1
|
Peña OA, Martin P. Cellular and molecular mechanisms of skin wound healing. Nat Rev Mol Cell Biol 2024; 25:599-616. [PMID: 38528155 DOI: 10.1038/s41580-024-00715-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2024] [Indexed: 03/27/2024]
Abstract
Wound healing is a complex process that involves the coordinated actions of many different tissues and cell lineages. It requires tight orchestration of cell migration, proliferation, matrix deposition and remodelling, alongside inflammation and angiogenesis. Whereas small skin wounds heal in days, larger injuries resulting from trauma, acute illness or major surgery can take several weeks to heal, generally leaving behind a fibrotic scar that can impact tissue function. Development of therapeutics to prevent scarring and successfully repair chronic wounds requires a fuller knowledge of the cellular and molecular mechanisms driving wound healing. In this Review, we discuss the current understanding of the different phases of wound healing, from clot formation through re-epithelialization, angiogenesis and subsequent scar deposition. We highlight the contribution of different cell types to skin repair, with emphasis on how both innate and adaptive immune cells in the wound inflammatory response influence classically studied wound cell lineages, including keratinocytes, fibroblasts and endothelial cells, but also some of the less-studied cell lineages such as adipocytes, melanocytes and cutaneous nerves. Finally, we discuss newer approaches and research directions that have the potential to further our understanding of the mechanisms underpinning tissue repair.
Collapse
Affiliation(s)
- Oscar A Peña
- School of Biochemistry, University of Bristol, Bristol, UK.
| | - Paul Martin
- School of Biochemistry, University of Bristol, Bristol, UK.
| |
Collapse
|
2
|
Yu D, Lu Z, Chong Y. Integrins as a bridge between bacteria and cells: key targets for therapeutic wound healing. BURNS & TRAUMA 2024; 12:tkae022. [PMID: 39015251 PMCID: PMC11250365 DOI: 10.1093/burnst/tkae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 12/17/2023] [Accepted: 04/22/2024] [Indexed: 07/18/2024]
Abstract
Integrins are heterodimers composed of α and β subunits that are bonded through non-covalent interactions. Integrins mediate the dynamic connection between extracellular adhesion molecules and the intracellular actin cytoskeleton. Integrins are present in various tissues and organs where these heterodimers participate in diverse physiological and pathological responses at the molecular level in living organisms. Wound healing is a crucial process in the recovery from traumatic diseases and comprises three overlapping phases: inflammation, proliferation and remodeling. Integrins are regulated during the entire wound healing process to enhance processes such as inflammation, angiogenesis and re-epithelialization. Prolonged inflammation may result in failure of wound healing, leading to conditions such as chronic wounds. Bacterial colonization of a wound is one of the primary causes of chronic wounds. Integrins facilitate the infectious effects of bacteria on the host organism, leading to chronic inflammation, bacterial colonization, and ultimately, the failure of wound healing. The present study investigated the role of integrins as bridges for bacteria-cell interactions during wound healing, evaluated the role of integrins as nodes for bacterial inhibition during chronic wound formation, and discussed the challenges and prospects of using integrins as therapeutic targets in wound healing.
Collapse
Affiliation(s)
- Dong Yu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Middle Road, Yangzhou 225000, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Middle Road, Yangzhou 225000, Jiangsu, China
| | - Zhaoyu Lu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Middle Road, Yangzhou 225000, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Middle Road, Yangzhou 225000, Jiangsu, China
| | - Yang Chong
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Middle Road, Yangzhou 225000, Jiangsu, China
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368 Hanjiang Middle Road, Yangzhou 225000, Jiangsu, China
| |
Collapse
|
3
|
Takada YK, Yu J, Ye X, Wu CY, Felding BH, Fujita M, Takada Y. The heparin-binding domain of VEGF165 directly binds to integrin αvβ3 and VEGFR2/KDR D1: a potential mechanism of negative regulation of VEGF165 signaling by αvβ3. Front Cell Dev Biol 2024; 12:1347616. [PMID: 38803393 PMCID: PMC11128890 DOI: 10.3389/fcell.2024.1347616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/04/2024] [Indexed: 05/29/2024] Open
Abstract
VEGF-A is a key cytokine in tumor angiogenesis and a major therapeutic target for cancer. VEGF165 is the predominant isoform of VEGF-A, and it is the most potent angiogenesis stimulant. VEGFR2/KDR domains 2 and 3 (D2D3) bind to the N-terminal domain (NTD, residues 1-110) of VEGF165. Since removal of the heparin-binding domain (HBD, residues 111-165) markedly reduced the mitogenic activity of the growth factor, it has been proposed that the HBD plays a critical role in the mitogenicity of VEGF165. Here, we report that αvβ3 specifically bound to the isolated VEGF165 HBD but not to VEGF165 NTD. Based on docking simulation and mutagenesis, we identified several critical amino acid residues within the VEGF165 HBD required for αvβ3 binding, i.e., Arg123, Arg124, Lys125, Lys140, Arg145, and Arg149. We discovered that VEGF165 HBD binds to the KDR domain 1 (D1) and identified that Arg123 and Arg124 are critical for KDR D1 binding by mutagenesis, indicating that the KDR D1-binding and αvβ3-binding sites overlap in the HBD. Full-length VEGF165 mutant (R123A/R124A/K125A/K140A/R145A/R149A) defective in αvβ3 and KDR D1 binding failed to induce ERK1/2 phosphorylation, integrin β3 phosphorylation, and KDR phosphorylation and did not support proliferation of endothelial cells, although the mutation did not affect the KDR D2D3 interaction with VEGF165. Since β3-knockout mice are known to show enhanced VEGF165 signaling, we propose that the binding of KDR D1 to the VEGF165 HBD and KDR D2D3 binding to the VEGF165 NTD are critically involved in the potent mitogenicity of VEGF165. We propose that binding competition between KDR and αvβ3 to the VEGF165 HBD endows integrin αvβ3 with regulatory properties to act as a negative regulator of VEGF165 signaling.
Collapse
Affiliation(s)
- Yoko K. Takada
- The Department of Dermatology, Sacramento, CA, United States
| | - Jessica Yu
- The Department of Dermatology, Sacramento, CA, United States
| | - Xiaojin Ye
- The Department of Dermatology, Sacramento, CA, United States
| | - Chun-Yi Wu
- The Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Brunie H. Felding
- The Scripps Research Institute, Department of Molecular and Experimental Medicine, La Jolla, CA, United States
| | - Masaaki Fujita
- The Department of Dermatology, Sacramento, CA, United States
| | - Yoshikazu Takada
- The Department of Dermatology, Sacramento, CA, United States
- The Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
4
|
Wang Y, Pan Z, Cui J, Zhang X, Li D, Sun H, Yang B, Li Y. Adhesive hydrogel releases protocatechualdehyde-Fe 3+ complex to promote three healing stages for accelerated therapy of oral ulcers. Acta Biomater 2024; 178:68-82. [PMID: 38452962 DOI: 10.1016/j.actbio.2024.02.047] [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: 12/08/2023] [Revised: 02/25/2024] [Accepted: 02/28/2024] [Indexed: 03/09/2024]
Abstract
Oral ulcers can significantly reduce the life quality of patients and even lead to malignant transformations. Local treatments using topical agents are often ineffective because of the wet and dynamic environment of the oral cavity. Current clinical treatments for oral ulcers, such as corticosteroids, have limitations and side effects for long-term usage. Here, we develop adhesive hydrogel patches (AHPs) that effectively promote the healing of oral ulcers in a rat model. The AHPs are comprised of the quaternary ammonium salt of chitosan, aldehyde-functionalized hyaluronic acid, and a tridentate complex of protocatechualdehyde and Fe3+ (PF). The AHPs exhibit tunable mechanical properties, self-healing ability, and wet adhesion on the oral mucosa. Through controlling the formula of the AHPs, PF released from the AHPs in a temporal manner. We further show that the AHPs have good biocompatibility and the capability to heal oral ulcers rapidly. Both in vitro and in vivo experiments indicate that the PF released from AHPs facilitated ulcer healing by suppressing inflammation, promoting macrophage polarization, enhancing cell proliferation, and inducing epithelial-mesenchymal transition involving inflammation, proliferation, and maturation stages. This study provides insights into the healing of oral ulcers and presents an effective therapeutic biomaterial for the treatment of oral ulcers. STATEMENT OF SIGNIFICANCE: By addressing the challenges associated with current clinical treatments for oral ulcers, the development of adhesive hydrogel patches (AHPs) presents an effective approach. These AHPs possess unique properties, such as tunable mechanical characteristics, self-healing ability, and strong adhesion to the mucosa. Through controlled release of protocatechualdehyde-Fe3+ complex, the AHPs facilitate the healing process by suppressing inflammation, promoting cell proliferation, and inducing epithelial-mesenchymal transition. The study not only provides valuable insights into the healing mechanisms of oral ulcers but also introduces a promising therapeutic biomaterial. This work holds significant scientific interest and demonstrates the potential to greatly improve the treatment outcomes and quality of life for individuals suffering from oral ulcers.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, China
| | - Ziyi Pan
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, China
| | - Jing Cui
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, China
| | - Xu Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, China
| | - Daowei Li
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, China.
| | - Hongchen Sun
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, China
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, China; Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, China
| | - Yunfeng Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, China; Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, China.
| |
Collapse
|
5
|
El-Sayed AFM, Khaled AA, Hamdan AM, Makled SO, Hafez EE, Saleh AA. The role of antifreeze genes in the tolerance of cold stress in the Nile tilapia (Oreochromis niloticus). BMC Genomics 2023; 24:476. [PMID: 37612592 PMCID: PMC10464439 DOI: 10.1186/s12864-023-09569-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/09/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Tilapia is one of the most essential farmed fishes in the world. It is a tropical and subtropical freshwater fish well adapted to warm water but sensitive to cold weather. Extreme cold weather could cause severe stress and mass mortalities in tilapia. The present study was carried out to investigate the effects of cold stress on the up-regulation of antifreeze protein (AFP) genes in Nile tilapia (Oreochromis niloticus). Two treatment groups of fish were investigated (5 replicates of 15 fish for each group in fibreglass tanks/70 L each): 1) a control group; the fish were acclimated to lab conditions for two weeks and the water temperature was maintained at 25 °C during the whole experimental period with feeding on a commercial diet (30% crude protein). 2) Cold stress group; the same conditions as the control group except for the temperature. Initially, the temperature was decreased by one degree every 12 h. The fish started showing death symptoms when the water temperature reached 6-8 °C. In this stage the tissue (muscle) samples were taken from both groups. The immune response of fish exposed to cold stress was detected and characterized using Differential Display-PCR (DD-PCR). RESULTS The results indicated that nine different up-regulation genes were detected in the cold-stressed fish compared to the control group. These genes are Integrin-alpha-2 (ITGA-2), Gap junction gamma-1 protein-like (GJC1), WD repeat-containing protein 59 isoform X2 (WDRP59), NUAK family SNF1-like kinase, G-protein coupled receptor-176 (GPR-176), Actin cytoskeleton-regulatory complex protein pan1-like (PAN-1), Whirlin protein (WHRN), Suppressor of tumorigenicity 7 protein isoform X2 (ST7P) and ATP-binding cassette sub-family A member 1-like isoform X2 (ABCA1). The antifreeze gene type-II amplification using a specific PCR product of 600 bp, followed by cloning and sequencing analysis revealed that the identified gene is antifreeze type-II, with similarity ranging from 70 to 95%. The in-vitro transcribed gene induced an antifreeze protein with a molecular size of 22 kDa. The antifreeze gene, ITGA-2 and the WD repeat protein belong to the lectin family (sugar-protein). CONCLUSIONS In conclusion, under cold stress, Nile tilapia express many defence genes, an antifreeze gene consisting of one open reading frame of approximately 0.6 kbp.
Collapse
Affiliation(s)
| | - Asmaa A Khaled
- Animal and Fish Production Department, Faculty of Agriculture (Saba Basha), Alexandria University, Alexandria City, 21531, Egypt
| | - Amira M Hamdan
- Oceanography Department, Faculty of Science, Alexandria University, Alexandria City, Egypt
| | - Sara O Makled
- Oceanography Department, Faculty of Science, Alexandria University, Alexandria City, Egypt
| | - Elsayed E Hafez
- Arid Lands Cultivation Research Institute, City of Scientific Research and Technological Applications, New Borg El Arab, Alexandria City, 21934, Egypt
| | - Ahmed A Saleh
- Animal and Fish Production Department, Faculty of Agriculture (Alshatby), Alexandria University, Alexandria City, 11865, Egypt.
| |
Collapse
|
6
|
Pensalfini M, Tepole AB. Mechano-biological and bio-mechanical pathways in cutaneous wound healing. PLoS Comput Biol 2023; 19:e1010902. [PMID: 36893170 PMCID: PMC10030043 DOI: 10.1371/journal.pcbi.1010902] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 03/21/2023] [Accepted: 01/27/2023] [Indexed: 03/10/2023] Open
Abstract
Injuries to the skin heal through coordinated action of fibroblast-mediated extracellular matrix (ECM) deposition, ECM remodeling, and wound contraction. Defects involving the dermis result in fibrotic scars featuring increased stiffness and altered collagen content and organization. Although computational models are crucial to unravel the underlying biochemical and biophysical mechanisms, simulations of the evolving wound biomechanics are seldom benchmarked against measurements. Here, we leverage recent quantifications of local tissue stiffness in murine wounds to refine a previously-proposed systems-mechanobiological finite-element model. Fibroblasts are considered as the main cell type involved in ECM remodeling and wound contraction. Tissue rebuilding is coordinated by the release and diffusion of a cytokine wave, e.g. TGF-β, itself developed in response to an earlier inflammatory signal triggered by platelet aggregation. We calibrate a model of the evolving wound biomechanics through a custom-developed hierarchical Bayesian inverse analysis procedure. Further calibration is based on published biochemical and morphological murine wound healing data over a 21-day healing period. The calibrated model recapitulates the temporal evolution of: inflammatory signal, fibroblast infiltration, collagen buildup, and wound contraction. Moreover, it enables in silico hypothesis testing, which we explore by: (i) quantifying the alteration of wound contraction profiles corresponding to the measured variability in local wound stiffness; (ii) proposing alternative constitutive links connecting the dynamics of the biochemical fields to the evolving mechanical properties; (iii) discussing the plausibility of a stretch- vs. stiffness-mediated mechanobiological coupling. Ultimately, our model challenges the current understanding of wound biomechanics and mechanobiology, beside offering a versatile tool to explore and eventually control scar fibrosis after injury.
Collapse
Affiliation(s)
- Marco Pensalfini
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana, United States of America
- Institute for Mechanical Systems (IMES), Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
- Laboratori de Càlcul Numèric (LaCàN), Universitat Politècnica de Catalunya-BarcelonaTech, Barcelona, Spain
| | - Adrian Buganza Tepole
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana, United States of America
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
| |
Collapse
|
7
|
Yun J, Park S, Park HY, Lee KA. Efficacy of Polydeoxyribonucleotide in Promoting the Healing of Diabetic Wounds in a Murine Model of Streptozotocin-Induced Diabetes: A Pilot Experiment. Int J Mol Sci 2023; 24:ijms24031932. [PMID: 36768255 PMCID: PMC9916466 DOI: 10.3390/ijms24031932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/20/2023] Open
Abstract
We assessed the efficacy of polydeoxyribonucleotide (PDRN) in accelerating the healing of diabetic wounds in a murine model of streptozotocin (STZ)-induced diabetes. After the creation of diabetic wounds, the mice of the PDRN SC, PDRN IP and PBS groups received a subcutaneous, an intra-peritoneal injection of PDRN and a subcutaneous injection of PBS, respectively. After euthanasia, time-dependent changes in the wound diameter and histologic scores were measured and vascular endothelial growth factor (VEGF), transforming growth factor-β1 (TGF-β1) and collagen types I and III were assessed for their expression levels. The PDRN SC and the PDRN IP groups showed a significantly smaller diameter of diabetic wounds, significantly higher histologic scores, a significantly greater expression of VEGF, a significantly lower expression of TGF-β1 and a significantly greater expression of collagen types I and III as compared with the PBS group (p < 0.05 or 0.0001). In conclusion, PDRN might be effective in promoting the healing of diabetic wounds in a murine model of STZ-induced diabetes.
Collapse
Affiliation(s)
- Jiyoung Yun
- Department of Plastic and Reconstructive Surgery, Inje University Busan Paik Hospital, College of Medicine, Inje University, Busan 47392, Republic of Korea
| | - SaeGwang Park
- Department of Microbiology and Immunology, College of Medicine, Inje University, Busan 47392, Republic of Korea
- Innovative Therapeutic Research Institute, Inje University Busan Paik Hospital, College of Medicine, Inje University, Busan 47392, Republic of Korea
| | - Ha Young Park
- Department of Pathology, Inje University Busan Paik Hospital, College of Medicine, Inje University, Busan 47392, Republic of Korea
| | - Kyung Ah Lee
- Department of Plastic and Reconstructive Surgery, Inje University Haeundae Paik Hospital, College of Medicine, Inje University, Busan 48108, Republic of Korea
- Correspondence:
| |
Collapse
|
8
|
Fu YN, Li Y, Deng B, Yu Y, Liu F, Wang L, Chen G, Tao L, Wei Y, Wang X. Spatiotemporally dynamic therapy with shape-adaptive drug-gel for the improvement of tissue regeneration with ordered structure. Bioact Mater 2022; 8:165-176. [PMID: 34541394 PMCID: PMC8424390 DOI: 10.1016/j.bioactmat.2021.06.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/08/2021] [Accepted: 06/11/2021] [Indexed: 12/30/2022] Open
Abstract
A spatiotemporally dynamic therapy (SDT) is proposed as a powerful therapeutic modality that provides spatially dynamic responses of drug-carriers for adapting to the wound microenvironment. Herein, dynamic chitosan-poly (ethylene glycol) (CP) Schiff-base linkages are employed to perform SDT by directly converting a liquid drug Kangfuxin (KFX) into a gel formation. The obtained KFX-CP drug-gel with shape-adaptive property is used to treat a representative oral mucositis (OM) model in a spatiotemporally dynamic manner. The KFX-CP drug-gel creates an instructive microenvironment to regulate signaling biomolecules and endogenous cells behavior, thereby promoting OM healing by the rule of dynamically adjusting shape to fit the irregular OM regions first, and then provides space for tissue regeneration, over KFX potion control and the general hydrogel group of CP hydrogel and KFX-F127. Most interestingly, the regenerated tissue has ordered structure like healthy tissue. Therefore, the SDT provides a new approach for the design of next generation of wound dressing and tissue engineering materials.
Collapse
Affiliation(s)
- Ya-nan Fu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yongsan Li
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
- The Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Bo Deng
- Department of Oncology of Integrative Chinese and Western Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yingjie Yu
- Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen, 518035, China
| | - Fang Liu
- Department of Oncology of Integrative Chinese and Western Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Lei Wang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Guang Chen
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Lei Tao
- The Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Yen Wei
- The Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Xing Wang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
9
|
Zhang Y, Wang Y, Zeng L, Liu Y, Sun H, Li S, Wang S, Shu L, Liu N, Yin S, Wang J, Ni D, Wu Y, Yang Y, He L, Meng B, Yang X. Amphibian-derived peptide homodimer OA-GL17d promotes skin wound regeneration through the miR-663a/TGF-β1/Smad axis. BURNS & TRAUMA 2022; 10:tkac032. [PMID: 35832307 PMCID: PMC9273405 DOI: 10.1093/burnst/tkac032] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/08/2022] [Indexed: 11/29/2022]
Abstract
Background Amphibian-derived peptides exhibit considerable potential in the discovery and development of new therapeutic interventions for clinically challenging chronic skin wounds. MicroRNAs (miRNAs) are also considered promising targets for the development of effective therapies against skin wounds. However, further research in this field is anticipated. This study aims to identify and provide a new peptide drug candidate, as well as to explore the underlying miRNA mechanisms and possible miRNA drug target for skin wound healing. Methods A combination of Edman degradation, mass spectrometry and cDNA cloning were adopted to determine the amino acid sequence of a peptide that was fractionated from the secretion of Odorrana andersonii frog skin using gel-filtration and reversed-phase high-performance liquid chromatography. The toxicity of the peptide was evaluated by Calcein-AM/propidium iodide (PI) double staining against human keratinocytes (HaCaT cells), hemolytic activity against mice blood cells and acute toxicity against mice. The stability of the peptide in plasma was also evaluated. The prohealing potency of the peptide was determined by MTS, scratch healing and a Transwell experiment against HaCaT cells, full-thickness injury wounds and scald wounds in the dorsal skin of mice. miRNA transcriptome sequencing analysis, enzyme-linked immunosorbent assay, real-time polymerase chain reaction and western blotting were performed to explore the molecular mechanisms. Results A novel peptide homodimer (named OA-GL17d) that contains a disulfide bond between the 16th cysteine residue of the peptide monomer and the sequence ‘GLFKWHPRCGEEQSMWT’ was identified. Analysis showed that OA-GL17d exhibited no hemolytic activity or acute toxicity, but effectively promoted keratinocyte proliferation and migration and strongly stimulated the repair of full-thickness injury wounds and scald wounds in the dorsal skin of mice. Mechanistically, OA-GL17d decreased the level of miR-663a to increase the level of transforming growth factor-β1 (TGF-β1) and activate the subsequent TGF-β1/Smad signaling pathway, thereby resulting in accelerated skin wound re-epithelialization and granular tissue formation. Conclusions Our results suggest that OA-GL17d is a new peptide drug candidate for skin wound repair. This study emphasizes the importance of exogenous peptides as molecular probes for exploring competing endogenous RNA mechanisms and indicates that miR-663a may be an effective target for promoting skin repair.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Anatomy and Histology & Embryology , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
- Kunming Medical University , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
| | - Ying Wang
- Key Laboratory of Chemistry in Ethnic Medicinal Resources & Key Laboratory of Natural Products Synthetic Biology of Ethnic Medicinal Endophytes , State Ethnic Affairs Commission & Ministry of Education, School of Ethnomedicine and Ethnopharmacy, , Kunming 650504, Yunnan, China
- Yunnan MinZu University , State Ethnic Affairs Commission & Ministry of Education, School of Ethnomedicine and Ethnopharmacy, , Kunming 650504, Yunnan, China
| | - Lin Zeng
- Institutional Center for Shared Technologies and Facilities of Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming 650223, Yunnan, China
| | - Yixiang Liu
- Key Laboratory of Chemistry in Ethnic Medicinal Resources & Key Laboratory of Natural Products Synthetic Biology of Ethnic Medicinal Endophytes , State Ethnic Affairs Commission & Ministry of Education, School of Ethnomedicine and Ethnopharmacy, , Kunming 650504, Yunnan, China
- Yunnan MinZu University , State Ethnic Affairs Commission & Ministry of Education, School of Ethnomedicine and Ethnopharmacy, , Kunming 650504, Yunnan, China
| | - Huiling Sun
- Department of Anatomy and Histology & Embryology , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
- Kunming Medical University , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
| | - Shanshan Li
- Department of Anatomy and Histology & Embryology , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
- Kunming Medical University , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
| | - Siyu Wang
- Department of Anatomy and Histology & Embryology , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
- Kunming Medical University , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
| | - Longjun Shu
- Key Laboratory of Chemistry in Ethnic Medicinal Resources & Key Laboratory of Natural Products Synthetic Biology of Ethnic Medicinal Endophytes , State Ethnic Affairs Commission & Ministry of Education, School of Ethnomedicine and Ethnopharmacy, , Kunming 650504, Yunnan, China
- Yunnan MinZu University , State Ethnic Affairs Commission & Ministry of Education, School of Ethnomedicine and Ethnopharmacy, , Kunming 650504, Yunnan, China
| | - Naixin Liu
- Department of Anatomy and Histology & Embryology , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
- Kunming Medical University , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
| | - Saige Yin
- Department of Anatomy and Histology & Embryology , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
- Kunming Medical University , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
| | - Junsong Wang
- Department of Anatomy and Histology & Embryology , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
- Kunming Medical University , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
| | - Dan Ni
- Department of Anatomy and Histology & Embryology , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
- Kunming Medical University , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
| | - Yutong Wu
- Department of Anatomy and Histology & Embryology , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
- Kunming Medical University , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
| | - Ying Yang
- Endocrinology Department of Affiliated Hospital of Yunnan University , Kunming 650021, Yunnan, China
| | - Li He
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University , Kunming, 650500, Yunnan, China
| | - Buliang Meng
- Department of Anatomy and Histology & Embryology , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
- Kunming Medical University , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
| | - Xinwang Yang
- Department of Anatomy and Histology & Embryology , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
- Kunming Medical University , Faculty of Basic Medical Science, , Kunming 650500, Yunnan, China
| |
Collapse
|
10
|
Cationic, anionic and neutral polysaccharides for skin tissue engineering and wound healing applications. Int J Biol Macromol 2021; 192:298-322. [PMID: 34634326 DOI: 10.1016/j.ijbiomac.2021.10.013] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/25/2021] [Accepted: 10/03/2021] [Indexed: 12/17/2022]
Abstract
Today, chronic wound care and management can be regarded as a clinically critical issue. However, the limitations of current approaches for wound healing have encouraged researchers and physicians to develop more efficient alternative approaches. Advances in tissue engineering and regenerative medicine have resulted in the development of promising approaches that can accelerate wound healing and improve the skin regeneration rate and quality. The design and fabrication of scaffolds that can address the multifactorial nature of chronic wound occurrence and provide support for the healing process can be considered an important area requiring improvement. In this regard, polysaccharide-based scaffolds have distinctive properties such as biocompatibility, biodegradability, high water retention capacity and nontoxicity, making them ideal for wound healing applications. Their tunable structure and networked morphology could facilitate a number of functions, such as controlling their diffusion, maintaining wound moisture, absorbing a large amount of exudates and facilitating gas exchange. In this review, the wound healing process and the influential factors, structure and properties of carbohydrate polymers, physical and chemical crosslinking of polysaccharides, scaffold fabrication techniques, and the use of polysaccharide-based scaffolds in skin tissue engineering and wound healing applications are discussed.
Collapse
|
11
|
Kearney KJ, Ariëns RAS, Macrae FL. The Role of Fibrin(ogen) in Wound Healing and Infection Control. Semin Thromb Hemost 2021; 48:174-187. [PMID: 34428799 DOI: 10.1055/s-0041-1732467] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Fibrinogen, one of the most abundant plasma proteins playing a key role in hemostasis, is an important modulator of wound healing and host defense against microbes. In the current review, we address the role of fibrin(ogen) throughout the process of wound healing and subsequent tissue repair. Initially fibrin(ogen) acts as a provisional matrix supporting incoming leukocytes and acting as reservoir for growth factors. It later goes on to support re-epithelialization, angiogenesis, and fibroplasia. Importantly, removal of fibrin(ogen) from the wound is essential for wound healing to progress. We also discuss how fibrin(ogen) functions through several mechanisms to protect the host against bacterial infection by providing a physical barrier, entrapment of bacteria in fibrin(ogen) networks, and by directing immune cell function. The central role of fibrin(ogen) in defense against bacterial infection has made it a target of bacterial proteins, evolved to interact with fibrin(ogen) to manipulate clot formation and degradation for the purpose of promoting microbial virulence and survival. Further understanding of the dual roles of fibrin(ogen) in wound healing and infection could provide novel means of therapy to improve recovery from surgical or chronic wounds and help to prevent infection from highly virulent bacterial strains, including those resistant to antibiotics.
Collapse
Affiliation(s)
- Katherine J Kearney
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Robert A S Ariëns
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom.,Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Fraser L Macrae
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
12
|
Picoli CC, Gonçalves BÔP, Santos GSP, Rocha BGS, Costa AC, Resende RR, Birbrair A. Pericytes cross-talks within the tumor microenvironment. Biochim Biophys Acta Rev Cancer 2021; 1876:188608. [PMID: 34384850 DOI: 10.1016/j.bbcan.2021.188608] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 07/14/2021] [Accepted: 08/05/2021] [Indexed: 02/07/2023]
Abstract
Cancer cells are embedded within the tumor microenvironment and interact dynamically with its components during tumor progression. Understanding the molecular mechanisms by which the tumor microenvironment components communicate is crucial for the success of therapeutic applications. Recent studies show, by using state-of-the-art technologies, including sophisticated in vivo inducible Cre/loxP mediated systems and CRISPR-Cas9 gene editing, that pericytes communicate with cancer cells. The arising knowledge on cross-talks within the tumor microenvironment will be essential for the development of new therapies against cancer. Here, we review recent progress in our understanding of pericytes roles within tumors.
Collapse
Affiliation(s)
- Caroline C Picoli
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Bryan Ô P Gonçalves
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gabryella S P Santos
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Beatriz G S Rocha
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alinne C Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rodrigo R Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
13
|
Cui H, Liu M, Yu W, Cao Y, Zhou H, Yin J, Liu H, Que S, Wang J, Huang C, Gong C, Zhao G. Copper Peroxide-Loaded Gelatin Sponges for Wound Dressings with Antimicrobial and Accelerating Healing Properties. ACS APPLIED MATERIALS & INTERFACES 2021; 13:26800-26807. [PMID: 34096255 DOI: 10.1021/acsami.1c07409] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Catalytic conversion of hydrogen peroxide (H2O2) to more toxic hydroxyl radicals (•OH) is a good choice for sterilization and anti-infection, but endogenous H2O2 is insufficient to achieve satisfactory sterilization efficacy. Despite great efforts, designing and developing antimicrobial materials that specifically and effectively self-supply H2O2 at the wound site remain as tremendous challenges. Here, we report a pH-responsive copper peroxide-loaded wound dressing made from copper hydroxide and gelatin sponge and then reacted with H2O2. In vitro experiments show that the prepared wound dressing has good bactericidal properties against Escherichia coli (E. coli), Staphylococcus aureus (S. aureus), and Pseudomonas aeruginosa (P. aeruginosa). Moreover, the as-prepared wound dressing can release •OH specifically in the bacterial-infected skin wound, rather than in normal tissues, and in vivo skin wound-healing experiments proved that the synthesized copper peroxide-loaded gelatin sponge could combat E. coli effectively; in addition, Cu2+ released from the gelatin sponge could stimulate angiogenesis and collagen deposition simultaneously. The study provides a strategy to improve antibacterial efficacy and reduce the toxic side effects through the release of •OH by bacterial self-activation.
Collapse
Affiliation(s)
- Haiyan Cui
- State Key Laboratory of Applied Organic Chemistry, Institute of Biochemical Engineering & Environmental Technology, College of Chemistry and Chemical Engineering, Lanzhou University, 222 Tianshui Road, Lanzhou 730000, P. R. China
- State Key Laboratory for Marine Corrosion and Protection, Luoyang Ship Material Research Institute (LSMRI), Xiamen 361101, P. R. China
| | - Mingsheng Liu
- State Key Laboratory of Applied Organic Chemistry, Institute of Biochemical Engineering & Environmental Technology, College of Chemistry and Chemical Engineering, Lanzhou University, 222 Tianshui Road, Lanzhou 730000, P. R. China
| | - Wenwen Yu
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, P. R. China
| | - Yufei Cao
- State Key Laboratory of Applied Organic Chemistry, Institute of Biochemical Engineering & Environmental Technology, College of Chemistry and Chemical Engineering, Lanzhou University, 222 Tianshui Road, Lanzhou 730000, P. R. China
| | - Haicun Zhou
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, P. R. China
| | - Juanjuan Yin
- State Key Laboratory of Applied Organic Chemistry, Institute of Biochemical Engineering & Environmental Technology, College of Chemistry and Chemical Engineering, Lanzhou University, 222 Tianshui Road, Lanzhou 730000, P. R. China
| | - Hongbin Liu
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, P. R. China
| | - Sheng Que
- Department of Chemistry, Minorities Teachers College, Qinghai Normal University, Xining 810008, P. R. China
| | - Jingjing Wang
- State Key Laboratory for Marine Corrosion and Protection, Luoyang Ship Material Research Institute (LSMRI), Xiamen 361101, P. R. China
| | - Congshu Huang
- State Key Laboratory for Marine Corrosion and Protection, Luoyang Ship Material Research Institute (LSMRI), Xiamen 361101, P. R. China
| | - Chenliang Gong
- State Key Laboratory of Applied Organic Chemistry, Institute of Biochemical Engineering & Environmental Technology, College of Chemistry and Chemical Engineering, Lanzhou University, 222 Tianshui Road, Lanzhou 730000, P. R. China
| | - Guanghui Zhao
- State Key Laboratory of Applied Organic Chemistry, Institute of Biochemical Engineering & Environmental Technology, College of Chemistry and Chemical Engineering, Lanzhou University, 222 Tianshui Road, Lanzhou 730000, P. R. China
| |
Collapse
|
14
|
Zhang Z, Li Z, Li Y, Wang Y, Yao M, Zhang K, Chen Z, Yue H, Shi J, Guan F, Ma S. Sodium alginate/collagen hydrogel loaded with human umbilical cord mesenchymal stem cells promotes wound healing and skin remodeling. Cell Tissue Res 2021; 383:809-821. [PMID: 33159581 DOI: 10.1007/s00441-020-03321-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 10/12/2020] [Indexed: 02/03/2023]
Abstract
Stem cell transplantation is a promising therapy for wound healing, but the low retention and survival of transplanted stem cells limit their application. Injectable hydrogels exert beneficial effects in skin tissue engineering. In this study, an injectable hydrogel composed of sodium alginate (SA) and collagen type I (Col) was synthesized as a tissue scaffold to improve the efficacy of stem cells in a full-thickness excision wound model. Our results showed that SA/Col hydrogel was injectable, biodegradable, and exhibited low immunogenicity, which could promote the retention and survival of hUC-MSCs in vivo. SA/Col loaded with hUC-MSCs showed reduced wound size (p < 0.05). Histological and immunofluorescence results confirmed that SA/Col loaded with hUC-MSCs significantly promoted the formation of granulation, enhanced collagen deposition and angiogenesis, increased VEGF and TGF-β1 expression (p < 0.05), and mitigated inflammation evidenced by lower production of TNF-α and IL-1β and higher release of IL-4 and IL-10 (p < 0.05). Furthermore, SA/Col loaded with hUC-MSCs significantly lowered the expression of NLRP3 inflammasome-related proteins (p < 0.05). Taken together, our results suggest that SA/Col loaded with hUC-MSCs promotes skin wound healing via partly inhibiting NLRP3 pathway, which has potential to the treatment of skin wounds.
Collapse
Affiliation(s)
- Zhenkun Zhang
- School of Life Sciences, Zhengzhou University, No. 100 Science Avenue, Zhengzhou, 450001, Henan, China
| | - Zhe Li
- School of Life Sciences, Zhengzhou University, No. 100 Science Avenue, Zhengzhou, 450001, Henan, China
| | - Ya Li
- School of Life Sciences, Zhengzhou University, No. 100 Science Avenue, Zhengzhou, 450001, Henan, China
| | - Yingying Wang
- School of Life Sciences, Zhengzhou University, No. 100 Science Avenue, Zhengzhou, 450001, Henan, China
| | - Minghao Yao
- School of Life Sciences, Zhengzhou University, No. 100 Science Avenue, Zhengzhou, 450001, Henan, China
| | - Kun Zhang
- School of Life Sciences, Zhengzhou University, No. 100 Science Avenue, Zhengzhou, 450001, Henan, China
| | - Zhenyu Chen
- School of Life Sciences, Zhengzhou University, No. 100 Science Avenue, Zhengzhou, 450001, Henan, China
| | - Han Yue
- Stem Cell Research Center, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
| | - Jijing Shi
- Central Lab of the First People's Hospital of Zhengzhou, Zhengzhou, 450001, Henan, China
| | - Fangxia Guan
- School of Life Sciences, Zhengzhou University, No. 100 Science Avenue, Zhengzhou, 450001, Henan, China
- Stem Cell Research Center, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
| | - Shanshan Ma
- School of Life Sciences, Zhengzhou University, No. 100 Science Avenue, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
15
|
Mezu-Ndubuisi OJ, Maheshwari A. The role of integrins in inflammation and angiogenesis. Pediatr Res 2021; 89:1619-1626. [PMID: 33027803 PMCID: PMC8249239 DOI: 10.1038/s41390-020-01177-9] [Citation(s) in RCA: 165] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/18/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023]
Abstract
Integrins are heterodimeric transmembrane cell adhesion molecules made up of alpha (α) and beta (β) subunits arranged in numerous dimeric pairings. These complexes have varying affinities to extracellular ligands. Integrins regulate cellular growth, proliferation, migration, signaling, and cytokine activation and release and thereby play important roles in cell proliferation and migration, apoptosis, tissue repair, as well as in all processes critical to inflammation, infection, and angiogenesis. This review presents current evidence from human and animal studies on integrin structure and molecular signaling, with particular emphasis on signal transduction in infants. We have included evidence from our own laboratory studies and from an extensive literature search in databases PubMed, EMBASE, Scopus, and the electronic archives of abstracts presented at the annual meetings of the Pediatric Academic Societies. To avoid bias in identification of existing studies, key words were short-listed prior to the actual search both from anecdotal experience and from PubMed's Medical Subject Heading (MeSH) thesaurus. IMPACT: Integrins are a family of ubiquitous αβ heterodimeric receptors that interact with numerous ligands in physiology and disease. Integrins play a key role in cell proliferation, tissue repair, inflammation, infection, and angiogenesis. This review summarizes current evidence from human and animal studies on integrin structure and molecular signaling and promising role in diseases of inflammation, infection, and angiogenesis in infants. This review shows that integrin receptors and ligands are novel therapeutic targets of clinical interest and hold promise as novel therapeutic targets in the management of several neonatal diseases.
Collapse
Affiliation(s)
- Olachi J. Mezu-Ndubuisi
- grid.14003.360000 0001 2167 3675Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI USA
| | - Akhil Maheshwari
- grid.21107.350000 0001 2171 9311Department of Pediatrics, Johns Hopkins University, Baltimore, MD USA
| |
Collapse
|
16
|
Wang L, Yang J, Yang X, Hou Q, Liu S, Zheng W, Long Y, Jiang X. Mercaptophenylboronic Acid-Activated Gold Nanoparticles as Nanoantibiotics against Multidrug-Resistant Bacteria. ACS APPLIED MATERIALS & INTERFACES 2020; 12:51148-51159. [PMID: 33155812 DOI: 10.1021/acsami.0c12597] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Multidrug-resistant (MDR) bacteria-induced infections are becoming challenging issues threatening human health and life. Current antibiotics can hardly tackle this problem. Herein, we present a strategy to prepare mercaptophenylboronic acid (MBA)-activated gold nanoparticles (Au NPs) as an antibacterial agent against MDR bacteria. Both Au NPs and MBA cannot serve as antibiotics. However, when MBA attaches on Au NPs, the Au_MBA NPs show potent antibacterial activities against Gram-positive MDR clinical isolates (e.g., MDR Staphyloccocus aureus, MDR S. aureus; MDR Staphyloccocus epidermidis, MDR S. epidermidis). Furthermore, Au_MBA NPs show an extremely high median lethal dose (LD50,i.v., 960 mg/kg), which is much higher than those of most of the clinically used antibiotics. As an application example, we dope Au_MBA NPs with electrospun poly(ε-caprolactone) (PCL)/gelatin nanofibrous membranes as wound dressings, which show striking ability to remedy S. aureus- or MDR S. aureus-infected full-thickness skin wounds on rats. Our study provides a novel strategy for treating MDR bacteria-infected wounds in a simple, low-cost, and efficient way, which holds promise for broad clinical applications.
Collapse
Affiliation(s)
- Le Wang
- School of Life Science and Technology, Harbin Institute of Technology, 2 Yikuang Road, Nangang District, Harbin 150001, China
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong 518055, China
- Collaborative Innovation Center for Nanomaterials & Devices, College of Physics, Qingdao University, Qingdao 266071, China
| | - Junchuan Yang
- GBA Research Innovation Institute for Nanotechnology, CAS Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology, Beijing 100190, China
| | - Xinglong Yang
- GBA Research Innovation Institute for Nanotechnology, CAS Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology, Beijing 100190, China
| | - Qinghong Hou
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong 518055, China
| | - Shaoqin Liu
- School of Life Science and Technology, Harbin Institute of Technology, 2 Yikuang Road, Nangang District, Harbin 150001, China
| | - Wenfu Zheng
- GBA Research Innovation Institute for Nanotechnology, CAS Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology, Beijing 100190, China
| | - Yunze Long
- Collaborative Innovation Center for Nanomaterials & Devices, College of Physics, Qingdao University, Qingdao 266071, China
| | - Xingyu Jiang
- Department of Biomedical Engineering, Southern University of Science and Technology, No. 1088 Xueyuan Road, Nanshan District, Shenzhen, Guangdong 518055, China
| |
Collapse
|
17
|
Jo SB, Park CY, Kang HK, Jung SY, Min BM. The laminin-211-derived PPFEGCIWN motif accelerates wound reepithelialization and increases phospho-FAK-Tyr397 and Rac1-GTP levels in a rat excisional wound splinting model. J Tissue Eng Regen Med 2020; 14:1100-1112. [PMID: 32592615 DOI: 10.1002/term.3084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/31/2020] [Accepted: 06/03/2020] [Indexed: 11/11/2022]
Abstract
We previously reported that the PPFEGCIWN motif (Ln2-LG3-P2-DN3), residues 2678-2686 of the human laminin α2 chain, promotes cell attachment of normal human epidermal keratinocytes (NHEKs) and dermal fibroblasts (NHDFs); however, its in vivo effects on cutaneous wound healing have not yet been examined. In this study, we sought to determine whether Ln2-LG3-P2-DN3 could promote full-thickness cutaneous wound healing by accelerating wound reepithelialization and wound closure in vivo. Ln2-LG3-P2-DN3 had significantly higher cell attachment and spreading activities than vehicle or scrambled peptide control in both NHEKs and NHDFs in vitro. The wound area was significantly smaller in rats treated with Ln2-LG3-P2-DN3 than in those treated with vehicle or scrambled peptide in the early phase of wound healing. Furthermore, Ln2-LG3-P2-DN3 significantly accelerated wound reepithelialization relative to vehicle or scrambled peptide and promoted FAK-Tyr397 phosphorylation and Rac1 activation. Collectively, our findings suggest that the PPFEGCIWN motif has potential as a therapeutic agent for cutaneous regeneration via the acceleration of wound reepithelization and wound closure.
Collapse
Affiliation(s)
- Seung Bin Jo
- Department of Oral Biochemistry and Program in Cancer and Developmental Biology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, South Korea
| | - Cho Yeon Park
- Department of Oral Biochemistry and Program in Cancer and Developmental Biology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, South Korea
| | - Hyun Ki Kang
- Department of Oral Biochemistry and Program in Cancer and Developmental Biology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, South Korea
| | - Sung Youn Jung
- Department of Oral Biochemistry and Program in Cancer and Developmental Biology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, South Korea
| | - Byung-Moo Min
- Department of Oral Biochemistry and Program in Cancer and Developmental Biology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, South Korea
| |
Collapse
|
18
|
Später T, Menger MM, Nickels RM, Menger MD, Laschke MW. Macrophages promote network formation and maturation of transplanted adipose tissue-derived microvascular fragments. J Tissue Eng 2020; 11:2041731420911816. [PMID: 32313616 PMCID: PMC7153185 DOI: 10.1177/2041731420911816] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 02/18/2020] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue–derived microvascular fragments rapidly reassemble into microvascular networks within implanted scaffolds. Herein, we analyzed the contribution of macrophages to this process. C57BL/6 mice received clodronate (clo)-containing liposomes for macrophage depletion, whereas animals treated with phosphate-buffered-saline-containing liposomes served as controls. Microvascular fragments were isolated from clo- and phosphate-buffered-saline-treated donor mice and seeded onto collagen–glycosaminoglycan matrices, which were implanted into dorsal skinfold chambers of clo- and phosphate-buffered-saline-treated recipient mice. The implants’ vascularization and incorporation were analyzed by stereomicroscopy, intravital fluorescence microscopy, histology, and immunohistochemistry. Compared to controls, matrices within clo-treated animals exhibited a significantly reduced functional microvessel density. Moreover, they contained a lower fraction of microvessels with an α-smooth muscle actin (SMA)+ cell layer, indicating impaired vessel maturation. This was associated with a deteriorated implant incorporation. These findings demonstrate that macrophages not only promote the reassembly of microvascular fragments into microvascular networks, but also improve their maturation during this process.
Collapse
Affiliation(s)
- Thomas Später
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Maximilian M Menger
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany.,Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Ruth M Nickels
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| |
Collapse
|
19
|
The HIPPO Transducer YAP and Its Targets CTGF and Cyr61 Drive a Paracrine Signalling in Cold Atmospheric Plasma-Mediated Wound Healing. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4910280. [PMID: 32104533 PMCID: PMC7040405 DOI: 10.1155/2020/4910280] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 12/15/2022]
Abstract
Reactive species play a pivotal role in orchestrating wound healing responses. They act as secondary messengers and drive redox-signalling pathways that are involved in the homeostatic, inflammatory, proliferative, and remodelling phases of wound healing. The application of Cold Atmospheric Plasma (CAP) to the wound site produces a profusion of short- and long-lived reactive species that have been demonstrated to be effective in promoting wound healing; however, knowledge of the mechanisms underlying CAP-mediated wound healing remains scarce. To address this, an in vitro coculture model was used to study the effects of CAP on wound healing and on paracrine crosstalk between dermal keratinocytes and fibroblasts. Using this coculture model, we observed a stimulatory effect on the migration ability of HaCaT cells that were cocultured with dermal fibroblasts. Additionally, CAP treatment resulted in an upregulation of the HIPPO transcription factor YAP in HaCaTs and fibroblasts. Downstream effectors of the HIPPO signalling pathway (CTGF and Cyr61) were also upregulated in dermal fibroblasts, and the administration of antioxidants could inhibit CAP-mediated wound healing and abrogate the gene expression of the HIPPO downstream effectors. Interestingly, we observed that HaCaT cells exhibited an improved cell migration rate when incubated with CAP-treated fibroblast-conditioned media compared to that observed after incubation with untreated media. An induction of CTGF and Cyr61 secretion was also observed upon CAP treatment in the fibroblast-conditioned media. Finally, exposure to recombinant CTGF and Cyr61 could also significantly improve HaCaT cell migration. In summary, our results validated that CAP activates a regenerative signalling pathway at the onset of wound healing. Additionally, CAP also stimulated a reciprocal communication between dermal fibroblasts and keratinocytes, resulting in improved keratinocyte wound healing in coculture.
Collapse
|
20
|
Sigaroodi F, Shafaei H, Karimipour M, Dolatkhah MA, Delazar A. Aloe Vera/Collagen Mixture Induces Integrin α1β1 and PECAM-1 Genes Expression in Human Adipose-Derived Stem Cells. Adv Pharm Bull 2019; 9:662-667. [PMID: 31857972 PMCID: PMC6912176 DOI: 10.15171/apb.2019.077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/06/2019] [Accepted: 06/15/2019] [Indexed: 12/25/2022] Open
Abstract
Purpose: Natural biomaterials are a key base in tissue engineering, and collagen, as the main content of the extracellular matrix (ECM), is frequently used in tissue engineering. Aloe vera has some therapeutic effects on ulcers, therefore, the use of this natural resource has always been considered for improving collagen function. We aimed to evaluate the effect of Aloe vera/ Collagen blended on cell viability, cell attachment, and angiogenic potential by determining of integrin α1β1 and platelet endothelial cell adhesion molecule (PECAM-1) genes expression in human adipose-derived stem cells (hASCs). Methods: In this study, hASCs after harvesting of adipose tissues from abdominal subcutaneous adipose tissue and isolation, were cultured in four groups of control, collagen gel, Aloe vera gel, and Aloe vera/collagen blended in vitro environment at 24h and then cell viability was assessed by MTT (3-(4,5-dimethylthiazol 2-yl)-2,5-diphenyltetrazolium) assay. Integrin α1β1 and PECAM-1 genes expression were evaluated by real-time RT-PCR. Results: The results of MTT showed that the combination of Aloe vera/collagen was retained the cell viability at the normal range and improved it. In real-time RT-PCR results, integrin α1β1 and PECAM-1 gene expression were increased in the Aloe vera/collagen blended group compared to the control group. Conclusion: For tissue engineering purposes, Aloe vera improves collagen properties in the culture of hASCs by increasing the expression of the integrin α1β1 and PECAM-1 genes.
Collapse
Affiliation(s)
- Faraz Sigaroodi
- Stem Cells Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajar Shafaei
- Stem Cells Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Karimipour
- Stem Cells Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Amin Dolatkhah
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Delazar
- Faculty of Pharmacy and Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
21
|
LRG1 Promotes Keratinocyte Migration and Wound Repair through Regulation of HIF-1α Stability. J Invest Dermatol 2019; 140:455-464.e8. [PMID: 31344385 DOI: 10.1016/j.jid.2019.06.143] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/13/2019] [Accepted: 06/30/2019] [Indexed: 01/10/2023]
Abstract
Re-epithelialization is a complex process during skin wound healing, and cell migration is an integral part of this phenomenon. Here we identified a role for LRG1 as a key regulator of epidermal keratinocyte migration where LRG1 acts via enhancement of HIF-1α stability. We showed that LRG1 is upregulated at murine skin wound edges and that addition of recombinant human LRG1 accelerates keratinocyte migration and skin wound healing. Furthermore, we identified transcription factor ELK3 as a downstream effector of LRG1. We confirmed that elevated ELK3 levels manipulated by LRG1 can promote cell migration through upregulation of HIF-1α stability. Because hyperglycemia complicatedly affects HIF-1α stability and activation, our findings provide insights into the molecular controls of wound-associated cell migration and identify potential therapeutic targets for the treatment of chronic diabetic wounds. In conclusion, we demonstrated that LRG1 promotes wound repair through keratinocyte migration and is important for normalization of an abnormal process of diabetic wound healing where HIF-1α stability is insufficient.
Collapse
|
22
|
Chang LY, Fan SMY, Liao YC, Wang WH, Chen YJ, Lin SJ. Proteomic Analysis Reveals Anti-Fibrotic Effects of Blue Light Photobiomodulation on Fibroblasts. Lasers Surg Med 2019; 52:358-372. [PMID: 31321797 DOI: 10.1002/lsm.23137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND OBJECTIVES This study was aimed at determining the effects of blue light photobiomodulation on primary adult mouse dermal fibroblasts (AMDFs) and the associated signaling pathways. STUDY DESIGN/MATERIALS AND METHODS Cultured AMDFs from adult C57BL/6 mice were irradiated by blue light from a light-emitting diode (wavelength = 463 ± 50 nm; irradiance = 5 mW/cm2 ; energy density = 4-8 J/cm2 ). The cells were analyzed using mass spectrometry for proteomics/phosphoproteomics, AlamarBlue assay for mitochondrial activity, time-lapse video for cell migration, quantitative polymerase chain reaction for gene expression, and immunofluorescence for protein expression. RESULTS Proteomic/phosphoproteomic analysis showed inhibition of extracellular signal-regulated kinases/mammalian target of rapamycin and casein kinase 2 pathways, cell motility-related networks, and multiple metabolic processes, including carbon metabolism, biosynthesis of amino acid, glycolysis/gluconeogenesis, and the pentose phosphate pathway. Functional analysis demonstrated inhibition of mitochondrial activities, cell migration, and mitosis. Expression of growth promoting insulin-like growth factor 1 and fibrosis-related genes, including transforming growth factor β1 (TGFβ1) and collagen type 1 ɑ2 chain diminished. Protein expression of α-smooth muscle actin, an important regulator of myofibroblast functions, was also suppressed. CONCLUSIONS Low-level blue light exerted suppressive effects on AMDFs, including suppression of mitochondrial activity, metabolism, cell motility, proliferation, TGFβ1 levels, and collagen I production. Low-level blue light can be a potential treatment for the prevention and reduction of tissue fibrosis, such as hypertrophic scar and keloids. Lasers Surg. Med. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lo-Yu Chang
- School of Medicine, College of Medicine, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Sabrina Mai-Yi Fan
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Yen-Chen Liao
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan.,Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Rd, Taipei 115, Taiwan
| | - Wei-Hung Wang
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Yu-Ju Chen
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan.,Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Rd, Taipei 115, Taiwan
| | - Sung-Jan Lin
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan.,Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, No. 7, Chung-Shan South Road, Taipei 100, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, No. 7, Chung-Shan South Road, Taipei 100, Taiwan
| |
Collapse
|
23
|
Coller BS. Foreword: A Brief History of Ideas About Platelets in Health and Disease. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.09988-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
24
|
Wang H, Li P, Yu D, Zhang Y, Wang Z, Liu C, Qiu H, Liu Z, Ren J, Qu X. Unraveling the Enzymatic Activity of Oxygenated Carbon Nanotubes and Their Application in the Treatment of Bacterial Infections. NANO LETTERS 2018; 18:3344-3351. [PMID: 29763562 DOI: 10.1021/acs.nanolett.7b05095] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Carbon nanotubes (CNTs) and their derivatives have emerged as a series of efficient biocatalysts to mimic the function of natural enzymes in recent years. However, the unsatisfiable enzymatic efficiency usually limits their practical usage ranging from materials science to biotechnology. Here, for the first time, we present the synthesis of several oxygenated-group-enriched carbon nanotubes (o-CNTs) via a facile but green approach, as well as their usage as high-performance peroxidase mimics for biocatalytic reaction. Exhaustive characterizations of the enzymatic activity of o-CNTs have been provided by exploring the accurate effect of various oxygenated groups on their surface including carbonyl, carboxyl, and hydroxyl groups. Because of the "competitive inhibition" effect among all of these oxygenated groups, the catalytic efficiency of o-CNTs is significantly enhanced by weakening the presence of noncatalytic sites. Furthermore, the admirable enzymatic activity of these o-CNTs has been successfully applied in the treatment of bacterial infections, and the results of both in vitro and in vivo nanozyme-mediated bacterial clearance clearly demonstrate the feasibility of o-CNTs as robust peroxidase mimics to effectively decrease the bacterial viability under physiological conditions. We believe that the present study will not only facilitate the construction of novel efficient nanozymes by rationally adjusting the degree of the "competitive inhibition" effect, but also broaden the biological usage of o-CNT-based nanomaterials via their satisfactory enzymatic activity.
Collapse
Affiliation(s)
- Huan Wang
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology , Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022 , P. R. China
- University of Science and Technology of China , Hefei 230029 , P. R. China
| | - Penghui Li
- MOE Key Laboratory of Green Chemistry, College of Chemistry , Sichuan University , Chengdu 610064 , P. R. China
| | - Dongqin Yu
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology , Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022 , P. R. China
- University of Science and Technology of China , Hefei 230029 , P. R. China
| | - Yan Zhang
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology , Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022 , P. R. China
- Graduate School of the Chinese Academy of Sciences , Beijing 100039 , P. R. China
| | - Zhenzhen Wang
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology , Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022 , P. R. China
- Graduate School of the Chinese Academy of Sciences , Beijing 100039 , P. R. China
| | - Chaoqun Liu
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology , Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022 , P. R. China
- Graduate School of the Chinese Academy of Sciences , Beijing 100039 , P. R. China
| | - Hao Qiu
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology , Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022 , P. R. China
- University of Science and Technology of China , Hefei 230029 , P. R. China
| | - Zhen Liu
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology , Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022 , P. R. China
| | - Jinsong Ren
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology , Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022 , P. R. China
| | - Xiaogang Qu
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology , Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022 , P. R. China
| |
Collapse
|
25
|
Schnittert J, Bansal R, Storm G, Prakash J. Integrins in wound healing, fibrosis and tumor stroma: High potential targets for therapeutics and drug delivery. Adv Drug Deliv Rev 2018; 129:37-53. [PMID: 29414674 DOI: 10.1016/j.addr.2018.01.020] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/16/2018] [Accepted: 01/29/2018] [Indexed: 12/20/2022]
Abstract
Wound healing is a complex process, which ultimately leads to fibrosis if not repaired well. Pathologically very similar to fibrosis is the tumor stroma, found in several solid tumors which are regarded as wounds that do not heal. Integrins are heterodimeric surface receptors which control various physiological cellular functions. Additionally, integrins also sense ECM-induced extracellular changes during pathological events, leading to cellular responses, which influence ECM remodeling. The purpose and scope of this review is to introduce integrins as key targets for therapeutics and drug delivery within the scope of wound healing, fibrosis and the tumor stroma. This review provides a general introduction to the biology of integrins including their types, ligands, means of signaling and interaction with growth factor receptors. Furthermore, we highlight integrins as key targets for therapeutics and drug delivery, based on their biological role, expression pattern within human tissues and at cellular level. Next, therapeutic approaches targeting integrins, with a focus on clinical studies, and targeted drug delivery strategies based on ligands are described.
Collapse
|
26
|
Gao SQ, Chang C, Niu XQ, Li LJ, Zhang Y, Gao JQ. Topical application of Hydroxysafflor Yellow A accelerates the wound healing in streptozotocin induced T1DM rats. Eur J Pharmacol 2018; 823:72-78. [PMID: 29408092 DOI: 10.1016/j.ejphar.2018.01.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/12/2018] [Accepted: 01/15/2018] [Indexed: 12/13/2022]
Abstract
To investigate the effects of Hydroxysafflor Yellow A (HSYA), which is derived from safflower, on the proliferation, migration and angiogenesis of cells in vitro and its potential efficacy in vivo when topically applied to a diabetic wound. Human umbilical vein endothelial cells (HUVECs) and mouse macrophage cells (RAW264.7) were used to evaluate angiogenesis and anti-inflammatory activities, respectively. The influence of HSYA on the wound scratch assay was investigated in keratinocytes. A splinted excisional wound model in rats with TIDM induced by streptozotocin was used to assess the effects of wound healing. Collagen disposition and secretion of vascular growth factors (VEGF) as well as transforming growth factor-β1 (TGF-β1) were evaluated by an ELISA assay and histological staining. The in vitro results showed that HSYA could significantly enhance both the neovascularization of HUVECs and the migration of keratinocytes. It showed the significant inhibitory effect on nitric oxide production, indicating the anti-inflammatory activity of HSYA. In vivo, the topical application of HSYA significantly enhanced the wound closure rate, and the time to complete wound closure was 17 days, whereas 30 days were needed with PBS treatment. Further, treatment with HSYA exhibited significant granulation tissue formation with higher collagen content, re-epithelialization and angiogenesis according to Masson's trichrome staining evaluation, VEGE and TGF-β1 ELISA measurement. In conclusion, HSYA application could be considered a promising therapeutic strategy for treating chronic non-healing diabetic foot ulcers.
Collapse
Affiliation(s)
- Si-Qian Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, PR China
| | - Chen Chang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, PR China
| | - Xiao-Qian Niu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, PR China
| | - Long-Jian Li
- Zhejiang Provincial Corps Hospital of Chinese People's Armed Police Forces, Jiaxing, Zhejiang, PR China
| | - Yan Zhang
- Zhejiang Provincial Corps Hospital of Chinese People's Armed Police Forces, Jiaxing, Zhejiang, PR China
| | - Jian-Qing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, PR China; Jiangsu Engineering Research Center for New-Type External and Transdermal Preparations, PR China.
| |
Collapse
|
27
|
Yang TL, Lee PL, Lee DY, Wang WL, Wei SY, Lee CI, Chiu JJ. Differential regulations of fibronectin and laminin in Smad2 activation in vascular endothelial cells in response to disturbed flow. J Biomed Sci 2018; 25:1. [PMID: 29295709 PMCID: PMC5749020 DOI: 10.1186/s12929-017-0402-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/22/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Atherosclerosis occurs in arterial curvatures and branches, where the flow is disturbed with low and oscillatory shear stress (OSS). The remodeling and alterations of extracellular matrices (ECMs) and their composition is the critical step in atherogenesis. In this study, we investigated the effects of different ECM proteins on the regulation of mechanotransduction in vascular endothelial cells (ECs) in response to OSS. METHODS Through the experiments ranging from in vitro cell culture studies on effects of OSS on molecular signaling to in vivo examinations on clinical specimens from patients with coronary artery disease (CAD), we elucidated the roles of integrins and different ECMs, i.e., fibronectin (FN) and laminin (LM), in transforming growth factor (TGF)-β receptor (TβR)-mediated Smad2 activation and nuclear factor-κB (NF-κB) signaling in ECs in response to OSS and hence atherogenesis. RESULTS OSS at 0.5±12 dynes/cm2 induces sustained increases in the association of types I and II TβRs with β1 and β3 integrins in ECs grown on FN, but it only transient increases in ECs grown on LM. OSS induces a sustained activation of Smad2 in ECs on FN, but only a transient activation of Smad2 in ECs on LM. OSS-activation of Smad2 in ECs on FN regulates downstream NF-κB signaling and pro-inflammatory gene expression through the activation of β1 integrin and its association with TβRs. In contrast, OSS induces transient activations of β1 and β3 integrins in ECs on LM, which associate with type I TβR to regulate Smad2 phosphorylation, resulting in transient induction of NF-κB and pro-inflammatory gene expression. In vivo investigations on diseased human coronary arteries from CAD patients revealed that Smad2 is highly activated in ECs of atherosclerotic lesions, which is accompanied by the concomitant increase of FN rather than LM in the EC layer and neointimal region of atherosclerotic lesions. CONCLUSIONS Our findings provide new insights into the mechanisms of how OSS regulates Smad2 signaling and pro-inflammatory genes through the complex signaling networks of integrins, TβRs, and ECMs, thus illustrating the molecular basis of regional pro-inflammatory activation within disturbed flow regions in the arterial tree.
Collapse
Affiliation(s)
- Tung-Lin Yang
- Department of Life Sciences, National Central University, Jung-Li, Taoyuan, Taiwan.,Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Pei-Ling Lee
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Ding-Yu Lee
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan.,Departments of Food Science and Biological Science and Technology, China University of Science and Technology, Taipei, Taiwan
| | - Wei-Li Wang
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Shu-Yi Wei
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Chih-I Lee
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Jeng-Jiann Chiu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan. .,Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan. .,Institute of Biomedical Engineering, National Cheng-Kung University, Tainan, Taiwan. .,College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
28
|
Rapisarda V, Borghesan M, Miguela V, Encheva V, Snijders AP, Lujambio A, O'Loghlen A. Integrin Beta 3 Regulates Cellular Senescence by Activating the TGF-β Pathway. Cell Rep 2017; 18:2480-2493. [PMID: 28273461 PMCID: PMC5357738 DOI: 10.1016/j.celrep.2017.02.012] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 12/06/2016] [Accepted: 01/31/2017] [Indexed: 12/21/2022] Open
Abstract
Cellular senescence is an important in vivo mechanism that prevents the propagation of damaged cells. However, the precise mechanisms regulating senescence are not well characterized. Here, we find that ITGB3 (integrin beta 3 or β3) is regulated by the Polycomb protein CBX7. β3 expression accelerates the onset of senescence in human primary fibroblasts by activating the transforming growth factor β (TGF-β) pathway in a cell-autonomous and non-cell-autonomous manner. β3 levels are dynamically increased during oncogene-induced senescence (OIS) through CBX7 Polycomb regulation, and downregulation of β3 levels overrides OIS and therapy-induced senescence (TIS), independently of its ligand-binding activity. Moreover, cilengitide, an αvβ3 antagonist, has the ability to block the senescence-associated secretory phenotype (SASP) without affecting proliferation. Finally, we show an increase in β3 levels in a subset of tissues during aging. Altogether, our data show that integrin β3 subunit is a marker and regulator of senescence.
Collapse
Affiliation(s)
- Valentina Rapisarda
- Epigenetics & Cellular Senescence Group, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK
| | - Michela Borghesan
- Epigenetics & Cellular Senescence Group, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK
| | - Veronica Miguela
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA
| | - Vesela Encheva
- Protein Analysis and Proteomics Group, The Francis Crick Institute, South Mimms EN6 3LD, UK
| | - Ambrosius P Snijders
- Protein Analysis and Proteomics Group, The Francis Crick Institute, South Mimms EN6 3LD, UK
| | - Amaia Lujambio
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA
| | - Ana O'Loghlen
- Epigenetics & Cellular Senescence Group, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK.
| |
Collapse
|
29
|
Yang X, Yang J, Wang L, Ran B, Jia Y, Zhang L, Yang G, Shao H, Jiang X. Pharmaceutical Intermediate-Modified Gold Nanoparticles: Against Multidrug-Resistant Bacteria and Wound-Healing Application via an Electrospun Scaffold. ACS NANO 2017; 11:5737-5745. [PMID: 28531351 DOI: 10.1021/acsnano.7b01240] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Remedying a multidrug-resistant (MDR) bacteria wound infection is a major challenge due to the inability of conventional antibiotics to treat such infections against MDR bacteria. Thus, developing wound dressings for wound care, particularly against MDR bacteria, is in huge demand. Here, we present a strategy in designing wound dressings: we use a small molecule (6-aminopenicillanic acid, APA)-coated gold nanoparticles (AuNPs) to inhibit MDR bacteria. We dope the AuNPs into electrospun fibers of poly(ε-caprolactone) (PCL)/gelatin to yield materials that guard against wound infection by MDR bacteria. We systematically evaluate the bactericidal activity of the AuNPs and wound-healing capability via the electrospun scaffold. APA-modified AuNPs (Au_APA) exhibit remarkable antibacterial activity even when confronted with MDR bacteria. Meanwhile, Au_APA has outstanding biocompatibility. Moreover, an in vivo bacteria-infected wound-healing experiment indicates that it has a striking ability to remedy a MDR bacteria wound infection. This wound scaffold can assist the wound care for bacterial infections.
Collapse
Affiliation(s)
- Xinglong Yang
- Natural Products Research Center, Chengdu Institute of Biology, Chinese Academy of Sciences , Chengdu, Sichuan 610041, China
- CAS Center for Excellence in Nanoscience, CAS Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology , ZhongGuanCun BeiYiTiao, Beijing 100190, China
- University of Chinese Academy of Science , Beijing 100049, China
| | - Junchuan Yang
- CAS Center for Excellence in Nanoscience, CAS Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology , ZhongGuanCun BeiYiTiao, Beijing 100190, China
- National Engineering Research Center for Nano-Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology , Wuhan 430074, China
| | - Le Wang
- CAS Center for Excellence in Nanoscience, CAS Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology , ZhongGuanCun BeiYiTiao, Beijing 100190, China
| | - Bei Ran
- CAS Center for Excellence in Nanoscience, CAS Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology , ZhongGuanCun BeiYiTiao, Beijing 100190, China
| | - Yuexiao Jia
- CAS Center for Excellence in Nanoscience, CAS Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology , ZhongGuanCun BeiYiTiao, Beijing 100190, China
| | - Lingmin Zhang
- CAS Center for Excellence in Nanoscience, CAS Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology , ZhongGuanCun BeiYiTiao, Beijing 100190, China
| | - Guang Yang
- National Engineering Research Center for Nano-Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology , Wuhan 430074, China
| | - Huawu Shao
- Natural Products Research Center, Chengdu Institute of Biology, Chinese Academy of Sciences , Chengdu, Sichuan 610041, China
| | - Xingyu Jiang
- CAS Center for Excellence in Nanoscience, CAS Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology , ZhongGuanCun BeiYiTiao, Beijing 100190, China
| |
Collapse
|
30
|
An SY, Jang YJ, Lim HJ, Han J, Lee J, Lee G, Park JY, Park SY, Kim JH, Do BR, Han C, Park HK, Kim OH, Song MJ, Kim SJ, Kim JH. Milk Fat Globule-EGF Factor 8, Secreted by Mesenchymal Stem Cells, Protects Against Liver Fibrosis in Mice. Gastroenterology 2017; 152:1174-1186. [PMID: 27956229 DOI: 10.1053/j.gastro.2016.12.003] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 11/16/2016] [Accepted: 12/03/2016] [Indexed: 01/10/2023]
Abstract
BACKGROUND & AIMS Mesenchymal stem cells (MSCs) mediate tissue repair and might be used to prevent or reduce liver fibrosis. However, little is known about the anti-fibrotic factors secreted from MSCs or their mechanisms. METHODS Umbilical cord-derived MSCs (UCMSCs) were differentiated into hepatocyte-like cells (hpUCMSCs), medium was collected, and secretome proteins were identified and quantified using nanochip-liquid chromatography/quadrupole time-of-flight mass spectrometry. Liver fibrosis was induced in mice by intraperitoneal injection of thioacetamide or CCl4; some mice were then given injections of secretomes or proteins. Liver tissues were collected and analyzed by histology or polymerase chain reaction array to analyze changes in gene expression patterns. We analyzed the effects of MSC secretomes and potential anti-fibrotic proteins on transforming growth factor β 1 (TGFβ1)-mediated activation of human hepatic stellate cell (HSC) lines (hTert-HSC and LX2) and human primary HSCs. Liver tissues were collected from 16 patients with liver cirrhosis and 16 individuals without cirrhosis (controls) in Korea and analyzed by immunohistochemistry and immunoblots. RESULTS In mice with fibrosis, accumulation of extracellular matrix proteins was significantly reduced 3 days after injecting secretomes from UCMSCs, and to a greater extent from hpUCMSCs; numbers of activated HSCs that expressed the myogenic marker α-smooth muscle actin (α-SMA, encoded by ACTA2 [actin, alpha 2, smooth muscle]) were also reduced. Secretomes from UCMSCs, and to a greater extent from hpUCMSCs, reduced liver expression of multiple fibrotic factors, collagens, metalloproteinases, TGFβ, and Smad proteins in the TGFβ signaling pathways. In HSC cell lines and primary HSCs, TGFβ1-stimulated upregulation of α-SMA was significantly inhibited (and SMAD2 phosphorylation reduced) by secretomes from UCMSCs, and to a greater extent from hpUCMSCs. We identified 32 proteins in secretomes of UCMSCs that were more highly concentrated in secretomes from hpUCMSCs and inhibited TGFβ-mediated activation of HSCs. One of these, milk fat globule-EGF factor 8 (MFGE8), was a strong inhibitor of activation of human primary HSCs. We found MFGE8 to down-regulate expression of TGFβ type I receptor by binding to αvβ3 integrin on HSCs and to be secreted by MSCs from umbilical cord, teeth, and bone marrow. In mice, injection of recombinant human MFGE8 had anti-fibrotic effects comparable to those of the hpUCMSC secretome, reducing extracellular matrix deposition and HSC activation. Co-injection of an antibody against MFGE8 reduced the anti-fibrotic effects of the hpUCMSC secretome in mice. Levels of MFGE8 were reduced in cirrhotic liver tissue from patients compared with controls. CONCLUSIONS MFGE8 is an anti-fibrotic protein in MSC secretomes that strongly inhibits TGFβ signaling and reduces extracellular matrix deposition and liver fibrosis in mice.
Collapse
Affiliation(s)
- Su Yeon An
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Yu Jin Jang
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Hee-Joung Lim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Jiyou Han
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Jaehun Lee
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Gyunggyu Lee
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Ji Young Park
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Seo-Young Park
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Ji Hyang Kim
- Biotechnology Research Institute, HurimBioCell Inc., Seoul, Republic of Korea
| | - Byung-Rok Do
- Biotechnology Research Institute, HurimBioCell Inc., Seoul, Republic of Korea
| | - Choongseong Han
- Department of Oral Medicine and Oral Diagnosis, Seoul National University Dental Hospital, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea; Nexel, Co., Ltd, Seoul, Republic of Korea
| | - Hee-Kyung Park
- Department of Oral Medicine and Oral Diagnosis, Seoul National University Dental Hospital, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Ok-Hee Kim
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daejeon, Republic of Korea
| | - Myeong Jun Song
- Division of Hepatology, Department of Internal Medicine, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daejeon, Republic of Korea
| | - Say-June Kim
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daejeon, Republic of Korea
| | - Jong-Hoon Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
31
|
Khan Z, Marshall JF. The role of integrins in TGFβ activation in the tumour stroma. Cell Tissue Res 2016; 365:657-73. [PMID: 27515461 PMCID: PMC5010607 DOI: 10.1007/s00441-016-2474-y] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/07/2016] [Indexed: 12/27/2022]
Abstract
TGFβ1 is the most pleiotropic of all known cytokines and thus, to avoid uncontrolled TGFβ-activated processes, its activity is tightly regulated. Studies in fibrosis have led to the discovery that αv integrins are the major regulators of the local activation of latent TGFβ in our tissues. Since all cells can express one or more types of αv integrins, this raises the possibility that, in the complex milieu of a developing cancer, multiple cell types including both cancer cells and stromal cells activate TGFβ. In normal tissues, TGFβ1 is a tumour suppressor through its ability to suppress epithelial cell division, whereas in cancer, in which tumour cells develop genetic escape mechanisms to become resistant to TGFβ growth suppression, TGFβ signalling creates a tumour-permissive environment by activating fibroblast-to-myofibroblast transition, by promoting angiogenesis, by suppressing immune cell populations and by promoting the secretion of both matrix proteins and proteases. In addition, TGFβ drives epithelial-to-mesenchymal transition (EMT) increasing the potential for metastasis. Since αv integrins activate TGFβ, they almost certainly drive TGFβ-dependent cancer progression. In this review, we discuss the data that are helping to develop this hypothesis and describe the evidence that αv integrins regulate the TGFβ promotion of cancer. Graphical Abstract Mechanisms of integrin-mediated transforming growth factor beta (TGFβ) activation and its effect on stromal processes. 1 Matrix-bound latent LAP-TGFβ1 binds αv integrins expressed by epithelial cells or fibroblasts (LAP latency-associated peptide). TGFβ1 becomes exposed. 2 Active TGFβ1 binds the TGFβ receptor in an autocrine or paracrine fashion. 3 TGFβ1 signalling increases integrin expression, LAP-TGFβ1 secretion and trans-differentiation of fibroblasts into contractile cells that secrete collagens and collagen cross-linking proteins. By contracting the matrix, latent TGFβ1 is stretched making the activation of latent TGFβ1 easier and creating a continuous cycle of TGFβ1 signalling. TGFβ1 promotes cancer progression by promoting angiogenesis, immune suppression and epithelial-to-mesenchymal transition (EMT).
Collapse
Affiliation(s)
- Zareen Khan
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - John F. Marshall
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
32
|
Singel KL, Segal BH. Neutrophils in the tumor microenvironment: trying to heal the wound that cannot heal. Immunol Rev 2016; 273:329-43. [PMID: 27558344 PMCID: PMC5477672 DOI: 10.1111/imr.12459] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neutrophils are the first responders to infection and injury and are critical for antimicrobial host defense. Through the generation of reactive oxidants, activation of granular constituents and neutrophil extracellular traps, neutrophils target microbes and prevent their dissemination. While these pathways are beneficial in the context of trauma and infection, their off-target effects in the context of tumor are variable. Tumor-derived factors have been shown to reprogram the marrow, skewing toward the expansion of myelopoiesis. This can result in stimulation of both neutrophilic leukocytosis and the release of immature granulocytic populations that accumulate in circulation and in the tumor microenvironment. While activated neutrophils have been shown to kill tumor cells, there is growing evidence for neutrophil activation driving tumor progression and metastasis through a number of pathways, including stimulation of thrombosis and angiogenesis, stromal remodeling, and impairment of T cell-dependent anti-tumor immunity. There is also growing appreciation of neutrophil heterogeneity in cancer, with distinct neutrophil populations promoting cancer control or progression. In addition to the effects of tumor on neutrophil responses, anti-neoplastic treatment, including surgery, chemotherapy, and growth factors, can influence neutrophil responses. Future directions for research are expected to result in more mechanistic knowledge of neutrophil biology in the tumor microenvironment that may be exploited as prognostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Kelly L. Singel
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Brahm H. Segal
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
- Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| |
Collapse
|
33
|
Santurdes N, González-Gómez A, Martín del Campo-Fierro M, Rosales-Ibáñez R, Oros-Ovalle C, Vázquez-Lasa B, San Román J. Development of bioresorbable bilayered systems for application as affordable wound dressings. J BIOACT COMPAT POL 2016. [DOI: 10.1177/0883911516635840] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The objective of this work was the preparation and evaluation of a bioresorbable bilayered system for application in the treatment of dermal lesions. The system was based on a polyesterurethane as the external layer and a gelatin membrane as the internal layer. The polyesterurethane was synthesized from poly(ε-caprolactone), polyethylene glycol of 1 or 10 kDa as a hydrophilic component or Pluronic F127 as an amphiphilic component and l-lysine ethyl ester diisocyanate as an urethane precursor. Gelatin membrane was obtained by crosslinking with the naturally occurring crosslinker genipin. Three important points were addressed in this study: the physicochemical characterization of the system, the in vitro behaviour and the in vivo performance on a full-thickness wound defect of rat. The polyesterurethane containing polyethylene glycol of 10 kDa presented the optimum properties for the designed application as to be tested in animal experiments. The in vivo results showed good healing of the lesion with the formation of epidermis similar to normal rat skin. These promising results suggest the potential of this system to be used as an affordable wound dressing in the treatment of different dermal lesions.
Collapse
Affiliation(s)
- N Santurdes
- Institute of Polymer Science and Technology, CSIC, Madrid, Spain
| | - A González-Gómez
- Institute of Polymer Science and Technology, CSIC, Madrid, Spain
- CIBER, Carlos III Health Institute, Madrid, Spain
| | | | - R Rosales-Ibáñez
- Faculty of Stomatology, Autonomous University of San Luis Potosi, San Luis Potosi, Mexico
| | - C Oros-Ovalle
- Faculty of Medicine, Autonomous University of San Luis Potosi and Hospital Central ‘Dr. Ignacio Morones Prieto’, San Luis Potosi, Mexico
| | - B Vázquez-Lasa
- Institute of Polymer Science and Technology, CSIC, Madrid, Spain
- CIBER, Carlos III Health Institute, Madrid, Spain
| | - J San Román
- Institute of Polymer Science and Technology, CSIC, Madrid, Spain
- CIBER, Carlos III Health Institute, Madrid, Spain
| |
Collapse
|
34
|
Sialylation of vitronectin regulates stress fiber formation and cell spreading of dermal fibroblasts via a heparin-binding site. Glycoconj J 2016; 33:227-36. [PMID: 26979432 DOI: 10.1007/s10719-016-9660-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/10/2016] [Accepted: 03/01/2016] [Indexed: 10/22/2022]
Abstract
Vitronectin (VN) plays an important role in tissue regeneration. We previously reported that VN from partial hepatectomized (PH) rats results in a decrease of sialylation of VN and de-sialylation of VN decreases the cell spreading of hepatic stellate cells. In this study, we analyzed the mechanism how sialylation of VN regulates the properties of mouse primary cultured dermal fibroblasts (MDF) and a dermal fibroblast cell line, Swiss 3T3 cells. At first, we confirmed that VN from PH rats or de-sialylated VN also decreased cell spreading in MDF and Swiss 3T3 cells. The de-sialylation suppressed stress fiber formation in Swiss 3T3 cells. Next, we analyzed the effect of the de-sialylation of VN on stress fiber formation in Swiss 3T3 cells. RGD peptide, an inhibitor for a cell binding site of VN, did not affect the cell attachment of Swiss 3T3 cells on untreated VN but significantly decreased it on de-sialylated VN, suggesting that the de-sialylation attenuates the binding activity of an RGD-independent binding site in VN. To analyze a candidate RGD-independent binding site, an inhibition experiment of stress fiber formation for a heparin binding site was performed. The addition of heparin and treatment of cells with heparinase decreased stress fiber formation in Swiss 3T3 cells. Furthermore, de-sialylation increased the binding activity of VN to heparin, as detected by surface plasmon resonance (SPR). These results demonstrate that sialylation of VN glycans regulates stress fiber formation and cell spreading of dermal fibroblast cells via a heparin binding site.
Collapse
|
35
|
Allen J, Ryu J, Maggi A, Flores B, Greer JR, Desai T. Tunable Microfibers Suppress Fibrotic Encapsulation via Inhibition of TGFβ Signaling. Tissue Eng Part A 2015; 22:142-50. [PMID: 26507808 DOI: 10.1089/ten.tea.2015.0087] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Fibrotic encapsulation limits the efficacy and lifetime of implantable biomedical devices. Microtopography has shown promise in the regulation of myofibroblast differentiation, a key driver of fibrotic encapsulation. However, existing studies have not systematically isolated the requisite geometric parameters for suppression of myofibroblast differentiation via microtopography, and there has not been in vivo validation of this technology to date. To address these issues, a novel lamination method was developed to afford more control over topography dimensions. Specifically, in this study we focus on fiber length and its effect on myofibroblast differentiation. Fibroblasts cultured on films with microfibers exceeding 16 μm in length lost the characteristic morphology associated with myofibroblast differentiation, while shorter microfibers of 6 μm length failed to produce this phenotype. This increase in length corresponded to a 50% decrease in fiber stiffness, which acts as a mechanical cue to influence myofibroblast differentiation. Longer microfiber films suppressed expression of myofibroblast-specific genes (αSMA, Col1α2, and Col3α1) and TGFβ signaling components (TGFβ1, TβR2, and Smad3). About 16 μm long microfiber films subcutaneously implanted in a mouse wound-healing model generated a substantially thinner fibrotic capsule and less deposition of collagen in the wound bed. Together, these results identify a critical feature length threshold for microscale topography-mediated repression of fibrotic encapsulation. This study also demonstrates a simple and powerful strategy to improve surface biocompatibility and reduce fibrotic encapsulation around implanted materials.
Collapse
Affiliation(s)
- Jessica Allen
- 1 UCSF Department of Bioengineering and Therapeutic Sciences, San Francisco , California
| | - Jubin Ryu
- 2 UCSF Department of Dermatology, San Francisco , California
| | - Alessandro Maggi
- 3 California Institute of Technology , Department of Medical Engineering, Pasadena, California
| | - Bianca Flores
- 1 UCSF Department of Bioengineering and Therapeutic Sciences, San Francisco , California
| | - Julia R Greer
- 4 California Institute of Technology, Division of Engineering and Applied Science, Kavli Nanoscience Institute , Pasadena, California
| | - Tejal Desai
- 1 UCSF Department of Bioengineering and Therapeutic Sciences, San Francisco , California
| |
Collapse
|
36
|
Mesenchymal stem cell-laden anti-inflammatory hydrogel enhances diabetic wound healing. Sci Rep 2015; 5:18104. [PMID: 26643550 PMCID: PMC4672289 DOI: 10.1038/srep18104] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 11/11/2015] [Indexed: 12/17/2022] Open
Abstract
The purpose of this study was to permit bone marrow mesenchymal stem cells (BMSCs) to reach their full potential in the treatment of chronic wounds. A biocompatible multifunctional crosslinker based temperature sensitive hydrogel was developed to deliver BMSCs, which improve the chronic inflammation microenvironments of wounds. A detailed in vitro investigation found that the hydrogel is suitable for BMSC encapsulation and can promote BMSC secretion of TGF-β1 and bFGF. In vivo, full-thickness skin defects were made on the backs of db/db mice to mimic diabetic ulcers. It was revealed that the hydrogel can inhibit pro-inflammatory M1 macrophage expression. After hydrogel association with BMSCs treated the wound, significantly greater wound contraction was observed in the hydrogel + BMSCs group. Histology and immunohistochemistry results confirmed that this treatment contributed to the rapid healing of diabetic skin wounds by promoting granulation tissue formation, angiogenesis, extracellular matrix secretion, wound contraction, and re-epithelialization. These results show that a hydrogel laden with BMSCs may be a promising therapeutic strategy for the management of diabetic ulcers.
Collapse
|
37
|
Acceleration of diabetic wound healing using a novel protease-anti-protease combination therapy. Proc Natl Acad Sci U S A 2015; 112:15226-31. [PMID: 26598687 DOI: 10.1073/pnas.1517847112] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Nonhealing chronic wounds are major complications of diabetes resulting in >70,000 annual lower-limb amputations in the United States alone. The reasons the diabetic wound is recalcitrant to healing are not fully understood, and there are limited therapeutic agents that could accelerate or facilitate its repair. We previously identified two active forms of matrix metalloproteinases (MMPs), MMP-8 and MMP-9, in the wounds of db/db mice. We argued that the former might play a role in the body's response to wound healing and that the latter is the pathological consequence of the disease with detrimental effects. Here we demonstrate that the use of compound ND-336, a novel highly selective inhibitor of gelatinases (MMP-2 and MMP-9) and MMP-14, accelerates diabetic wound healing by lowering inflammation and by enhancing angiogenesis and re-epithelialization of the wound, thereby reversing the pathological condition. The detrimental role of MMP-9 in the pathology of diabetic wounds was confirmed further by the study of diabetic MMP-9-knockout mice, which exhibited wounds more prone to healing. Furthermore, topical administration of active recombinant MMP-8 also accelerated diabetic wound healing as a consequence of complete re-epithelialization, diminished inflammation, and enhanced angiogenesis. The combined topical application of ND-336 (a small molecule) and the active recombinant MMP-8 (an enzyme) enhanced healing even more, in a strategy that holds considerable promise in healing of diabetic wounds.
Collapse
|
38
|
Coulson-Thomas VJ, Chang SH, Yeh LK, Coulson-Thomas YM, Yamaguchi Y, Esko J, Liu CY, Kao W. Loss of corneal epithelial heparan sulfate leads to corneal degeneration and impaired wound healing. Invest Ophthalmol Vis Sci 2015; 56:3004-14. [PMID: 26024086 DOI: 10.1167/iovs.14-15341] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Heparan sulfate (HS) is a highly modified glycosaminoglycan (GAG) bound to a core protein to form heparan sulfate proteoglycans (HSPGs) that are vital in many cellular processes ranging from development to adult physiology, as well as in disease, through interactions with various protein ligands. This study aimed to elucidate the role of HS in corneal epithelial homeostasis and wound healing. METHODS An inducible quadruple transgenic mouse model was generated to excise Ext1 and Ndst1, which encode the critical HS chain elongation enzyme and N-deacetylase/N-sulfotransferase, respectively, in keratin 14-positive cells upon doxycycline induction. RESULTS EXT(Δ/ΔCEpi) mice (deletion of Ext1 in corneal epithelium) induced at P20 presented progressive thinning of the corneal epithelium with a significant loss in the number of epithelial layers by P55. EXT(Δ/ΔCEpi) mice presented tight junction disruption, loss of cell-basement membrane adhesion complexes, and impaired wound healing. Interestingly, EXT(Δ/ΔCEpi) and NDST(Δ/ΔCEpi) mice presented an increase in cell proliferation, which was assayed by both Ki67 staining and 5-ethynyl-2'-deoxyuridine (EdU) incorporation. Moreover, EXT(Δ/ΔCEpi) mice presented compromised epithelial stratification 7 days after a debridement wound. The conditional knockout of HS from keratocytes using the keratocan promoter led to no corneal abnormalities or any disruption in wound healing. CONCLUSIONS Corneal epithelial cells require HS for maintaining corneal homeostasis, and the loss of epithelial HS leads to both impaired wound healing and impaired corneal stratification.
Collapse
Affiliation(s)
| | - Shao-Hsuan Chang
- Department of Ophthalmology University of Cincinnati, Cincinnati, Ohio, United States
| | - Lung-Kun Yeh
- Department of Ophthalmology, Chang-Gung Memorial Hospital, Chang-Gung University College of Medicine, Linko, Taiwan
| | | | - Yu Yamaguchi
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States
| | - Jeffrey Esko
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California-San Diego, La Jolla, California, United States
| | - Chia-Yang Liu
- Department of Ophthalmology University of Cincinnati, Cincinnati, Ohio, United States
| | - Winston Kao
- Department of Ophthalmology University of Cincinnati, Cincinnati, Ohio, United States
| |
Collapse
|
39
|
Merna N, Fung KM, Wang JJ, King CR, Hansen KC, Christman KL, George SC. Differential β3 Integrin Expression Regulates the Response of Human Lung and Cardiac Fibroblasts to Extracellular Matrix and Its Components. Tissue Eng Part A 2015; 21:2195-205. [PMID: 25926101 PMCID: PMC4528988 DOI: 10.1089/ten.tea.2014.0337] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 04/27/2015] [Indexed: 11/12/2022] Open
Abstract
Extracellular matrix (ECM) derived from whole organ decellularization has been successfully used in a variety of tissue engineering applications. ECM contains a complex mixture of functional and structural molecules that are ideally suited for the tissue from which the ECM is harvested. However, decellularization disrupts the structural properties and protein composition of the ECM, which may impact function when cells such as the fibroblast are reintroduced during recellularization. We hypothesized that the ECM structure and composition, fibroblast source, and integrin expression would influence the fibroblast phenotype. Human cardiac fibroblasts (HCFs) and normal human lung fibroblasts (NHLFs) were cultured on intact cardiac ECM, collagen gels, and coatings composed of cardiac ECM, lung ECM, and individual ECM components (collagen and fibronectin [FN]) for 48 h. COL1A expression of HCFs and NHLFs cultured on ECM and FN coatings decreased to <50% of that of untreated cells; COL1A expression for HCFs cultured on ECM coatings was one- to twofold higher than HCFs cultured on intact ECM. NHLFs cultured on ECM and FN coatings expressed 12- to 31-fold more alpha-smooth muscle actin (αSMA) than HCFs; the αSMA expression for HCFs and NHLFs cultured on ECM coatings was ∼2- to 5-fold higher than HCFs and NHLFs cultured on intact ECM. HCFs expressed significantly higher levels of β3 and β4 integrins when compared to NHLFs. Inhibition of the β3 integrin, but not β4, resulted in a 16- to 26-fold increase in αSMA expression in HCFs cultured on ECM coatings and FN. Our results demonstrate that β3 integrin expression depends on the source of the fibroblast and that its expression inhibits αSMA expression (and thus the myofibroblast phenotype). We conclude that the fibroblast source and integrin expression play important roles in regulating the fibroblast phenotype.
Collapse
Affiliation(s)
- Nick Merna
- Department of Biomedical Engineering, University of California, Irvine, California
| | - Kelsey M. Fung
- Department of Biomedical Engineering, University of California, Irvine, California
| | - Jean J. Wang
- Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, California
| | - Cristi R. King
- Department of Biomedical Engineering, Washington University in St. Louis, Missouri
| | - Kirk C. Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, Colorado
| | - Karen L. Christman
- Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, California
| | - Steven C. George
- Department of Biomedical Engineering, Washington University in St. Louis, Missouri
| |
Collapse
|
40
|
Cichon MA, Radisky DC. Extracellular matrix as a contextual determinant of transforming growth factor-β signaling in epithelial-mesenchymal transition and in cancer. Cell Adh Migr 2015; 8:588-94. [PMID: 25482625 PMCID: PMC4594483 DOI: 10.4161/19336918.2014.972788] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Extracellular matrix (ECM) provides both structural support and contextual information to cells within tissues and organs. The combination of biochemical and biomechanical signals from the ECM modulates responses to extracellular signals toward differentiation, proliferation, or apoptosis; alterations in the ECM are necessary for development and remodeling processes, but aberrations in the composition and organization of ECM are associated with disease pathology and can predispose to development of cancer. The primary cell surface sensors of the ECM are the integrins, which provide the physical connection between the ECM and the cytoskeleton and also convey biochemical information about the composition of the ECM. Transforming growth factor-β (TGF-β) is an extracellular signaling molecule that is a powerful controller of a variety of cellular functions, and that has been found to induce very different outcomes according to cell type and cellular context. It is becoming clear that ECM-mediated signaling through integrins is reciprocally influenced by TGF-β: integrin expression, activation, and responses are affected by cellular exposure to TGF-β, and TGF-β activation and cellular responses are in turn controlled by signaling from the ECM through integrins. Epithelial-mesenchymal transition (EMT), a physiological process that is activated by TGF-β in normal development and in cancer, is also affected by the composition and structure of the ECM. Here, we will outline how signaling from the ECM controls the contextual response to TGF-β, and how this response is selectively modulated during disease, with an emphasis on recent findings, current challenges, and future opportunities.
Collapse
|
41
|
Li Y, Drabsch Y, Pujuguet P, Ren J, van Laar T, Zhang L, van Dam H, Clément-Lacroix P, Ten Dijke P. Genetic depletion and pharmacological targeting of αv integrin in breast cancer cells impairs metastasis in zebrafish and mouse xenograft models. Breast Cancer Res 2015; 17:28. [PMID: 25849225 PMCID: PMC4381510 DOI: 10.1186/s13058-015-0537-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Accepted: 02/11/2015] [Indexed: 12/21/2022] Open
Abstract
Introduction Increased expression of αv integrins is frequently associated with tumor cell adhesion, migration, invasion and metastasis, and correlates with poor prognosis in breast cancer. However, the mechanism by which αv integrins can enhance breast cancer progression is still largely unclear. The effects of therapeutic targeting of αv integrins in breast cancer also have yet to be investigated. Methods We knocked down αv integrin in MDA-MB-231 and MCF10A-M4 breast cancer cells, or treated these cells with the αv antagonist GLPG0187. The effects of αv integrin depletion on mesenchymal markers, transforming growth factor-β (TGF-β)/Smad signaling and TGF-β-induced target gene expression were analyzed in MDA-MB-231 cells by RNA analysis or Western blotting. The function of αv integrin on breast cancer cell migration was investigated by transwell assay in vitro, and its effect on breast cancer progression was assessed by both zebrafish and mouse xenografts in vivo. In the mouse model, GLPG0187 was administered separately, or in combination with the standard-of-care anti-resorptive agent zoledronate and the chemotherapeutic drug paclitaxel, to study the effects of combinational treatments on breast cancer metastasis. Results Genetic interference and pharmacological targeting of αv integrin with GLPG0187 in different breast cancer cell lines inhibited invasion and metastasis in the zebrafish or mouse xenograft model. Depletion of αv integrin in MDA-MB-231 cells inhibited the expression of mesenchymal markers and the TGF-β/Smad response. TGF-β induced αv integrin mRNA expression and αv integrin was required for TGF-β-induced breast cancer cell migration. Moreover, treatment of MDA-MB-231 cells with non-peptide RGD antagonist GLPG0187 decreased TGF-β signaling. In the mouse xenografts GLPG0187 inhibited the progression of bone metastasis. Maximum efficacy of inhibition of bone metastasis was achieved when GLPG0187 was combined with the standard-of-care metastatic breast cancer treatments. Conclusion These findings show that αv integrin is required for efficient TGF-β/Smad signaling and TGF-β-induced breast cancer cell migration, and for maintaining a mesenchymal phenotype of the breast cancer cells. Our results also provide evidence that targeting αv integrin could be an effective therapeutic approach for treatment of breast cancer tumors and/or metastases that overexpress αv integrin. Electronic supplementary material The online version of this article (doi:10.1186/s13058-015-0537-8) contains supplementary material, which is available to authorized users.
Collapse
|
42
|
Liang M, Wang Y, Liang A, Dong JF, Du J, Cheng J. Impaired integrin β3 delays endothelial cell regeneration and contributes to arteriovenous graft failure in mice. Arterioscler Thromb Vasc Biol 2015; 35:607-15. [PMID: 25614287 DOI: 10.1161/atvbaha.114.305089] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Neointima formation is associated with stenosis and subsequent thrombosis in arteriovenous grafts (AVGs). A role of integrin β3 in the neointima formation of AVGs remains poorly understood. APPROACH AND RESULTS In integrin β3(-/-) mice, we found significantly accelerated occlusion of AVGs compared with the wild-type mice. This is caused by the development of neointima and lack of endothelial regeneration. The latter is a direct consequence of impaired functions of circulating angiogenic cells (CACs) and platelets in integrin β3(-/-) mice. Evidence suggests the involvement of platelet regulating CAC homing to and differentiation at graft sites via transforming growth factor-β1 and Notch signaling pathway. First, CACs deficient of integrin β3 impaired adhesion activity toward exposed subendothelium. Second, platelets from integrin β3(-/-) mice failed to sufficiently stimulate CACs to differentiate into mature endothelial cells. Finally, we found that transforming growth factor-β1 level was increased in platelets from integrin β3(-/-) mice and resulted in enhanced Notch1 activation in CACs in AVGs. These results demonstrate that integrin β3 is critical for endothelial cell homing and differentiation. The increased transforming growth factor-β1 and Notch1 signaling mediates integrin β3(-/-)-induced AVG occlusion. This accelerated occlusion of AVGs was reversed in integrin β3(-/-) mice transplanted with the bone marrow from wild-type mice. CONCLUSIONS Our results suggest that boosting integrin β3 function in the endothelial cells and platelets could prevent neointima and thrombosis in AVGs.
Collapse
Affiliation(s)
- Ming Liang
- From the Department of Nephrology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China (M.L.); Department of Cell Biology, Third Military Medical University, Chongqing, China (Y.W.); Puget Sound Blood Research Institute, Hematology Division, Department of Medicine, University of Washington, Seattle (J.-F.D.); Beijing Anzhen Hospital Affiliated to the Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China (J.D.); and Nephrology Division, Baylor College of Medicine, Houston, TX (M.L., Y.W., A.L., J.C.)
| | - Yun Wang
- From the Department of Nephrology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China (M.L.); Department of Cell Biology, Third Military Medical University, Chongqing, China (Y.W.); Puget Sound Blood Research Institute, Hematology Division, Department of Medicine, University of Washington, Seattle (J.-F.D.); Beijing Anzhen Hospital Affiliated to the Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China (J.D.); and Nephrology Division, Baylor College of Medicine, Houston, TX (M.L., Y.W., A.L., J.C.)
| | - Anlin Liang
- From the Department of Nephrology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China (M.L.); Department of Cell Biology, Third Military Medical University, Chongqing, China (Y.W.); Puget Sound Blood Research Institute, Hematology Division, Department of Medicine, University of Washington, Seattle (J.-F.D.); Beijing Anzhen Hospital Affiliated to the Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China (J.D.); and Nephrology Division, Baylor College of Medicine, Houston, TX (M.L., Y.W., A.L., J.C.)
| | - Jin-Fei Dong
- From the Department of Nephrology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China (M.L.); Department of Cell Biology, Third Military Medical University, Chongqing, China (Y.W.); Puget Sound Blood Research Institute, Hematology Division, Department of Medicine, University of Washington, Seattle (J.-F.D.); Beijing Anzhen Hospital Affiliated to the Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China (J.D.); and Nephrology Division, Baylor College of Medicine, Houston, TX (M.L., Y.W., A.L., J.C.)
| | - Jie Du
- From the Department of Nephrology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China (M.L.); Department of Cell Biology, Third Military Medical University, Chongqing, China (Y.W.); Puget Sound Blood Research Institute, Hematology Division, Department of Medicine, University of Washington, Seattle (J.-F.D.); Beijing Anzhen Hospital Affiliated to the Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China (J.D.); and Nephrology Division, Baylor College of Medicine, Houston, TX (M.L., Y.W., A.L., J.C.)
| | - Jizhong Cheng
- From the Department of Nephrology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China (M.L.); Department of Cell Biology, Third Military Medical University, Chongqing, China (Y.W.); Puget Sound Blood Research Institute, Hematology Division, Department of Medicine, University of Washington, Seattle (J.-F.D.); Beijing Anzhen Hospital Affiliated to the Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China (J.D.); and Nephrology Division, Baylor College of Medicine, Houston, TX (M.L., Y.W., A.L., J.C.).
| |
Collapse
|
43
|
Chen S, Zhang M, Shao X, Wang X, Zhang L, Xu P, Zhong W, Zhang L, Xing M, Zhang L. A laminin mimetic peptide SIKVAV-conjugated chitosan hydrogel promoting wound healing by enhancing angiogenesis, re-epithelialization and collagen deposition. J Mater Chem B 2015; 3:6798-6804. [DOI: 10.1039/c5tb00842e] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Angiogenesis and re-epithelialization are critical factors in skin wound healing.
Collapse
Affiliation(s)
- Shixuan Chen
- School of Basic Medical Sciences
- Southern Medical University
- Guangzhou 510515
- China
- Departments of Mechanical Engineering
| | - Min Zhang
- School of Basic Medical Sciences
- Southern Medical University
- Guangzhou 510515
- China
| | - Xuebing Shao
- Departments of Mechanical Engineering
- Biochemistry and Medical Genetics
- University of Manitoba, and Manitoba Children's Hospital of Research Institute
- Winnipeg
- Canada
| | - Xueer Wang
- School of Basic Medical Sciences
- Southern Medical University
- Guangzhou 510515
- China
| | - Lei Zhang
- School of Basic Medical Sciences
- Southern Medical University
- Guangzhou 510515
- China
| | - Pengcheng Xu
- School of Basic Medical Sciences
- Southern Medical University
- Guangzhou 510515
- China
| | - Wen Zhong
- Department of Biosystem Engineering
- University of Manitoba
- Winnipeg
- Canada
| | - Lu Zhang
- School of Basic Medical Sciences
- Southern Medical University
- Guangzhou 510515
- China
- Elderly Health Services Research Center
| | - Malcolm Xing
- Departments of Mechanical Engineering
- Biochemistry and Medical Genetics
- University of Manitoba, and Manitoba Children's Hospital of Research Institute
- Winnipeg
- Canada
| | - Lin Zhang
- School of Basic Medical Sciences
- Southern Medical University
- Guangzhou 510515
- China
| |
Collapse
|
44
|
Koivisto L, Heino J, Häkkinen L, Larjava H. Integrins in Wound Healing. Adv Wound Care (New Rochelle) 2014; 3:762-783. [PMID: 25493210 DOI: 10.1089/wound.2013.0436] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Indexed: 01/06/2023] Open
Abstract
Significance: Regulation of cell adhesions during tissue repair is fundamentally important for cell migration, proliferation, and protein production. All cells interact with extracellular matrix proteins with cell surface integrin receptors that convey signals from the environment into the nucleus, regulating gene expression and cell behavior. Integrins also interact with a variety of other proteins, such as growth factors, their receptors, and proteolytic enzymes. Re-epithelialization and granulation tissue formation are crucially dependent on the temporospatial function of multiple integrins. This review explains how integrins function in wound repair. Recent Advances: Certain integrins can activate latent transforming growth factor beta-1 (TGF-β1) that modulates wound inflammation and granulation tissue formation. Dysregulation of TGF-β1 function is associated with scarring and fibrotic disorders. Therefore, these integrins represent targets for therapeutic intervention in fibrosis. Critical Issues: Integrins have multifaceted functions and extensive crosstalk with other cell surface receptors and molecules. Moreover, in aberrant healing, integrins may assume different functions, further increasing the complexity of their functionality. Discovering and understanding the role that integrins play in wound healing provides an opportunity to identify the mechanisms for medical conditions, such as excessive scarring, chronic wounds, and even cancer. Future Directions: Integrin functions in acute and chronic wounds should be further addressed in models better mimicking human wounds. Application of any products in acute or chronic wounds will potentially alter integrin functions that need to be carefully considered in the design.
Collapse
Affiliation(s)
- Leeni Koivisto
- Laboratory of Periodontal Biology, Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Jyrki Heino
- Department of Biochemistry, University of Turku, Turku, Finland
| | - Lari Häkkinen
- Laboratory of Periodontal Biology, Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Hannu Larjava
- Laboratory of Periodontal Biology, Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| |
Collapse
|
45
|
Winograd-Katz SE, Fässler R, Geiger B, Legate KR. The integrin adhesome: from genes and proteins to human disease. Nat Rev Mol Cell Biol 2014; 15:273-88. [PMID: 24651544 DOI: 10.1038/nrm3769] [Citation(s) in RCA: 445] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The adhesive interactions of cells with their environment through the integrin family of transmembrane receptors have key roles in regulating multiple aspects of cellular physiology, including cell proliferation, viability, differentiation and migration. Consequently, failure to establish functional cell adhesions, and thus the assembly of associated cytoplasmic scaffolding and signalling networks, can have severe pathological effects. The roles of specific constituents of integrin-mediated adhesions, which are collectively known as the 'integrin adhesome', in diverse pathological states are becoming clear. Indeed, the prominence of mutations in specific adhesome molecules in various human diseases is now appreciated, and experimental as well as in silico approaches provide insights into the molecular mechanisms underlying these pathological conditions.
Collapse
Affiliation(s)
- Sabina E Winograd-Katz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Benjamin Geiger
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Kyle R Legate
- 1] Department of Molecular Medicine, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany. [2] Center for Nanosciences, Department of Applied Physics, Ludwig-Maximilians University, 80799 Munich, Germany
| |
Collapse
|
46
|
Parekh R, Lorenzo MK, Shin SY, Pozzi A, Clark AL. Integrin α1β1 differentially regulates cytokine-mediated responses in chondrocytes. Osteoarthritis Cartilage 2014; 22:499-508. [PMID: 24418674 PMCID: PMC4028170 DOI: 10.1016/j.joca.2013.12.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 12/07/2013] [Accepted: 12/25/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To elucidate the role of integrin α1β1 in chondrocyte responses to inflammatory interleukin-1α (IL-1) and anabolic transforming growth factor-β1 (TGF-β1) in the knee. METHODS Intracellular calcium transient responses to IL-1 and TGF-β1 were measured in wild type and integrin α1-null chondrocytes using real time ex vivo confocal microscopy, and immunohistochemistry was performed to analyze TGF-β1-mediated activation of Smad2/3 in tibial and femoral chondrocytes. RESULTS Loss of integrin α1β1 reduces intracellular calcium transient response to IL-1, while it enhances chondrocyte responses to TGF-β1 as measured by intracellular calcium transients and activation of downstream Smad2/3. CONCLUSIONS Integrin α1β1 plays a vital role in mediating chondrocyte responses to two contrasting factors that are critical players in the onset and progression of osteoarthritis - inflammatory IL-1 and anabolic TGF-β. Further investigation into the molecular mechanisms by which integrin α1β1 mediates these responses will be an important next step in understanding the influence of increased expression of integrin α1β1 during the early stages of osteoarthritis on disease progression.
Collapse
Affiliation(s)
- R Parekh
- Faculty of Science, The University of Calgary, Calgary, AB, Canada
| | - MK Lorenzo
- Faculty of Science, The University of Calgary, Calgary, AB, Canada
| | - SY Shin
- Faculty of Kinesiology, The University of Calgary, Calgary, AB, Canada
| | - A Pozzi
- Department of Medicine, Vanderbilt University, Nashville, TN, USA,Department of Medicine, Veterans Affairs Hospital, Nashville, TN, USA
| | - AL Clark
- Faculty of Kinesiology, The University of Calgary, Calgary, AB, Canada,Department of Surgery, Faculty of Medicine, The University of Calgary, Calgary, AB, Canada
| |
Collapse
|
47
|
Gardner A, Borthwick LA, Fisher AJ. Lung epithelial wound healing in health and disease. Expert Rev Respir Med 2014; 4:647-60. [DOI: 10.1586/ers.10.62] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
48
|
Gagen D, Faralli JA, Filla MS, Peters DM. The role of integrins in the trabecular meshwork. J Ocul Pharmacol Ther 2013; 30:110-20. [PMID: 24266581 DOI: 10.1089/jop.2013.0176] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Integrins are a family of heterodimeric transmembrane receptors that mediate adhesion to the extracellular matrix (ECM). However, integrins are not just adhesion receptors. They can act as "bidirectional signal transducers" that coordinate a large number of cellular activities in response to the extracellular environment and intracellular signaling events. Among the activities regulated by integrins are cell adhesion, assembly of the ECM, growth factor signaling, apoptosis, organization of the cytoskeleton, and cytoskeleton-mediated processes such as contraction, endocytosis, and phagocytosis. Integrins regulate these activities through a complex network of intracellular signaling kinases and adaptor proteins that associate with the transmembrane and cytoplasmic domains of the integrin subunits. In this review, we will discuss how some of the known integrin-mediated activities can control the function of the trabecular meshwork. We will also discuss how integrin activity is a tightly regulated process that involves conformation changes within the heterodimer which are mediated by specific integrin-binding proteins.
Collapse
Affiliation(s)
- Debjani Gagen
- 1 Department of Pathology and Laboratory Medicine, Medical Science Center, University of Wisconsin , Madison, Wisconsin
| | | | | | | |
Collapse
|
49
|
Gerber EE, Gallo EM, Fontana SC, Davis EC, Wigley FM, Huso DL, Dietz HC. Integrin-modulating therapy prevents fibrosis and autoimmunity in mouse models of scleroderma. Nature 2013; 503:126-30. [PMID: 24107997 PMCID: PMC3992987 DOI: 10.1038/nature12614] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 08/28/2013] [Indexed: 12/13/2022]
Abstract
In systemic sclerosis (SSc), a common and aetiologically mysterious form of scleroderma (defined as pathological fibrosis of the skin), previously healthy adults acquire fibrosis of the skin and viscera in association with autoantibodies. Familial recurrence is extremely rare and causal genes have not been identified. Although the onset of fibrosis in SSc typically correlates with the production of autoantibodies, whether they contribute to disease pathogenesis or simply serve as a marker of disease remains controversial and the mechanism for their induction is largely unknown. The study of SSc is hindered by a lack of animal models that recapitulate the aetiology of this complex disease. To gain a foothold in the pathogenesis of pathological skin fibrosis, we studied stiff skin syndrome (SSS), a rare but tractable Mendelian disorder leading to childhood onset of diffuse skin fibrosis with autosomal dominant inheritance and complete penetrance. We showed previously that SSS is caused by heterozygous missense mutations in the gene (FBN1) encoding fibrillin-1, the main constituent of extracellular microfibrils. SSS mutations all localize to the only domain in fibrillin-1 that harbours an Arg-Gly-Asp (RGD) motif needed to mediate cell-matrix interactions by binding to cell-surface integrins. Here we show that mouse lines harbouring analogous amino acid substitutions in fibrillin-1 recapitulate aggressive skin fibrosis that is prevented by integrin-modulating therapies and reversed by antagonism of the pro-fibrotic cytokine transforming growth factor β (TGF-β). Mutant mice show skin infiltration of pro-inflammatory immune cells including plasmacytoid dendritic cells, T helper cells and plasma cells, and also autoantibody production; these findings are normalized by integrin-modulating therapies or TGF-β antagonism. These results show that alterations in cell-matrix interactions are sufficient to initiate and sustain inflammatory and pro-fibrotic programmes and highlight new therapeutic strategies.
Collapse
Affiliation(s)
- Elizabeth E Gerber
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Duperret EK, Ridky TW. Focal adhesion complex proteins in epidermis and squamous cell carcinoma. Cell Cycle 2013; 12:3272-85. [PMID: 24036537 DOI: 10.4161/cc.26385] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Focal adhesions (FAs) are large, integrin-containing, multi-protein assemblies spanning the plasma membrane that link the cellular cytoskeleton to surrounding extracellular matrix. They play critical roles in adhesion and cell signaling and are major regulators of epithelial homeostasis, tissue response to injury, and tumorigenesis. Most integrin subunits and their associated FA proteins are expressed in skin, and murine genetic models have provided insight into the functional roles of FAs in normal and neoplastic epidermis. Here, we discuss the roles of these proteins in normal epidermal proliferation, adhesion, wound healing, and cancer. While many downstream signaling mechanisms remain unclear, the critically important roles of FAs are highlighted by the development of therapeutics targeting FAs for human cancer.
Collapse
|