] [Indexed: 01/06/2025]
Abstract
Ocular surface chemical injuries often result in permanent visual impairment and necessitate complex, long-term treatments. Immediate and extensive irrigation serves as the first-line intervention, followed by various therapeutic protocols applied throughout different stages of the condition. To optimize outcomes, conventional regimens increasingly incorporate biological agents and surgical techniques. In recent years, nanotechnology has made significant strides, revolutionizing the management of ocular surface chemical injuries by enabling sustained drug release, enhancing treatment efficacy, and minimizing side effects. This review provides a comprehensive analysis of the etiology, epidemiology, classification, and conventional therapies for ocular chemical burns, with a special focus on nanotechnology-based drug delivery systems in managing ocular surface chemical injuries. Twelve categories of nanocarrier platforms are examined, including liposomes, nanoemulsions, nanomicelles, nanowafers, nanostructured lipid carriers, nanoparticles, hydrogels, dendrimers, nanocomplexes, nanofibers, nanozymes, and nanocomposite materials, highlighting their advantages in targeted delivery, biocompatibility, and improved healing efficacy. Additionally, current challenges and limitations in the field are discussed and the future potential of nanotechnology in treating ocular diseases is explored. This review presents the most extensive examination of this topic to date, aiming to link recent advancements with broader therapeutic strategies.
Collapse
Affiliation(s)
- Qiaoran Qi
- Department of OphthalmologyNinth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyCenter for Basic Medical Research and Innovation in Visual System DiseasesMinistry of EducationShanghai200011China
| | - Dai Su
- Department of OphthalmologyNinth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyCenter for Basic Medical Research and Innovation in Visual System DiseasesMinistry of EducationShanghai200011China
| | - Shuqin Zhuang
- Department of OphthalmologyNinth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyCenter for Basic Medical Research and Innovation in Visual System DiseasesMinistry of EducationShanghai200011China
| | - Sunyuan Yao
- Department of OphthalmologyNinth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyCenter for Basic Medical Research and Innovation in Visual System DiseasesMinistry of EducationShanghai200011China
| | - Ludwig M. Heindl
- Department of OphthalmologyFaculty of Medicine and University Hospital CologneUniversity of Cologne50937CologneGermany
- Center for Integrated Oncology (CIO)Aachen‐Bonn‐Cologne‐DuesseldorfCologneGermany
| | - Xianqun Fan
- Department of OphthalmologyNinth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyCenter for Basic Medical Research and Innovation in Visual System DiseasesMinistry of EducationShanghai200011China
| | - Ming Lin
- Department of OphthalmologyNinth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyCenter for Basic Medical Research and Innovation in Visual System DiseasesMinistry of EducationShanghai200011China
| | - Jin Li
- Department of OphthalmologyNinth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyCenter for Basic Medical Research and Innovation in Visual System DiseasesMinistry of EducationShanghai200011China
| | - Yan Pang
- Department of OphthalmologyNinth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyCenter for Basic Medical Research and Innovation in Visual System DiseasesMinistry of EducationShanghai200011China
- Shanghai Frontiers Science Center of Drug Target Identification and DeliverySchool of Pharmaceutical SciencesShanghai Jiao Tong UniversityShanghai200240China
| |
Collapse
|
3
|
Dong Z, Wang Q, Yan Y, Qiang LO, Liu M. Evolution and functional divergence of the Fidgetin family. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119870. [PMID: 39481482 DOI: 10.1016/j.bbamcr.2024.119870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024]
Abstract
The Fidgetin (FIGN) family, which comprises FIGN, Fidgetin-like 1 (FIGNL1), and Fidgetin-like 2 (FIGNL2), is a vital group of microtubule-severing proteins. These proteins feature a conserved AAA+ domain essential for ATPase activity and a hexameric assembly. This review provides an in-depth analysis of the evolution and functional divergence of the FIGN family members, highlighting their role in the dynamic organization of the cytoskeleton. We further explore their broader biological functions across various species, systems, and subcellular localization. Although the FIGN family is conserved, each member exhibits unique structural characteristics and functions that reflect their evolutionary adaptations. FIGNL1 is found across animal species, while FIGNL2 is specific to vertebrates, thereby indicating its more recent evolutionary origin. Moreover, synteny analysis has revealed that FIGN is located in a more conserved genomic region compared to FIGNL2, which has undergone substantial evolutionary changes. The expression patterns of the FIGN members also vary across organisms and tissues. For example, FIGNL2 shows a notably reduced expression in the mammalian nervous system compared to that in lower vertebrates. The FIGN family members have distinct roles in microtubule severing, cell division, and DNA repair. Specifically, FIGN is involved in cell division and neuronal regeneration, FIGNL1 in axonal growth and DNA repair, and FIGNL2 in cell migration and vascular development. Their involvement in these processes underscores their role as potential biomarkers for certain cancers as well as therapeutic targets for diseases affecting the nervous system and cardiovascular development. All these evolutionary insights and functional distinctions of the FIGN family offer a comprehensive framework for understanding cytoskeletal regulation and its implications in health and disease.
Collapse
Affiliation(s)
- Zhangji Dong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Qing Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Yingying Yan
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Liang Oscar Qiang
- Department of Neurobiology & Anatomy at Drexel University College of Medicine, Philadelphia, PA 19104, USA
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China.
| |
Collapse
|
4
|
Chai AC, Siegwart DJ, Wang RC. Nucleic Acid Therapy for the Skin. J Invest Dermatol 2024:S0022-202X(24)02062-1. [PMID: 39269387 DOI: 10.1016/j.jid.2024.07.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 07/16/2024] [Indexed: 09/15/2024]
Abstract
Advances in sequencing technologies have facilitated the identification of the genes and mechanisms for many inherited skin diseases. Although targeted nucleic acid therapeutics for diseases in other organs have begun to be deployed in patients, the goal of precise therapeutics for skin diseases has not yet been realized. First, we review the current and emerging nucleic acid-based gene-editing and delivery modalities. Next, current and emerging viral and nanoparticle vehicles for the delivery of gene therapies are reviewed. Finally, specific skin diseases that could benefit optimally from nucleic acid therapies are highlighted. By adopting the latest technologies and addressing specific barriers related to skin biology, nucleic acid therapeutics have the potential to revolutionize treatments for patients with skin disease.
Collapse
Affiliation(s)
- Andreas C Chai
- Department of Dermatology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Medical Scientist Training Program, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Harmon Center for Regenerative Science and Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - Daniel J Siegwart
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Program in Genetic Drug Engineering, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Richard C Wang
- Department of Dermatology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA; Harold C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
5
|
Duan W, Zhao J, Gao Y, Xu K, Huang S, Zeng L, Shen JW, Zheng Y, Wu J. Porous silicon-based sensing and delivery platforms for wound management applications. J Control Release 2024; 371:530-554. [PMID: 38857787 DOI: 10.1016/j.jconrel.2024.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/28/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024]
Abstract
Wound management remains a great challenge for clinicians due to the complex physiological process of wound healing. Porous silicon (PSi) with controlled pore morphology, abundant surface chemistry, unique photonic properties, good biocompatibility, easy biodegradation and potential bioactivity represent an exciting class of materials for various biomedical applications. In this review, we focus on the recent progress of PSi in the design of advanced sensing and delivery systems for wound management applications. Firstly, we comprehensively introduce the common type, normal healing process, delaying factors and therapeutic drugs of wound healing. Subsequently, the typical fabrication, functionalization and key characteristics of PSi have been summarized because they provide the basis for further use as biosensing and delivery materials in wound management. Depending on these properties, the rise of PSi materials is evidenced by the examples in literature in recent years, which has emphasized the robust potential of PSi for wound monitoring, treatment and theranostics. Finally, challenges and opportunities for the future development of PSi-based sensors and delivery systems for wound management applications are proposed and summarized. We hope that this review will help readers to better understand current achievements and future prospects on PSi-based sensing and delivery systems for advanced wound management.
Collapse
Affiliation(s)
- Wei Duan
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Lab of Nanomedicine and Omic-based Diagnostics, Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou 310058, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Jingwen Zhao
- Lab of Nanomedicine and Omic-based Diagnostics, Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou 310058, PR China
| | - Yue Gao
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Keying Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Sheng Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Longhuan Zeng
- Department of Geriatric Medicine, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, PR China
| | - Jia-Wei Shen
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, PR China.
| | - Yongke Zheng
- Department of Geriatric Medicine, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, PR China.
| | - Jianmin Wu
- Lab of Nanomedicine and Omic-based Diagnostics, Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou 310058, PR China.
| |
Collapse
|
6
|
Smart K, Sharp DJ. The fidgetin family: Shaking things up among the microtubule-severing enzymes. Cytoskeleton (Hoboken) 2024; 81:151-166. [PMID: 37823563 DOI: 10.1002/cm.21799] [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] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023]
Abstract
The microtubule cytoskeleton is required for several crucial cellular processes, including chromosome segregation, cell polarity and orientation, and intracellular transport. These functions rely on microtubule stability and dynamics, which are regulated by microtubule-binding proteins (MTBPs). One such type of regulator is the microtubule-severing enzymes (MSEs), which are ATPases Associated with Diverse Cellular Activities (AAA+ ATPases). The most recently identified family are the fidgetins, which contain three members: fidgetin, fidgetin-like 1 (FL1), and fidgetin-like 2 (FL2). Of the three known MSE families, the fidgetins have the most diverse range of functions in the cell, spanning mitosis/meiosis, development, cell migration, DNA repair, and neuronal function. Furthermore, they offer intriguing novel therapeutic targets for cancer, cardiovascular disease, and wound healing. In the two decades since their first report, there has been great progress in our understanding of the fidgetins; however, there is still much left unknown about this unusual family. This review aims to consolidate the present body of knowledge of the fidgetin family of MSEs and to inspire deeper exploration into the fidgetins and the MSEs as a whole.
Collapse
Affiliation(s)
- Karishma Smart
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - David J Sharp
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
- Microcures, Inc., Bronx, New York, USA
| |
Collapse
|
7
|
Lu Y, Sathiyaseelan A, Zhang X, Zhang L, Han K, Wang MH. Synthesis of Starch-Based Ag 2[Fe (CN) 5NO] Nanoparticles for Utilization in Antibacterial and Wound-Dressing Applications. Antioxidants (Basel) 2024; 13:154. [PMID: 38397752 PMCID: PMC10886034 DOI: 10.3390/antiox13020154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Bacterial infections can lead to the formation of chronic wounds and delay the wound-healing process. Therefore, it is important to explore safe and efficient antimicrobial agents that have wound-healing and biocompatible properties. In this study, novel starch-fabricated silver nitroprusside nanoparticles (S-AgNP NPs) were prepared for biocompatible wound-healing applications. The study showed that S-AgNP NPs are spherical, with an average size of 356 ± 22.28 d. nm and zeta potential of -27.8 ± 2.80 mV, respectively. Furthermore, the FTIR and XRD results showed that S-AgNP NPs have functional groups and crystal structures from the silver nitroprusside nanoparticles (AgNP NPs) and starch. Additionally, S-AgNP NPs showed excellent bacterial and biofilm inhibition on B. cereus (15.6 μg/mL), L. monocytogenes (15.6 μg/mL), S. aureus (31.3 μg/mL), E. coli (31.3 μg/mL) and S. enterica (62.5 μg/mL). Moreover, S-AgNP NPs promoted cell migration and proliferation at a concentration of 62.5 μg/mL compared to AgNP NPs. Meanwhile, S-AgNP NPs had good biocompatibility and low cytotoxicity compared to AgNP NPs. Therefore, this study provided new ideas for the development of wound-healing agents with bacteriostatic properties in chronic wounds.
Collapse
Affiliation(s)
- Yuting Lu
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea; (Y.L.); (A.S.); (X.Z.); (L.Z.); (K.H.)
| | - Anbazhagan Sathiyaseelan
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea; (Y.L.); (A.S.); (X.Z.); (L.Z.); (K.H.)
| | - Xin Zhang
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea; (Y.L.); (A.S.); (X.Z.); (L.Z.); (K.H.)
| | - Lina Zhang
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea; (Y.L.); (A.S.); (X.Z.); (L.Z.); (K.H.)
| | - Kiseok Han
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea; (Y.L.); (A.S.); (X.Z.); (L.Z.); (K.H.)
| | - Myeong Hyeon Wang
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea; (Y.L.); (A.S.); (X.Z.); (L.Z.); (K.H.)
- KIIT (Kangwon Institute of Inclusive Technology), Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
8
|
Ivanova E, Hue-Beauvais C, Chaulot-Talmon A, Castille J, Laubier J, De Casanove C, Aubert-Frambourg A, Germon P, Jammes H, Le Provost F. DNA methylation and gene expression changes in mouse mammary tissue during successive lactations: part II - the impact of lactation rank. Epigenetics 2023; 18:2215620. [PMID: 37219968 PMCID: PMC10208124 DOI: 10.1080/15592294.2023.2215620] [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: 12/01/2022] [Revised: 05/09/2023] [Accepted: 05/12/2023] [Indexed: 05/25/2023] Open
Abstract
Mastitis is among the main reasons women cease breastfeeding. In farm animals, mastitis results in significant economic losses and the premature culling of some animals. Nevertheless, the effect of inflammation on the mammary gland is not completely understood. This article discusses the changes to DNA methylation in mouse mammary tissue caused by lipopolysaccharide-induced inflammation after in vivo intramammary challenges and the differences in DNA methylation between 1st and 2nd lactations. Lactation rank induces 981 differential methylations of cytosines (DMCs) in mammary tissue. Inflammation in 1st lactation compared to inflammation in 2nd lactation results in the identification of 964 DMCs. When comparing inflammation in 1st vs. 2nd lactations with previous inflammation history, 2590 DMCs were identified. Moreover, Fluidigm PCR data show changes in the expression of several genes related to mammary function, epigenetic regulation, and the immune response. We show that the epigenetic regulation of two successive physiological lactations is not the same in terms of DNA methylation and that the effect of lactation rank on DNA methylation is stronger than that of the onset of inflammation. The conditions presented here show that few DMCs are shared between comparisons, suggesting a specific epigenetic response depending on lactation rank, the presence of inflammation, and even whether the cells had previously suffered inflammation. In the long term, this information could lead to a better understanding of the epigenetic regulation of lactation in both physiological and pathological conditions.Abbreviations: RRBS, reduced representation bisulphite sequencing; RT-qPCR, real-time quantitative polymerase chain reaction; MEC, mammary epithelial cells; MaSC, mammary stem cell; TSS, transcription start site; TTS, transcription termination site; UTR, untranslated region; SINE, short interspersed nuclear element; LINE, long interspersed nuclear element; CGI, CpG island; DEG, differentially expressed gene; DMC, differentially methylated cytosine; DMR, differentially methylated region; GO term, gene ontology term; MF, molecular function; BP, biological process.
Collapse
Affiliation(s)
- E Ivanova
- INRAE, AgroParisTech, GABI, Université Paris-Saclay, Jouy-en-Josas, France
| | - C Hue-Beauvais
- INRAE, AgroParisTech, GABI, Université Paris-Saclay, Jouy-en-Josas, France
| | - A Chaulot-Talmon
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France
- BREED, Ecole Nationale Vétérinaire d’Alfort, Maisons-Alfort, France
| | - J Castille
- INRAE, AgroParisTech, GABI, Université Paris-Saclay, Jouy-en-Josas, France
| | - J Laubier
- INRAE, AgroParisTech, GABI, Université Paris-Saclay, Jouy-en-Josas, France
| | - C De Casanove
- INRAE, AgroParisTech, GABI, Université Paris-Saclay, Jouy-en-Josas, France
| | - A Aubert-Frambourg
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France
- BREED, Ecole Nationale Vétérinaire d’Alfort, Maisons-Alfort, France
| | - P Germon
- INRAE, Université de Tours, Nouzilly, France
| | - H Jammes
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France
- BREED, Ecole Nationale Vétérinaire d’Alfort, Maisons-Alfort, France
| | - F Le Provost
- INRAE, AgroParisTech, GABI, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
9
|
Correia M, Lopes J, Lopes D, Melero A, Makvandi P, Veiga F, Coelho JFJ, Fonseca AC, Paiva-Santos AC. Nanotechnology-based techniques for hair follicle regeneration. Biomaterials 2023; 302:122348. [PMID: 37866013 DOI: 10.1016/j.biomaterials.2023.122348] [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: 06/09/2023] [Revised: 09/26/2023] [Accepted: 10/05/2023] [Indexed: 10/24/2023]
Abstract
The hair follicle (HF) is a multicellular complex structure of the skin that contains a reservoir of multipotent stem cells. Traditional hair repair methods such as drug therapies, hair transplantation, and stem cell therapy have limitations. Advances in nanotechnology offer new approaches for HF regeneration, including controlled drug release and HF-specific targeting. Until recently, embryogenesis was thought to be the only mechanism for forming hair follicles. However, in recent years, the phenomenon of wound-induced hair neogenesis (WIHN) or de novo HF regeneration has gained attention as it can occur under certain conditions in wound beds. This review covers HF-specific targeting strategies, with particular emphasis on currently used nanotechnology-based strategies for both hair loss-related diseases and HF regeneration. HF regeneration is discussed in several modalities: modulation of the hair cycle, stimulation of progenitor cells and signaling pathways, tissue engineering, WIHN, and gene therapy. The HF has been identified as an ideal target for nanotechnology-based strategies for hair regeneration. However, some regulatory challenges may delay the development of HF regeneration nanotechnology based-strategies, which will be lastly discussed.
Collapse
Affiliation(s)
- Mafalda Correia
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
| | - Joana Lopes
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
| | - Daniela Lopes
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
| | - Ana Melero
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia (Campus de Burjassot), Av. Vicente A. Estelles s/n, 46100, Burjassot, Valencia, Spain
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, 324000, Quzhou, Zhejiang, China
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
| | - Jorge F J Coelho
- CEMMPRE - Department of Chemical Engineering, University of Coimbra, 3030-790, Coimbra, Portugal
| | - Ana C Fonseca
- CEMMPRE - Department of Chemical Engineering, University of Coimbra, 3030-790, Coimbra, Portugal.
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal.
| |
Collapse
|
10
|
Smart K, Kramer AH, Smart S, Hodgson L, Sharp DJ. Fidgetin-like 2 depletion enhances cell migration by regulating GEF-H1, RhoA, and FAK. Biophys J 2023; 122:3600-3610. [PMID: 36523161 PMCID: PMC10541466 DOI: 10.1016/j.bpj.2022.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/31/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
The microtubule (MT) cytoskeleton and its dynamics play an important role in cell migration. Depletion of the microtubule-severing enzyme Fidgetin-like 2 (FL2), a regulator of MT dynamics at the leading edge of migrating cells, leads to faster and more efficient cell migration. Here we examine how siRNA knockdown of FL2 increases cell motility. Förster resonance energy transfer biosensor studies shows that FL2 knockdown decreases activation of the p21 Rho GTPase, RhoA, and its activator GEF-H1. Immunofluorescence studies reveal that GEF-H1 is sequestered by the increased MT density resulting from FL2 depletion. Activation of the Rho GTPase, Rac1, however, does not change after FL2 knockdown. Furthermore, FL2 depletion leads to an increase in focal adhesion kinase activation at the leading edge, as shown by immunofluorescence studies, but no change in actin dynamics, as shown by fluorescence recovery after photobleaching. We believe these results expand our understanding of the role of MT dynamics in cell migration and offer new insights into RhoA and Rac1 regulation.
Collapse
Affiliation(s)
- Karishma Smart
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| | - Adam H Kramer
- Microcures, Inc., Research and Development, Bronx, New York
| | | | - Louis Hodgson
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York.
| | - David J Sharp
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York; Microcures, Inc., Research and Development, Bronx, New York.
| |
Collapse
|
11
|
Birnbaum R, Biswas J, Singer RH, Sharp DJ. mRNA Localization and Local Translation of the Microtubule Severing Enzyme, Fidgetin-Like 2, in Polarization, Migration and Outgrowth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.537087. [PMID: 37131812 PMCID: PMC10153175 DOI: 10.1101/2023.04.17.537087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Cell motility requires strict spatiotemporal control of protein expression. During cell migration, mRNA localization and local translation in subcellular areas like the leading edge and protrusions are particularly advantageous for regulating the reorganization of the cytoskeleton. Fidgetin-Like 2 (FL2), a microtubule severing enzyme (MSE) that restricts migration and outgrowth, localizes to the leading edge of protrusions where it severs dynamic microtubules. FL2 is primarily expressed during development but in adulthood, is spatially upregulated at the leading edge minutes after injury. Here, we show mRNA localization and local translation in protrusions of polarized cells are responsible for FL2 leading edge expression after injury. The data suggests that the RNA binding protein IMP1 is involved in the translational regulation and stabilization of FL2 mRNA, in competition with the miRNA let-7. These data exemplify the role of local translation in microtubule network reorganization during migration and elucidate an unexplored MSE protein localization mechanism.
Collapse
Affiliation(s)
- Rayna Birnbaum
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jeetayu Biswas
- Present address: Department of Medicine, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10021, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Robert H. Singer
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - David J. Sharp
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Microcures, Inc., Research and Development, Bronx, NY, 10461, USA
| |
Collapse
|
12
|
Singh SK, Dwivedi SD, Yadav K, Shah K, Chauhan NS, Pradhan M, Singh MR, Singh D. Novel Biotherapeutics Targeting Biomolecular and Cellular Approaches in Diabetic Wound Healing. Biomedicines 2023; 11:biomedicines11020613. [PMID: 36831151 PMCID: PMC9952895 DOI: 10.3390/biomedicines11020613] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/12/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Wound healing responses play a major role in chronic inflammation, which affects millions of people around the world. One of the daunting tasks of creating a wound-healing drug is finding equilibrium in the inflammatory cascade. In this study, the molecular and cellular mechanisms to regulate wound healing are explained, and recent research is addressed that demonstrates the molecular and cellular events during diabetic wound healing. Moreover, a range of factors or agents that facilitate wound healing have also been investigated as possible targets for successful treatment. It also summarises the various advances in research findings that have revealed promising molecular targets in the fields of therapy and diagnosis of cellular physiology and pathology of wound healing, such as neuropeptides, substance P, T cell immune response cDNA 7, miRNA, and treprostinil growth factors such as fibroblast growth factor, including thymosin beta 4, and immunomodulators as major therapeutic targets.
Collapse
Affiliation(s)
- Suraj Kumar Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur 492010, Chhattisgarh, India
| | - Shradha Devi Dwivedi
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur 492010, Chhattisgarh, India
| | - Krishna Yadav
- Raipur Institute of Pharmaceutical Educations and Research, Sarona, Raipur 492010, Chhattisgarh, India
| | - Kamal Shah
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, Uttar Pradesh, India
| | | | - Madhulika Pradhan
- Gracious College of Pharmacy Abhanpur Raipur, Village-Belbhata, Taluka, Abhanpur 493661, Chhattisgarh, India
| | - Manju Rawat Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur 492010, Chhattisgarh, India
| | - Deependra Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur 492010, Chhattisgarh, India
- Correspondence:
| |
Collapse
|
13
|
Sharma P, Kumar A, Agarwal T, Dey AD, Moghaddam FD, Rahimmanesh I, Ghovvati M, Yousefiasl S, Borzacchiello A, Mohammadi A, Yella VR, Moradi O, Sharifi E. Nucleic acid-based therapeutics for dermal wound healing. Int J Biol Macromol 2022; 220:920-933. [PMID: 35987365 DOI: 10.1016/j.ijbiomac.2022.08.099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/02/2022] [Accepted: 08/14/2022] [Indexed: 02/06/2023]
Abstract
Non-healing wounds have long been the subject of scientific and clinical investigations. Despite breakthroughs in understanding the biology of delayed wound healing, only limited advances have been made in properly treating wounds. Recently, research into nucleic acids (NAs) such as small-interfering RNA (siRNA), microRNA (miRNA), plasmid DNA (pDNA), aptamers, and antisense oligonucleotides (ASOs) has resulted in the development of a latest therapeutic strategy for wound healing. In this regard, dendrimers, scaffolds, lipid nanoparticles, polymeric nanoparticles, hydrogels, and metal nanoparticles have all been explored as NA delivery techniques. However, the translational possibility of NA remains a substantial barrier. As a result, different NAs must be identified, and their distribution method must be optimized. This review explores the role of NA-based therapeutics in various stages of wound healing and provides an update on the most recent findings in the development of NA-based nanomedicine and biomaterials, which may offer the potential for the invention of novel therapies for this long-term condition. Further, the challenges and potential for miRNA-based techniques to be translated into clinical applications are also highlighted.
Collapse
Affiliation(s)
- Preety Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India; Government Pharmacy College Kangra, Nagrota Bhagwan, Himachal Pradesh, India
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Tarun Agarwal
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, AP, India
| | - Asmita Deka Dey
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Farnaz Dabbagh Moghaddam
- Institute for Photonics and Nanotechnologies, National Research Council, Via Fosso del Cavaliere, 100, 00133 Rome, Italy
| | - Ilnaz Rahimmanesh
- Applied Physiology Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8174673461, Iran
| | - Mahsa Ghovvati
- Department of Radiological Sciences, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA 90095, USA
| | - Satar Yousefiasl
- School of Dentistry, Hamadan University of Medical Sciences, Hamadan 6517838736, Iran
| | - Assunta Borzacchiello
- Institute for Polymers, Composites, and Biomaterials (IPCB), National Research Council (CNR), Naples 80125, Italy
| | - Abbas Mohammadi
- Department of Chemistry, University of Isfahan, Isfahan 81746-73441, Iran
| | - Venkata Rajesh Yella
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, AP, India
| | - Omid Moradi
- Department of Chemistry, Shahr-e-Qods Branch, Islamic Azad University, 374-37515 Tehran, Iran
| | - Esmaeel Sharifi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan 6517838736, Iran.
| |
Collapse
|
14
|
Loloi J, Babar M, Davies KP, Suadicani SO. Nanotechnology as a tool to advance research and treatment of non-oncologic urogenital diseases. Ther Adv Urol 2022; 14:17562872221109023. [PMID: 35924206 PMCID: PMC9340423 DOI: 10.1177/17562872221109023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 05/31/2022] [Indexed: 11/16/2022] Open
Abstract
Nanotechnology represents an expanding area of research and innovation in almost every field of science, including Medicine, where nanomaterial-based products have been developed for diagnostic and therapeutic applications. Because of their small, nanoscale size, these materials exhibit unique physical and chemical properties that differ from those of each component when considered in bulk. In Nanomedicine, there is an increasing interest in harnessing these unique properties to engineer nanocarriers for the delivery of therapeutic agents. Nano-based drug delivery platforms have many advantages over conventional drug administration routes as this technology allows for local and transdermal applications of therapeutics that can bypass the first-pass metabolism, improves drug efficacy through encapsulation of hydrophobic drugs, and allows for a sustained and controlled release of encapsulated agents. In Urology, nano-based drug delivery platforms have been extensively investigated and implemented for cancer treatment. However, there is also great potential for use of nanotechnology to treat non-oncologic urogenital diseases. We provide an update on research that is paving the way for clinical translation of nanotechnology in the areas of erectile dysfunction (ED), overactive bladder (OAB), interstitial cystitis/bladder pain syndrome (IC/BPS), and catheter-associated urinary tract infections (CAUTIs). Overall, preclinical and clinical studies have proven the utility of nanomaterials both as vehicles for transdermal and intravesical delivery of therapeutic agents and for urinary catheter formulation with antimicrobial agents to treat non-oncologic urogenital diseases. Although clinical translation will be dependent on overcoming regulatory challenges, it is inevitable before there is universal adoption of this technology to treat non-oncologic urogenital diseases.
Collapse
|
15
|
Bhar B, Chouhan D, Pai N, Mandal BB. Harnessing Multifaceted Next-Generation Technologies for Improved Skin Wound Healing. ACS APPLIED BIO MATERIALS 2021; 4:7738-7763. [PMID: 35006758 DOI: 10.1021/acsabm.1c00880] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Dysregulation of sequential and synchronized events of skin regeneration often results in the impairment of chronic wounds. Conventional wound dressings fail to trigger the normal healing mechanism owing to the pathophysiological conditions. Tissue engineering approaches that deal with the fabrication of dressings using various biomaterials, growth factors, and stem cells have shown accelerated healing outcomes. However, most of these technologies are associated with difficulties in scalability and cost-effectiveness of the products. In this review, we survey the latest developments in wound healing strategies that have recently emerged through the multidisciplinary approaches of bioengineering, nanotechnology, 3D bioprinting, and similar cutting-edge technologies to overcome the limitations of conventional therapies. We also focus on the potential of wearable technology that supports complete monitoring of the changes occurring in the wound microenvironment. In addition, we review the role of advanced devices that can precisely enable the delivery of nanotherapeutics, oligonucleotides, and external stimuli in a controlled manner. These technological advancements offer the opportunity to actively influence the regeneration process to benefit the treatment regime further. Finally, the clinical relevance, trajectory, and prospects of this field have been discussed in brief that highlights their potential in providing a beneficial wound care solution at an affordable cost.
Collapse
Affiliation(s)
- Bibrita Bhar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Dimple Chouhan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Nakhul Pai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Biman B Mandal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.,Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.,School of Health Science and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| |
Collapse
|
16
|
Berger AG, Chou JJ, Hammond PT. Approaches to Modulate the Chronic Wound Environment Using Localized Nucleic Acid Delivery. Adv Wound Care (New Rochelle) 2021; 10:503-528. [PMID: 32496978 PMCID: PMC8260896 DOI: 10.1089/wound.2020.1167] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
Significance: Nonhealing wounds have been the subject of decades of basic and clinical research. Despite new knowledge about the biology of impaired wound healing, little progress has been made in treating chronic wounds, leaving patients with few therapeutic options. Diabetic ulcers are a particularly common form of nonhealing wound. Recent Advances: Recently, investigation of therapeutic nucleic acids (TNAs), including plasmid DNA, small interfering RNA, microRNA mimics, anti-microRNA oligonucleotides, messenger RNA, and antisense oligonucleotides, has created a new treatment strategy for chronic wounds. TNAs can modulate the wound toward a prohealing environment by targeting gene pathways associated with inflammation, proteases, cell motility, angiogenesis, epithelialization, and oxidative stress. A variety of delivery systems have been investigated for TNAs, including dendrimers, lipid nanoparticles (NPs), polymeric micelles, polyplexes, metal NPs, and hydrogels. This review summarizes recent developments in TNA delivery for therapeutic targets associated with chronic wounds, with an emphasis on diabetic ulcers. Critical Issues: Translational potential of TNAs remains a key challenge; we highlight some drug delivery approaches for TNAs that may hold promise. We also describe current commercial efforts to locally deliver nucleic acids to modulate the wound environment. Future Directions: Localized nucleic acid delivery holds promise for the treatment of nonhealing chronic wounds. Future efforts to improve targeting of these nucleic acid therapies in the wound with both spatial and temporal control through drug delivery systems will be crucial to successful clinical translation.
Collapse
Affiliation(s)
- Adam G. Berger
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Harvard-MIT Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jonathan J. Chou
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Paula T. Hammond
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
17
|
Jozic I, Abujamra BA, Elliott MH, Wikramanayake TC, Marjanovic J, Stone RC, Head CR, Pastar I, Kirsner RS, Andreopoulos FM, Musi JP, Tomic-Canic M. Glucocorticoid-mediated induction of caveolin-1 disrupts cytoskeletal organization, inhibits cell migration and re-epithelialization of non-healing wounds. Commun Biol 2021; 4:757. [PMID: 34145387 PMCID: PMC8213848 DOI: 10.1038/s42003-021-02298-5] [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: 03/11/2021] [Accepted: 05/28/2021] [Indexed: 12/17/2022] Open
Abstract
Although impaired keratinocyte migration is a recognized hallmark of chronic wounds, the molecular mechanisms underpinning impaired cell movement are poorly understood. Here, we demonstrate that both diabetic foot ulcers (DFUs) and venous leg ulcers (VLUs) exhibit global deregulation of cytoskeletal organization in genomic comparison to normal skin and acute wounds. Interestingly, we found that DFUs and VLUs exhibited downregulation of ArhGAP35, which serves both as an inactivator of RhoA and as a glucocorticoid repressor. Since chronic wounds exhibit elevated levels of cortisol and caveolin-1 (Cav1), we posited that observed elevation of Cav1 expression may contribute to impaired actin-cytoskeletal signaling, manifesting in aberrant keratinocyte migration. We showed that Cav1 indeed antagonizes ArhGAP35, resulting in increased activation of RhoA and diminished activation of Cdc42, which can be rescued by Cav1 disruption. Furthermore, we demonstrate that both inducible keratinocyte specific Cav1 knockout mice, and MβCD treated diabetic mice, exhibit accelerated wound closure. Taken together, our findings provide a previously unreported mechanism by which Cav1-mediated cytoskeletal organization prevents wound closure in patients with chronic wounds.
Collapse
Affiliation(s)
- Ivan Jozic
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Beatriz Abdo Abujamra
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michael H Elliott
- Departments of Ophthalmology, Physiology, and Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tongyu C Wikramanayake
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jelena Marjanovic
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rivka C Stone
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Cheyanne R Head
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Robert S Kirsner
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Fotios M Andreopoulos
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juan P Musi
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
18
|
Baker L, Tar M, Kramer AH, Villegas GA, Charafeddine RA, Vafaeva O, Nacharaju P, Friedman J, Davies KP, Sharp DJ. Fidgetin-like 2 negatively regulates axonal growth and can be targeted to promote functional nerve regeneration. JCI Insight 2021; 6:138484. [PMID: 33872220 PMCID: PMC8262307 DOI: 10.1172/jci.insight.138484] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 04/01/2021] [Indexed: 02/05/2023] Open
Abstract
The microtubule (MT) cytoskeleton plays a critical role in axon growth and guidance. Here, we identify the MT-severing enzyme fidgetin-like 2 (FL2) as a negative regulator of axon regeneration and a therapeutic target for promoting nerve regeneration after injury. Genetic knockout of FL2 in cultured adult dorsal root ganglion neurons resulted in longer axons and attenuated growth cone retraction in response to inhibitory molecules. Given the axonal growth-promoting effects of FL2 depletion in vitro, we tested whether FL2 could be targeted to promote regeneration in a rodent model of cavernous nerve (CN) injury. The CNs are parasympathetic nerves that regulate blood flow to the penis, which are commonly damaged during radical prostatectomy (RP), resulting in erectile dysfunction (ED). Application of FL2-siRNA after CN injury significantly enhanced functional nerve recovery. Remarkably, following bilateral nerve transection, visible and functional nerve regeneration was observed in 7 out of 8 animals treated with FL2-siRNA, while no control-treated animals exhibited regeneration. These studies identify FL2 as a promising therapeutic target for enhancing regeneration after peripheral nerve injury and for mitigating neurogenic ED after RP - a condition for which, at present, only poor treatment options exist.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - David J. Sharp
- Department of Physiology and Biophysics
- Dominick P. Purpura Department of Neuroscience, and
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
19
|
Hegde A, Ananthan ASHP, Kashyap C, Ghosh S. Wound Healing by Keratinocytes: A Cytoskeletal Perspective. J Indian Inst Sci 2021. [DOI: 10.1007/s41745-020-00219-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
20
|
Dong Z, Chen X, Li Y, Zhuo R, Lai X, Liu M. Microtubule Severing Protein Fignl2 Contributes to Endothelial and Neuronal Branching in Zebrafish Development. Front Cell Dev Biol 2021; 8:593234. [PMID: 33585441 PMCID: PMC7873885 DOI: 10.3389/fcell.2020.593234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/21/2020] [Indexed: 11/13/2022] Open
Abstract
Previously, fidgetin (fign) and its family members fidgetin-like 1 (fignl1) and fidgetin-like 2 (fignl2) were found to be highly expressed during zebrafish brain development, suggesting their functions in the nervous system. In this study, we report the effects of loss-of-function of these genes on development. We designed and identified single-guide RNAs targeted to generate fign, fignl1, and fignl2 mutants and then observed the overall morphological and behavioral changes. Our findings showed that while fign and fignl1 null mutants displayed no significant defects, fignl2 null zebrafish mutants displayed pericardial edema, reduced heart rate, and smaller eyes; fignl2 null mutants responded to the light-darkness shift with a lower swimming velocity. fignl2 mRNAs were identified in vascular endothelial cells by in situ hybridization and re-analysis of an online dataset of single-cell RNAseq results. Finally, we used morpholino oligonucleotides to confirm that fignl2 knockdown resulted in severe heart edema, which was caused by abnormal vascular branching. The zebrafish fignl2 morphants also showed longer axonal length and more branches of caudal primary neurons. Taken together, we summarize that Fignl2 functions on cellular branches in endothelial cells and neurons. This study reported for the first time that the microtubule-severing protein Fignl2 contributes to cell branching during development.
Collapse
Affiliation(s)
- Zhangji Dong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xu Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yuanyuan Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Run Zhuo
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xiaona Lai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
21
|
Wang J, Dey A, Kramer AH, Miao Y, Liu J, Baker L, Friedman JM, Nacharaju P, Chuck RS, Zhang C, Sharp DJ. A Novel Therapeutic Approach to Corneal Alkaline Burn Model by Targeting Fidgetin-Like 2, a Microtubule Regulator. Transl Vis Sci Technol 2021; 10:17. [PMID: 33510956 PMCID: PMC7804583 DOI: 10.1167/tvst.10.1.17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 12/11/2020] [Indexed: 12/26/2022] Open
Abstract
Purpose The purpose of this study was to determine the efficacy of nanoparticle-encapsulated Fidgetin-like 2 (FL2) siRNA (FL2-NPsi), a novel therapeutic agent targeting the FL2 gene, for the treatment of corneal alkaline chemical injury. Methods Eighty 12-week-old, male Sprague-Dawley rats were divided evenly into 8 treatment groups: prednisolone, empty nanoparticles, control-NPsi (1 µM, 10 µM, and 20 µM) and FL2-NPsi (1 µM, 10 µM, and 20 µM). An alkaline burn was induced onto the cornea of each rat, which was then treated for 14 days according to group assignment. Clinical, histopathologic, and immunohistochemical analyses were conducted to assess for wound healing. FL2-NPsi-mediated knockdown of FL2 was confirmed by in vitro quantitative polymerase chain reaction (qPCR). Toxicity assays were performed to assess for apoptosis (terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling [TUNEL] assay) and nerve damage (whole mount immunochemical staining). Statistical analyses were performed using Student's t-test and ANOVA. Results Compared with controls, FL2-NPsi-treated groups demonstrated enhanced corneal wound healing, with the 10 and 20 µM FL2-NPsi-treated groups demonstrating maximum rates of corneal re-epithelialization as assessed by ImageJ software, enhanced corneal transparency, and improved stromal organization on histology. Immunohistochemical analysis of vascular endothelial cells, macrophages, and neutrophils did not show significant differences between treatment groups. FL2-NPsi was not found to be toxic to nerves or induce apoptosis (p = 0.917). Conclusions Dose-response studies found both 10 and 20 µM FL2-NPsi to be efficacious in this rat model. FL2-NPsi may offer a novel treatment for corneal alkaline chemical injuries. Translational Relevance Basic cell biology findings about the microtubule cytoskeleton were used to design a therapeutic to enhance corneal cell migration, highlighting the promise of targeting microtubules to regulate corneal wound healing.
Collapse
Affiliation(s)
- Jessie Wang
- Department of Ophthalmology & Visual Sciences, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
- MicroCures, Inc., Bronx, NY, USA
| | | | | | - Yuan Miao
- Department of Ophthalmology & Visual Sciences, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Juan Liu
- Department of Ophthalmology & Visual Sciences, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Joel M. Friedman
- Department of Physiology & Biophysics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Parimala Nacharaju
- Department of Physiology & Biophysics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Roy S. Chuck
- Department of Ophthalmology & Visual Sciences, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Cheng Zhang
- Department of Ophthalmology & Visual Sciences, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - David J. Sharp
- Department of Ophthalmology & Visual Sciences, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
- MicroCures, Inc., Bronx, NY, USA
- Department of Physiology & Biophysics, Albert Einstein College of Medicine, Bronx, NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
22
|
Roles for microtubules in the proliferative and differentiated cells of stratified epithelia. Curr Opin Cell Biol 2020; 68:98-104. [PMID: 33186891 DOI: 10.1016/j.ceb.2020.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/24/2020] [Accepted: 10/05/2020] [Indexed: 12/27/2022]
Abstract
While microtubule dynamics and organization have been extensively studied invitro, both biochemically and in cultured cells, recent work has begun to extend this into tissues ex vivo and organisms in vivo. Advances in genetic tools and imaging technology have allowed studies on the dynamics, function, and organization of microtubules in the stratified epithelia of the epidermis. Here, we discuss recent work that highlights the varied roles that microtubules play in supporting epidermal function. These findings demonstrate that studying microtubules in tissues has revealed not only novel aspects of epidermal biology but also new principles of microtubule regulation.
Collapse
|
23
|
Dong Z, Li Y, Chen X, Lai X, Liu M. A comparative study of the expression patterns of Fign family members in zebrafish embryonic development. Comp Biochem Physiol B Biochem Mol Biol 2020; 251:110522. [PMID: 33069857 DOI: 10.1016/j.cbpb.2020.110522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 10/23/2022]
Abstract
During development, highly dynamic reconstruction of microtubules is involved in many cellular processes, including cell division, migration, morphological changes, and material transportation within cells. Microtubule severing proteins (MSPs), with the function of cutting microtubules into short parts, are important regulators in the reconstruction of microtubule arrays. Fidgetin (fign) and its family members fidgetin like 1 (fignl1) and fignl2 are MSPs, and knowledge on the expression patterns of fign family members will benefit our understanding of their primary roles in one specific stage during development. In this study, we compared the evolutionary relationships of fign family members and found that fignl2 is closer to fign than fignl1. We utilized the zebrafish model and in situ hybridization (ISH) to parallelly identify the expression features of fign family members. Our findings revealed that before 12 h post fertilization (hpf), the expression patterns of fign and fignl1 and fignl2 genes were similar, but differences arose thereafter. Fignl2 transcripts were present in more tissues and organs of zebrafish after 12 hpf and potentially exhibited more ubiquitous functions. This study is the first to assess systematic comparable data on the expression patterns of fign family members during development.
Collapse
Affiliation(s)
- Zhangji Dong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong Jiangsu 226001, China
| | - Yuanyuan Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong Jiangsu 226001, China
| | - Xu Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong Jiangsu 226001, China
| | - Xiaona Lai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong Jiangsu 226001, China
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong Jiangsu 226001, China.
| |
Collapse
|
24
|
Pharmacological activation of Nrf2 promotes wound healing. Eur J Pharmacol 2020; 886:173395. [PMID: 32710954 DOI: 10.1016/j.ejphar.2020.173395] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/09/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023]
Abstract
Wound repair and regeneration is a complex orchestrated process, comprising several phases interconnecting various cellular events and triggering multiple intracellular molecular pathways in damaged cells and tissues. In several metabolic disorders including diabetes mellitus, delay in wound healing due to elevated levels of cellular stress poses a key challenge. Several therapeutic wound dressing materials and strategies including hyperbaric oxygen therapy and negative pressure wound therapy have been developed to accelerate repair and restore cellular homeostasis at the wound site. Further, tremendous progress has been made in identification of transcriptional regulators involved in the process of wound healing. Nuclear factor erythroid 2-related factor 2 (Nrf2), a redox sensitive transcription factor, is the key regulator of intracellular redox homeostasis which induces the expression of cytoprotective genes and increases the production of antioxidants that scavenge free radicals. Activators of Nrf2 have been reported to combat oxidative stress and enhance the process of wound healing in several pathophysiological conditions, including diabetes and its complications such as diabetic foot ulcer, and chronic kidney disease, and diabetic nephropathy. Several bioactive compounds have been reported to reduce cellular stress, and thus accelerate cell proliferation, neovascularization results in repairing damaged tissues by the activation of the transcription factor, Nrf2. This review is focused on the strategies for diabetic wound healing and the highlights the role of bioactive compounds that activate the Nrf2 signaling and revitalize the cellular and molecular mechanism in the chronic wound niche, regulate and restore redox homeostasis thereby promoting wound repair and regeneration.
Collapse
|
25
|
Tezgel Ö, DiStasio N, Laghezza-Masci V, Taddei AR, Szarpak-Jankowska A, Auzély-Velty R, Navarro FP, Texier I. Collagen scaffold-mediated delivery of NLC/siRNA as wound healing materials. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2019.101421] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
26
|
Baas PW, Qiang L. Tau: It's Not What You Think. Trends Cell Biol 2019; 29:452-461. [PMID: 30929793 PMCID: PMC6527491 DOI: 10.1016/j.tcb.2019.02.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 02/12/2019] [Accepted: 02/21/2019] [Indexed: 12/12/2022]
Abstract
Tau is a multifunctional microtubule-associated protein in the neuron. For decades, tau's main function in neurons has been broadly accepted as stabilizing microtubules in the axon; however, this conclusion was reached mainly on the basis of studies performed in vitro and on ectopic expression of tau in non-neuronal cells. The idea has become so prevailing that some disease researchers are even seeking to use microtubule-stabilizing drugs to treat diseases in which tau dissociates from microtubules. Recent work suggests that tau is not a stabilizer of microtubules in the axon, but rather enables axonal microtubules to have long labile domains, in part by outcompeting genuine stabilizers. This new perspective on tau challenges long-standing dogma.
Collapse
Affiliation(s)
- Peter W Baas
- Drexel University College of Medicine, Department of Neurobiology and Anatomy, 2900 Queen Lane, Philadelphia, PA 19129, USA.
| | - Liang Qiang
- Drexel University College of Medicine, Department of Neurobiology and Anatomy, 2900 Queen Lane, Philadelphia, PA 19129, USA
| |
Collapse
|
27
|
O'Rourke BP, Kramer AH, Cao LL, Inayathullah M, Guzik H, Rajadas J, Nosanchuk JD, Sharp DJ. Fidgetin-Like 2 siRNA Enhances the Wound Healing Capability of a Surfactant Polymer Dressing. Adv Wound Care (New Rochelle) 2019; 8:91-100. [PMID: 30911440 PMCID: PMC6430983 DOI: 10.1089/wound.2018.0827] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/07/2018] [Indexed: 02/02/2023] Open
Abstract
Microtubules (MTs) are intracellular polymers that provide structure to the cell, serve as railways for intracellular transport, and regulate many cellular activities, including cell migration. The dynamicity and function of the MT cytoskeleton are determined in large part by its regulatory proteins, including the recently discovered MT severing enzyme Fidgetin-like 2 (FL2). Downregulation of FL2 expression with small interfering RNA (siRNA) results in a more than twofold increase in cell migration rate in vitro as well as translates into improved wound-healing outcomes in in vivo mouse models. Here we utilized a commercially available surfactant polymer dressing (SPD) as a vehicle to deliver FL2 siRNA. To this end we incorporated collagen microparticles containing FL2 siRNA into SPD (SPD-FL2-siRNA) for direct application to the injury site. Topical application of SPD-FL2 siRNA to murine models of full-thickness excision wounds and full-thickness burn wounds resulted in significant improvements in the rate and quality of wound healing, as measured clinically and histologically, compared with controls. Wound healing occurred more rapidly and with high fidelity, resulting in properly organized collagen substructure. Taken together, these findings indicate that the incorporation of FL2 siRNA into existing treatment options is a promising avenue to improve wound outcomes.
Collapse
Affiliation(s)
| | - Adam H. Kramer
- Physiology and Biophysics, and Albert Einstein College of Medicine, Bronx, New York
| | - Longyue L. Cao
- Department of Medicine, Children's Hospital Boston, Boston, Massachusetts
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Mohammed Inayathullah
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University, Palo Alto, California
| | - Hillary Guzik
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, New York
| | - Jayakumar Rajadas
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University, Palo Alto, California
| | - Joshua D. Nosanchuk
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - David J. Sharp
- MicroCures, Inc., Research and Development, Bronx, New York
- Physiology and Biophysics, and Albert Einstein College of Medicine, Bronx, New York
- Department of Ophthalmology and Visual Sciences, and Albert Einstein College of Medicine, Bronx, New York
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
28
|
Knockdown of Fidgetin Improves Regeneration of Injured Axons by a Microtubule-Based Mechanism. J Neurosci 2019; 39:2011-2024. [PMID: 30647150 DOI: 10.1523/jneurosci.1888-18.2018] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/21/2018] [Accepted: 12/24/2018] [Indexed: 12/16/2022] Open
Abstract
Fidgetin is a microtubule-severing protein that pares back the labile domains of microtubules in the axon. Experimental depletion of fidgetin results in elongation of the labile domains of microtubules and faster axonal growth. To test whether fidgetin knockdown assists axonal regeneration, we plated dissociated adult rat DRGs transduced using AAV5-shRNA-fidgetin on a laminin substrate with spots of aggrecan, a growth-inhibitory chondroitin sulfate proteoglycan. This cell culture assay mimics the glial scar formed after CNS injury. Aggrecan is more concentrated at the edge of the spot, such that axons growing from within the spot toward the edge encounter a concentration gradient that causes growth cones to become dystrophic and axons to retract or curve back on themselves. Fidgetin knockdown resulted in faster-growing axons on both laminin and aggrecan and enhanced crossing of axons from laminin onto aggrecan. Strikingly, axons from within the spot grew more avidly against the inhibitory aggrecan concentration gradient to cross onto laminin, without retracting or curving back. We also tested whether depleting fidgetin improves axonal regeneration in vivo after a dorsal root crush in adult female rats. Whereas control DRG neurons failed to extend axons across the dorsal root entry zone after injury, DRG neurons in which fidgetin was knocked down displayed enhanced regeneration of axons across the dorsal root entry zone into the spinal cord. Collectively, these results establish fidgetin as a novel therapeutic target to augment nerve regeneration and provide a workflow template by which microtubule-related targets can be compared in the future.SIGNIFICANCE STATEMENT Here we establish a workflow template from cell culture to animals in which microtubule-based treatments can be tested and compared with one another for their effectiveness in augmenting regeneration of injured axons relevant to spinal cord injury. The present work uses a viral transduction approach to knock down fidgetin from rat neurons, which coaxes nerve regeneration by elevating microtubule mass in their axons. Unlike previous strategies using microtubule-stabilizing drugs, fidgetin knockdown adds microtubule mass that is labile (rather than stable), thereby better recapitulating the growth status of a developing axon.
Collapse
|
29
|
Khashim Z, Samuel S, Duraisamy N, Krishnan K. Potential Biomolecules and Current Treatment Technologies for Diabetic Foot Ulcer: An Overview. Curr Diabetes Rev 2019; 15:2-14. [PMID: 28523994 DOI: 10.2174/1573399813666170519102406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/20/2017] [Accepted: 05/03/2017] [Indexed: 01/13/2023]
Abstract
BACKGROUND Diabetic foot ulceration remains a major challenge and is one of the most expensive and leading causes of major and minor amputations among patients with diabetic foot ulcer. Hence the purpose of this review is to emphasize on potential molecular markers involved in diabetic foot ulcer physiology, the efficacy of different types of dressing materials, adjunct therapy and newer therapeutic approach like nanoparticles for the treatment of diabetic foot ulcer. METHODS We conducted a systematic literature review search by using Pubmed and other web searches. The quality evidence of diabetic foot ulcer biomolecules and treatments was collected, summarized and compared with other studies. RESULTS The present investigation suggested that impaired wound healing in diabetic patients is an influence of several factors. All the advanced therapies and foot ulcer dressing materials are not suitable for all types of diabetic foot ulcers, however more prospective follow ups and in vivo and in vitro studies are needed to draw certain conclusion. Several critical wound biomolecules have been identified and are in need to be investigated in diabetic foot ulcers. The application of biocompatible nanoparticles holds a promising approach for designing dressing materials for the treatment of diabetic foot ulcer. CONCLUSION Understanding the cellular and molecular events and identifying the appropriate treatment strategies for different foot ulcer grades will reduce recurrence of foot ulcer and lower limb amputation.
Collapse
Affiliation(s)
- Zenith Khashim
- Department of Biotechnology, University of Madras, Chennai, India
| | - Shila Samuel
- Department of Biochemistry, VRR Institute of Biomedical Science, 1/7, MRB Avenue, Kattupakkam, Chennai-600056, India
| | | | | |
Collapse
|
30
|
Scharf C, Eymann C, Emicke P, Bernhardt J, Wilhelm M, Görries F, Winter J, von Woedtke T, Darm K, Daeschlein G, Steil L, Hosemann W, Beule A. Improved Wound Healing of Airway Epithelial Cells Is Mediated by Cold Atmospheric Plasma: A Time Course-Related Proteome Analysis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7071536. [PMID: 31223425 PMCID: PMC6541959 DOI: 10.1155/2019/7071536] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/28/2019] [Indexed: 01/08/2023]
Abstract
The promising potential of cold atmospheric plasma (CAP) treatment as a new therapeutic option in the field of medicine, particularly in Otorhinolaryngology and Respiratory medicine, demands primarily the assessment of potential risks and the prevention of any direct and future cell damages. Consequently, the application of a special intensity of CAP that is well tolerated by cells and tissues is of particular interest. Although improvement of wound healing by CAP treatment has been described, the underlying mechanisms and the molecular influences on human tissues are so far only partially characterized. In this study, human S9 bronchial epithelial cells were treated with cold plasma of atmospheric pressure plasma jet that was previously proven to accelerate the wound healing in a clinically relevant extent. We studied the detailed cellular adaptation reactions for a specified plasma intensity by time-resolved comparative proteome analyses of plasma treated vs. nontreated cells to elucidate the mechanisms of the observed improved wound healing and to define potential biomarkers and networks for the evaluation of plasma effects on human epithelial cells. K-means cluster analysis and time-related analysis of fold-change factors indicated concordantly clear differences between the short-term (up to 1 h) and long-term (24-72 h) adaptation reactions. Thus, the induction of Nrf2-mediated oxidative and endoplasmic reticulum stress response, PPAR-alpha/RXR activation as well as production of peroxisomes, and prevention of apoptosis already during the first hour after CAP treatment are important cell strategies to overcome oxidative stress and to protect and maintain cell integrity and especially microtubule dynamics. After resolving of stress, when stress adaptation was accomplished, the cells seem to start again with proliferation and cellular assembly and organization. The observed strategies and identification of marker proteins might explain the accelerated wound healing induced by CAP, and these indicators might be subsequently used for risk assessment and quality management of application of nonthermal plasma sources in clinical settings.
Collapse
Affiliation(s)
- Christian Scharf
- 1Department of Otorhinolaryngology, Head and Neck Surgery, University Medicine Greifswald, Germany
| | - Christine Eymann
- 1Department of Otorhinolaryngology, Head and Neck Surgery, University Medicine Greifswald, Germany
| | - Philipp Emicke
- 1Department of Otorhinolaryngology, Head and Neck Surgery, University Medicine Greifswald, Germany
| | - Jörg Bernhardt
- 2Institute for Microbiology, University of Greifswald, Germany
| | - Martin Wilhelm
- 1Department of Otorhinolaryngology, Head and Neck Surgery, University Medicine Greifswald, Germany
| | - Fabian Görries
- 1Department of Otorhinolaryngology, Head and Neck Surgery, University Medicine Greifswald, Germany
| | - Jörn Winter
- 3Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
| | - Thomas von Woedtke
- 3Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
- 4Department of Hygiene and Environmental Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Katrin Darm
- 1Department of Otorhinolaryngology, Head and Neck Surgery, University Medicine Greifswald, Germany
| | - Georg Daeschlein
- 5Department of Dermatology, University Medicine Greifswald, Greifswald, Germany
| | - Leif Steil
- 6Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Germany
| | - Werner Hosemann
- 1Department of Otorhinolaryngology, Head and Neck Surgery, University Medicine Greifswald, Germany
| | - Achim Beule
- 1Department of Otorhinolaryngology, Head and Neck Surgery, University Medicine Greifswald, Germany
- 7Department of Otorhinolaryngology, University Hospital Münster, Münster, Germany
| |
Collapse
|
31
|
McNally FJ, Roll-Mecak A. Microtubule-severing enzymes: From cellular functions to molecular mechanism. J Cell Biol 2018; 217:4057-4069. [PMID: 30373906 PMCID: PMC6279391 DOI: 10.1083/jcb.201612104] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 08/13/2018] [Accepted: 10/04/2018] [Indexed: 12/24/2022] Open
Abstract
McNally and Roll-Mecak review the molecular mechanism of microtubule-severing enzymes and their diverse roles in processes ranging from cell division to ciliogensis and morphogenesis. Microtubule-severing enzymes generate internal breaks in microtubules. They are conserved in eukaryotes from ciliates to mammals, and their function is important in diverse cellular processes ranging from cilia biogenesis to cell division, phototropism, and neurogenesis. Their mutation leads to neurodegenerative and neurodevelopmental disorders in humans. All three known microtubule-severing enzymes, katanin, spastin, and fidgetin, are members of the meiotic subfamily of AAA ATPases that also includes VPS4, which disassembles ESCRTIII polymers. Despite their conservation and importance to cell physiology, the cellular and molecular mechanisms of action of microtubule-severing enzymes are not well understood. Here we review a subset of cellular processes that require microtubule-severing enzymes as well as recent advances in understanding their structure, biophysical mechanism, and regulation.
Collapse
Affiliation(s)
- Francis J McNally
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA
| | - Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, MD .,Biochemistry and Biophysics Center, National Heart, Lung and Blood Institute, Bethesda, MD
| |
Collapse
|
32
|
Vemu A, Szczesna E, Zehr EA, Spector JO, Grigorieff N, Deaconescu AM, Roll-Mecak A. Severing enzymes amplify microtubule arrays through lattice GTP-tubulin incorporation. Science 2018; 361:eaau1504. [PMID: 30139843 PMCID: PMC6510489 DOI: 10.1126/science.aau1504] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 07/18/2018] [Indexed: 12/16/2022]
Abstract
Spastin and katanin sever and destabilize microtubules. Paradoxically, despite their destructive activity they increase microtubule mass in vivo. We combined single-molecule total internal reflection fluorescence microscopy and electron microscopy to show that the elemental step in microtubule severing is the generation of nanoscale damage throughout the microtubule by active extraction of tubulin heterodimers. These damage sites are repaired spontaneously by guanosine triphosphate (GTP)-tubulin incorporation, which rejuvenates and stabilizes the microtubule shaft. Consequently, spastin and katanin increase microtubule rescue rates. Furthermore, newly severed ends emerge with a high density of GTP-tubulin that protects them against depolymerization. The stabilization of the newly severed plus ends and the higher rescue frequency synergize to amplify microtubule number and mass. Thus, severing enzymes regulate microtubule architecture and dynamics by promoting GTP-tubulin incorporation within the microtubule shaft.
Collapse
Affiliation(s)
- Annapurna Vemu
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Ewa Szczesna
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Elena A Zehr
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Jeffrey O Spector
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | - Nikolaus Grigorieff
- Howard Hughes Medical Institute, Brandeis University, Waltham, MA 02454, USA
| | - Alexandra M Deaconescu
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02903, USA
| | - Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA.
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD 20892, USA
| |
Collapse
|
33
|
Kaplani K, Koutsi S, Armenis V, Skondra FG, Karantzelis N, Champeris Tsaniras S, Taraviras S. Wound healing related agents: Ongoing research and perspectives. Adv Drug Deliv Rev 2018; 129:242-253. [PMID: 29501699 DOI: 10.1016/j.addr.2018.02.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 01/28/2018] [Accepted: 02/26/2018] [Indexed: 02/07/2023]
Abstract
Wound healing response plays a central part in chronic inflammation, affecting millions of people worldwide. It is a dynamic process that can lead to fibrosis, if tissue damage is irreversible and wound resolution is not attained. It is clear that there is a tight interconnection among wound healing, fibrosis and a variety of chronic disease conditions, demonstrating the heterogeneity of this pathology. Based on our further understanding of the cellular and molecular mechanisms underpinning tissue repair, new therapeutic approaches have recently been developed that target different aspects of the wound healing process and fibrosis. Nevertheless, several issues still need to be taken into consideration when designing modern wound healing drug delivery formulations. In this review, we highlight novel pharmacological agents that hold promise for targeting wound repair and fibrosis. We also focus on drug-delivery systems that may enhance current and future therapies.
Collapse
Affiliation(s)
- Konstantina Kaplani
- Division of Stem Cells and Regenerative Medicine, Biomedical Postgraduate Programme, School of Medicine, University of Patras, Patras 26504, Greece; Department of Physiology, School of Medicine, University of Patras, Patras 26504, Greece
| | - Stamatina Koutsi
- Division of Stem Cells and Regenerative Medicine, Biomedical Postgraduate Programme, School of Medicine, University of Patras, Patras 26504, Greece; Department of Physiology, School of Medicine, University of Patras, Patras 26504, Greece
| | - Vasileios Armenis
- Division of Stem Cells and Regenerative Medicine, Biomedical Postgraduate Programme, School of Medicine, University of Patras, Patras 26504, Greece
| | - Foteini G Skondra
- Division of Stem Cells and Regenerative Medicine, Biomedical Postgraduate Programme, School of Medicine, University of Patras, Patras 26504, Greece
| | - Nickolas Karantzelis
- Department of Physiology, School of Medicine, University of Patras, Patras 26504, Greece
| | | | - Stavros Taraviras
- Division of Stem Cells and Regenerative Medicine, Biomedical Postgraduate Programme, School of Medicine, University of Patras, Patras 26504, Greece; Department of Physiology, School of Medicine, University of Patras, Patras 26504, Greece.
| |
Collapse
|
34
|
Sun J, Wang Z, Wang X. Suppression of LRRC19 promotes cutaneous wound healing in pressure ulcers in mice. Organogenesis 2018; 14:13-24. [PMID: 29461900 DOI: 10.1080/15476278.2018.1436924] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The ischemia-reperfusion (I/R) induced skin lesion has been identified as primary cause of pressure ulcer. Better understanding of the mechanism is required for new therapy development. Leucine rich repeat containing protein 19 (LRRC19) is a recently discovered transmembrane protein containing leucine-rich repeats and plays a role in immune response. To investigate the role of LRRC19 in pressure ulcers, mouse ulcer model was established with two cycles of I/R. The expression of LRRC19 was assessed during injury. siRNA mediated LRRC19 downregulation was applied to investigate the disease severity, immune cell infiltration and pro-inflammatory cytokines production. The primary skin fibroblasts were stimulated with IL-1β to dissect the molecular mechanism. LRRC19 was readily induced in I/R induced lesion site in a pattern mimicking the disease progress as measured by wound area. Knockdown of LRRC19 by siRNA significantly alleviated the disease severity and attenuated immune cell infiltration and pro-inflammatory cytokines production. In primary skin fibroblast model, siRNA knockdown of LRRC19 suppressed IL-1β mediated NFκB activation and its downstream cytokines production. LRRC19 was a novel factor for I/R-induced tissue damage by promoting NFκB dependent pro-inflammatory response. Our results supported that LRRC19 could be a potential therapeutic target for pressure ulcers.
Collapse
Affiliation(s)
- Jie Sun
- a The Third Department of Neurosurgery , Cangzhou Central Hospital , Xinhuaxi Road, Cangzhou City , China
| | - Zhijing Wang
- b Department of Anesthesiology , Cangzhou Central Hospital , Xinhuaxi Road, Cangzhou City , China
| | - Xirui Wang
- a The Third Department of Neurosurgery , Cangzhou Central Hospital , Xinhuaxi Road, Cangzhou City , China
| |
Collapse
|
35
|
Wang Y, Wu B, Lu P, Zhang D, Wu B, Varshney S, Del Monte-Nieto G, Zhuang Z, Charafeddine R, Kramer AH, Sibinga NE, Frangogiannis NG, Kitsis RN, Adams RH, Alitalo K, Sharp DJ, Harvey RP, Stanley P, Zhou B. Uncontrolled angiogenic precursor expansion causes coronary artery anomalies in mice lacking Pofut1. Nat Commun 2017; 8:578. [PMID: 28924218 PMCID: PMC5603578 DOI: 10.1038/s41467-017-00654-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 07/18/2017] [Indexed: 12/13/2022] Open
Abstract
Coronary artery anomalies may cause life-threatening cardiac complications; however, developmental mechanisms underpinning coronary artery formation remain ill-defined. Here we identify an angiogenic cell population for coronary artery formation in mice. Regulated by a DLL4/NOTCH1/VEGFA/VEGFR2 signaling axis, these angiogenic cells generate mature coronary arteries. The NOTCH modulator POFUT1 critically regulates this signaling axis. POFUT1 inactivation disrupts signaling events and results in excessive angiogenic cell proliferation and plexus formation, leading to anomalous coronary arteries, myocardial infarction and heart failure. Simultaneous VEGFR2 inactivation fully rescues these defects. These findings show that dysregulated angiogenic precursors link coronary anomalies to ischemic heart disease.Though coronary arteries are crucial for heart function, the mechanisms guiding their formation are largely unknown. Here, Wang et al. identify a unique, endocardially-derived angiogenic precursor cell population for coronary artery formation in mice and show that a DLL4/NOTCH1/VEGFA/VEGFR2 signaling axis is key for coronary artery development.
Collapse
Affiliation(s)
- Yidong Wang
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Bingruo Wu
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Pengfei Lu
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Donghong Zhang
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Brian Wu
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Shweta Varshney
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Gonzalo Del Monte-Nieto
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, 2010, Australia.,St. Vincent's Clinical School, University of New South Wales, Kensington, NSW, 2052, Australia
| | - Zhenwu Zhuang
- Department of Medicine, Yale University, New Haven, Connecticut, 06510, USA
| | - Rabab Charafeddine
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Adam H Kramer
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Nicolas E Sibinga
- Departments of Medicine, Developmental and Molecular Biology, Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Nikolaos G Frangogiannis
- Departments of Medicine, Microbiology and Immunology, Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Richard N Kitsis
- Departments of Medicine and Cell Biology, Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Röntgenstraße 20, and Faculty of Medicine, University of Münster, 48149, Münster, Germany
| | - Kari Alitalo
- Wihuri Research Institute, Biomedicum Helsinki, Haartmaninkatu 8, FI-00290, Helsinki, Finland
| | - David J Sharp
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Richard P Harvey
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, 2010, Australia.,St. Vincent's Clinical School, University of New South Wales, Kensington, NSW, 2052, Australia.,School of Biotechnology and Biomolecular Science, University of New South Wales, Kensington, NSW, 2052, Australia
| | - Pamela Stanley
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Bin Zhou
- Departments of Genetics, Pediatrics, and Medicine (Cardiology), Wilf Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, 10461, USA. .,Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
36
|
Austin TO, Matamoros AJ, Friedman JM, Friedman AJ, Nacharaju P, Yu W, Sharp DJ, Baas PW. Nanoparticle Delivery of Fidgetin siRNA as a Microtubule-based Therapy to Augment Nerve Regeneration. Sci Rep 2017; 7:9675. [PMID: 28852085 PMCID: PMC5575010 DOI: 10.1038/s41598-017-10250-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/04/2017] [Indexed: 12/28/2022] Open
Abstract
Microtubule-stabilizing drugs have gained popularity for treating injured adult axons, the rationale being that increased stabilization of microtubules will prevent the axon from retracting and fortify it to grow through inhibitory molecules associated with nerve injury. We have posited that a better approach would be not to stabilize the microtubules, but to increase labile microtubule mass to levels more conducive to axonal growth. Recent work on fetal neurons suggests this can be accomplished using RNA interference to reduce the levels of fidgetin, a microtubule-severing protein. Methods to introduce RNA interference into adult neurons, in vitro or in vivo, have been problematic and not translatable to human patients. Here we show that a novel nanoparticle approach, previously shown to deliver siRNA into tissues and organs, enables siRNA to gain entry into adult rat dorsal root ganglion neurons in culture. Knockdown of fidgetin is partial with this approach, but sufficient to increase the labile microtubule mass of the axon, thereby increasing axonal growth. The increase in axonal growth occurs on both a favorable substrate and a growth-inhibitory molecule associated with scar formation in injured spinal cord. The nanoparticles are readily translatable to in vivo studies on animals and ultimately to clinical applications.
Collapse
Affiliation(s)
- Timothy O Austin
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Andrew J Matamoros
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Joel M Friedman
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Adam J Friedman
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.,Department of Dermatology, George Washington School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Parimala Nacharaju
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Wenqian Yu
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - David J Sharp
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Peter W Baas
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA.
| |
Collapse
|
37
|
Huynh CT, Zheng Z, Nguyen MK, McMillan A, Yesilbag Tonga G, Rotello VM, Alsberg E. Cytocompatible Catalyst-Free Photodegradable Hydrogels for Light-Mediated RNA Release To Induce hMSC Osteogenesis. ACS Biomater Sci Eng 2017; 3:2011-2023. [DOI: 10.1021/acsbiomaterials.6b00796] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | | | | | | | - Gulen Yesilbag Tonga
- Department
of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Vincent M. Rotello
- Department
of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | | |
Collapse
|
38
|
Hamdan S, Pastar I, Drakulich S, Dikici E, Tomic-Canic M, Deo S, Daunert S. Nanotechnology-Driven Therapeutic Interventions in Wound Healing: Potential Uses and Applications. ACS CENTRAL SCIENCE 2017; 3:163-175. [PMID: 28386594 PMCID: PMC5364456 DOI: 10.1021/acscentsci.6b00371] [Citation(s) in RCA: 277] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Indexed: 05/09/2023]
Abstract
The chronic nature and associated complications of nonhealing wounds have led to the emergence of nanotechnology-based therapies that aim at facilitating the healing process and ultimately repairing the injured tissue. A number of engineered nanotechnologies have been proposed demonstrating unique properties and multiple functions that address specific problems associated with wound repair mechanisms. In this outlook, we highlight the most recently developed nanotechnology-based therapeutic agents and assess the viability and efficacy of each treatment, with emphasis on chronic cutaneous wounds. Herein we explore the unmet needs and future directions of current technologies, while discussing promising strategies that can advance the wound-healing field.
Collapse
Affiliation(s)
- Suzana Hamdan
- Department of Biochemistry
and Molecular Biology, Miller School of Medicine, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United
States
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program,
Department of Dermatology and Cutaneous Surgery, Miller School of
Medicine, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States
| | - Stefan Drakulich
- Wound Healing and Regenerative Medicine Research Program,
Department of Dermatology and Cutaneous Surgery, Miller School of
Medicine, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States
| | - Emre Dikici
- Department of Biochemistry
and Molecular Biology, Miller School of Medicine, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United
States
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program,
Department of Dermatology and Cutaneous Surgery, Miller School of
Medicine, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States
| | - Sapna Deo
- Department of Biochemistry
and Molecular Biology, Miller School of Medicine, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United
States
| | - Sylvia Daunert
- Department of Biochemistry
and Molecular Biology, Miller School of Medicine, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United
States
- E-mail:
| |
Collapse
|
39
|
Bailey ME, Jiang N, Dima RI, Ross JL. Invited review: Microtubule severing enzymes couple atpase activity with tubulin GTPase spring loading. Biopolymers 2017; 105:547-56. [PMID: 27037673 DOI: 10.1002/bip.22842] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/23/2016] [Accepted: 03/28/2016] [Indexed: 12/21/2022]
Abstract
Microtubules are amazing filaments made of GTPase enzymes that store energy used for their own self-destruction to cause a stochastically driven dynamics called dynamic instability. Dynamic instability can be reproduced in vitro with purified tubulin, but the dynamics do not mimic that observed in cells. This is because stabilizers and destabilizers act to alter microtubule dynamics. One interesting and understudied class of destabilizers consists of the microtubule-severing enzymes from the ATPases Associated with various cellular Activities (AAA+) family of ATP-enzymes. Here we review current knowledge about GTP-driven microtubule dynamics and how that couples to ATP-driven destabilization by severing enzymes. We present a list of challenges regarding the mechanism of severing, which require development of experimental and modeling approaches to shed light as to how severing enzymes can act to regulate microtubule dynamics in cells. © 2016 Wiley Periodicals, Inc. Biopolymers 105: 547-556, 2016.
Collapse
Affiliation(s)
- Megan E Bailey
- Department of Physiology and Biophysics, 1705 NE Pacific St., Seattle, WA 98195
| | - Nan Jiang
- Department of Chemistry, University of Cincinnati, Cincinnati OH 45221
| | - Ruxandra I Dima
- Department of Chemistry, University of Cincinnati, Cincinnati OH 45221
| | - Jennifer L Ross
- Department of Physics, 666 N. Pleasant St. University of Massachusetts, Amherst, MA 01003
| |
Collapse
|
40
|
Abstract
ABSTRACT
Three-dimensional (3D) cell motility underlies essential processes, such as embryonic development, tissue repair and immune surveillance, and is involved in cancer progression. Although the cytoskeleton is a well-studied regulator of cell migration, most of what we know about its functions originates from studies conducted in two-dimensional (2D) cultures. This research established that the microtubule network mediates polarized trafficking and signaling that are crucial for cell shape and movement in 2D. In parallel, developments in light microscopy and 3D cell culture systems progressively allowed to investigate cytoskeletal functions in more physiologically relevant settings. Interestingly, several studies have demonstrated that microtubule involvement in cell morphogenesis and motility can differ in 2D and 3D environments. In this Commentary, we discuss these differences and their relevance for the understanding the role of microtubules in cell migration in vivo. We also provide an overview of microtubule functions that were shown to control cell shape and motility in 3D matrices and discuss how they can be investigated further by using physiologically relevant models.
Collapse
Affiliation(s)
- Benjamin P. Bouchet
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, Utrecht 3584 CH, The Netherlands
| | - Anna Akhmanova
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, Utrecht 3584 CH, The Netherlands
| |
Collapse
|
41
|
Hu Z, Feng J, Bo W, Wu R, Dong Z, Liu Y, Qiang L, Liu M. Fidgetin regulates cultured astrocyte migration by severing tyrosinated microtubules at the leading edge. Mol Biol Cell 2016; 28:545-553. [PMID: 27974640 PMCID: PMC5305261 DOI: 10.1091/mbc.e16-09-0628] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 12/08/2016] [Accepted: 12/09/2016] [Indexed: 12/02/2022] Open
Abstract
Fign regulates cultured astrocyte migration by severing tyrosinated microtubules (MTs). Inhibition of cellular migration induced by Fign knockdown can be rescued with concomitant knockdown of kinesin-12. A working model is given for the MT reconfiguration underlying cellular migration elicited by the cooperation of two distinct MT-related proteins. Microtubule (MT) organization is essential for many cellular events, including mitosis, migration, and cell polarity. Fidgetin (Fign), an ATP-dependent, MT-severing protein, contributes to the regulation of MT configuration by cutting and trimming MT polymers. Functions of Fign have been indicated in neurite outgrowth, mitosis, meiosis, and cellular migration. Here we focus on migration of astrocytes. We find that Fign plays an essential role in cultured astrocyte migration by preferentially targeting MTs (or regions of MTs) that are rich in tyrosinated tubulin, a marker for especially dynamic MTs or especially dynamic regions of MTs. Inhibition of cellular migration induced by Fign knockdown can be rescued with concomitant knockdown of kinesin-12, a motor protein best known for its role in mitosis. We propose a novel working model for MT reconfiguration underlying cellular migration elicited by the functional cooperation of two distinct MT-related proteins.
Collapse
Affiliation(s)
- Zunlu Hu
- Key Laboratory of Neuroregeneration, Jiangsu, and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Jie Feng
- Key Laboratory of Neuroregeneration, Jiangsu, and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Weijuan Bo
- Key Laboratory of Neuroregeneration, Jiangsu, and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Ronghua Wu
- Key Laboratory of Neuroregeneration, Jiangsu, and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Zhangji Dong
- Key Laboratory of Neuroregeneration, Jiangsu, and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Yan Liu
- Key Laboratory of Neuroregeneration, Jiangsu, and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Liang Qiang
- Key Laboratory of Neuroregeneration, Jiangsu, and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China .,Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129
| | - Mei Liu
- Key Laboratory of Neuroregeneration, Jiangsu, and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| |
Collapse
|
42
|
Turner CT, Hasanzadeh Kafshgari M, Melville E, Delalat B, Harding F, Mäkilä E, Salonen JJ, Cowin AJ, Voelcker NH. Delivery of Flightless I siRNA from Porous Silicon Nanoparticles Improves Wound Healing in Mice. ACS Biomater Sci Eng 2016; 2:2339-2346. [DOI: 10.1021/acsbiomaterials.6b00550] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Christopher T. Turner
- Regenerative Medicine, Future
Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Morteza Hasanzadeh Kafshgari
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Future Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Elizabeth Melville
- Regenerative Medicine, Future
Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Bahman Delalat
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Future Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Francis Harding
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Future Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Ermei Mäkilä
- Department
of Physics and Astronomy, University of Turku, FI-20014 Turku, Finland
| | - Jarno J. Salonen
- Department
of Physics and Astronomy, University of Turku, FI-20014 Turku, Finland
| | - Allison J. Cowin
- Regenerative Medicine, Future
Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Nicolas H. Voelcker
- ARC
Centre of Excellence in Convergent Bio-Nano Science and Technology,
Future Industries Institute, University of South Australia, Adelaide, South Australia 5001, Australia
| |
Collapse
|
43
|
|
44
|
Charafeddine RA, Nosanchuk JD, Sharp DJ. Targeting Microtubules for Wound Repair. Adv Wound Care (New Rochelle) 2016; 5:444-454. [PMID: 27785378 DOI: 10.1089/wound.2015.0658] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 09/08/2015] [Indexed: 12/16/2022] Open
Abstract
Significance: Fast and seamless healing is essential for both deep and chronic wounds to restore the skin and protect the body from harmful pathogens. Thus, finding new targets that can both expedite and enhance the repair process without altering the upstream signaling milieu and causing serious side effects can improve the way we treat wounds. Since cell migration is key during the different stages of wound healing, it presents an ideal process and intracellular structural machineries to target. Recent Advances and Critical Issues: The microtubule (MT) cytoskeleton is rising as an important structural and functional regulator of wound healing. MTs have been reported to play different roles in the migration of the various cell types involved in wound healing. Specific microtubule regulatory proteins (MRPs) can be targeted to alter a section or subtype of the MT cytoskeleton and boost or hinder cell motility. However, inhibiting intracellular components can be challenging in vivo, especially using unstable molecules, such as small interfering RNA. Nanoparticles can be used to protect these unstable molecules and topically deliver them to the wound. Utilizing this approach, we recently showed that fidgetin-like 2, an uncharacterized MRP, can be targeted to enhance cell migration and wound healing. Future Directions: To harness the full potential of the current MRP therapeutic targets, studies should test them with different delivery platforms, dosages, and skin models. Screening for new MT effectors that boost cell migration in vivo would also help find new targets for skin repair.
Collapse
Affiliation(s)
- Rabab A. Charafeddine
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, New York
| | - Joshua D. Nosanchuk
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - David J. Sharp
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
45
|
Abstract
The dynamic instability of microtubules is characterised by slow growth phases stochastically interrupted by rapid depolymerisations called catastrophes. Rescue events can arrest the depolymerisation and restore microtubule elongation. However the origin of these rescue events remain unexplained. Here we show that microtubule lattice self-repair, in structurally damaged sites, is responsible for the rescue of microtubule growth. Tubulin photo-conversion in cells revealed that free tubulin dimers can incorporate along the shafts of microtubules, especially in regions where microtubules cross each other, form bundles or become bent due to mechanical constraints. These incorporation sites appeared to act as effective rescue sites ensuring microtubule rejuvenation. By securing damaged microtubule growth, the self-repair process supports a mechanosensitive growth by specifically promoting microtubule assembly in regions where they are subjected to physical constraints.
Collapse
|
46
|
Xiao Y, Ahadian S, Radisic M. Biochemical and Biophysical Cues in Matrix Design for Chronic and Diabetic Wound Treatment. TISSUE ENGINEERING PART B-REVIEWS 2016; 23:9-26. [PMID: 27405960 DOI: 10.1089/ten.teb.2016.0200] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Progress in biomaterial science and engineering and increasing knowledge in cell biology have enabled us to develop functional biomaterials providing appropriate biochemical and biophysical cues for tissue regeneration applications. Tissue regeneration is particularly important to treat chronic wounds of people with diabetes. Understanding and controlling the cellular microenvironment of the wound tissue are important to improve the wound healing process. In this study, we review different biochemical (e.g., growth factors, peptides, DNA, and RNA) and biophysical (e.g., topographical guidance, pressure, electrical stimulation, and pulsed electromagnetic field) cues providing a functional and instructive acellular matrix to heal diabetic chronic wounds. The biochemical and biophysical signals generally regulate cell-matrix interactions and cell behavior and function inducing the tissue regeneration for chronic wounds. Some technologies and devices have already been developed and used in the clinic employing biochemical and biophysical cues for wound healing applications. These technologies can be integrated with smart biomaterials to deliver therapeutic agents to the wound tissue in a precise and controllable manner. This review provides useful guidance in understanding molecular mechanisms and signals in the healing of diabetic chronic wounds and in designing instructive biomaterials to treat them.
Collapse
Affiliation(s)
- Yun Xiao
- 1 Department of Chemical Engineering and Applied Chemistry, University of Toronto , Toronto, Ontario, Canada .,2 Institute of Biomaterials and Biomedical Engineering, University of Toronto , Toronto, Ontario, Canada
| | - Samad Ahadian
- 2 Institute of Biomaterials and Biomedical Engineering, University of Toronto , Toronto, Ontario, Canada
| | - Milica Radisic
- 1 Department of Chemical Engineering and Applied Chemistry, University of Toronto , Toronto, Ontario, Canada .,2 Institute of Biomaterials and Biomedical Engineering, University of Toronto , Toronto, Ontario, Canada
| |
Collapse
|
47
|
DeMarino C, Schwab A, Pleet M, Mathiesen A, Friedman J, El-Hage N, Kashanchi F. Biodegradable Nanoparticles for Delivery of Therapeutics in CNS Infection. J Neuroimmune Pharmacol 2016; 12:31-50. [PMID: 27372507 DOI: 10.1007/s11481-016-9692-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 06/12/2016] [Indexed: 12/18/2022]
Abstract
Despite the significant advances in neurological medicine, it remains difficult to treat ailments directly involving the brain. The blood brain barrier (BBB) is a tightly regulated, selectively permeable barrier that restricts access from the blood into the brain extracellular fluid (BEF). Many conditions such as tumors or infections in the brain are difficult to treat due to the fact that drugs and other therapeutic agents are unable to easily pass through this relatively impermeable barrier. Human Immunodeficiency Virus (HIV) presents a particular problem as it is able to remain dormant in the brain for years protected from antiretroviral drugs by the BBB. The development of nanoscale carriers over the past few decades has made possible the delivery of therapies with the potential to overcome membrane barriers and provide specific, targeted delivery. This review seeks to provide a comprehensive overview of the various aspects of nanoparticle formulation and their applications in improving the delivery efficiency of drugs, specifically antiretroviral therapeutics to the brain to treat HIV.
Collapse
Affiliation(s)
- Catherine DeMarino
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Angela Schwab
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Michelle Pleet
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Allison Mathiesen
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Joel Friedman
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Nazira El-Hage
- Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Fatah Kashanchi
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, USA.
| |
Collapse
|
48
|
Huynh CT, Nguyen MK, Naris M, Tonga GY, Rotello VM, Alsberg E. Light-triggered RNA release and induction of hMSC osteogenesis via photodegradable, dual-crosslinked hydrogels. Nanomedicine (Lond) 2016; 11:1535-50. [PMID: 27246686 PMCID: PMC5827787 DOI: 10.2217/nnm-2016-0088] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 05/04/2016] [Indexed: 12/30/2022] Open
Abstract
AIM To engineer a photodegradable hydrogel system for actively controlled release of bioactive unmodified RNA at designated time points to induce hMSC osteogenesis. MATERIALS & METHODS RNA/polyethylenimine complexes were loaded into dual-crosslinked photodegradable hydrogels to examine the capacity of UV light application to trigger their release. The ability of released RNA to drive hMSC osteogenic differentiation was also investigated. RESULTS & CONCLUSION RNA release from photodegradable hydrogels was accelerated upon UV application, which was not observed in non-photodegradable hydrogels. Regardless of the presence of UV light, released siGFP exhibited high bioactivity by silencing GFP expression in HeLa cells. Importantly, siNoggin or miRNA-20a released from the hydrogels induced hMSC osteogenesis. This system provides a potentially valuable physician/patient-controlled 'on-demand' RNA delivery platform for biomedical applications.
Collapse
Affiliation(s)
- Cong Truc Huynh
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Minh Khanh Nguyen
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Mantas Naris
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Gulen Yesilbag Tonga
- Department of Orthopaedic Surgery, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Vincent M Rotello
- Department of Orthopaedic Surgery, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| |
Collapse
|
49
|
Parani M, Lokhande G, Singh A, Gaharwar AK. Engineered Nanomaterials for Infection Control and Healing Acute and Chronic Wounds. ACS APPLIED MATERIALS & INTERFACES 2016; 8:10049-69. [PMID: 27043006 DOI: 10.1021/acsami.6b00291] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Nanoengineered biomaterials have dramatically expanded the range of tools used for infection control and to accelerate wound healing. This review thoroughly describes the developments that are shaping this emerging field and evaluates the potential wound healing applications of recently developed engineered nanomaterials for both acute and chronic wounds. Specifically, we will assess the unique characteristics of engineered nanomaterials that render them applicable for wound healing and infection control. A range of engineered nanomaterials, including polymeric-, metallic- and ceramic-based nanomaterials, that could be used as therapeutic delivery agents to accelerate regeneration of damaged dermal and epidermal tissues are also detailed. Finally, we will detail the current state of engineered nanomaterials for wound regeneration and will identify promising new research directions in infection control.
Collapse
Affiliation(s)
- Madasamy Parani
- Genomics Laboratory, Department of Genetic Engineering, SRM University , Chennai, Tamil Nadu 603 203, India
| | | | - Ankur Singh
- Sibley School of Mechanical and Aerospace Engineering, Cornell University , Ithaca, New York 14853, United States
| | | |
Collapse
|
50
|
Abstract
Complex spatiotemporal interaction of Rho GTPases with microtubules (MTs) and MT-associated proteins drives directed cellular migration. In this issue, Charafeddine et al. describe a role for a novel MT-severing enzyme, fidgetin-like 2 (FL2), in directional migration of keratinocytes and fibroblasts. FL2 normally localizes to the leading edge of the cell cortex where it shears MTs, thus dictating the size and distribution of focal adhesions by regulating cytoskeletal remodeling. Small interfering RNA (siRNA)-directed knockdown of FL2 increases cell migration and focal adhesion area in vitro through possible interaction with Rho GTPases. Efficient FL2 knockdown in murine wounds was achieved using nanoparticles as a siRNA delivery vehicle, and this resulted in enhanced wound closure in vivo. Effective siRNA nanoparticle targeting of MT-severing enzymes offers promise of controlled and targeted delivery that may maximize therapeutic success for patients with burn wounds and chronic wound disorders.
Collapse
|