1
|
Park S, Yoon YJ, Hong Y, Yu J, Cho JM, Jeong YJ, Yu H, Jeong H, Lee H, Hwang S, Koh WG, Yang JY, Hyun KA, Jung HI, Lim JY. CD9-enriched extracellular vesicles from chemically reprogrammed basal progenitors of salivary glands mitigate salivary gland fibrosis. Bioact Mater 2025; 47:229-247. [PMID: 39925710 PMCID: PMC11803853 DOI: 10.1016/j.bioactmat.2025.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 12/07/2024] [Accepted: 01/16/2025] [Indexed: 02/11/2025] Open
Abstract
Extracellular vesicles (EVs) derived from stem cells offer promising potential for cell-free therapy. However, refining their cargo for precise disease targeting and delivery remains challenging. This study employed chemical reprogramming via dual inhibition of transforming growth factor beta (TGFβ) and bone morphogenetic protein (BMP) to expand salivary gland basal progenitor cells (sgBPCs). CD9-enriched (CD9+) EVs were then isolated from the sgBPC secretome concentrate using a dual microfluidic chip. Notably, CD9+ EVs demonstrated superior uptake by salivary epithelial cells compared to CD9-depleted (CD9-) EVs and total EVs. In vivo studies using a salivary gland (SG) obstruction mouse model and ex vivo studies in SG fibrosis organoids revealed that CD9+ EVs significantly enhanced anti-fibrotic effects over CD9- EVs and control treatments. The presence of miR-3162 and miR-1290 in CD9+ EVs supported their anti-fibrotic properties by downregulating ACVR1 expression. The chemical reprogramming culture method effectively expanded sgBPCs, enabling consistent and scalable EV production. Utilizing microfluidic chip-isolated CD9+ EVs and ductal delivery presents a targeted and efficient approach for anti-fibrotic SG regeneration.
Collapse
Affiliation(s)
- Sunyoung Park
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seoul, 03722, Republic of Korea
- The DABOM Inc., 50 Yonsei-ro, Seoul, 03722, Republic of Korea
| | - Yeo-Jun Yoon
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yongpyo Hong
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jianning Yu
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seoul, 03722, Republic of Korea
- Department of Biomedical Laboratory Science, Yonsei University, 1 Yeonsedae-gil, Wonju, Gangwon-do, 26493, Republic of Korea
| | - Jae-Min Cho
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Ye Jin Jeong
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Haeun Yu
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seoul, 03722, Republic of Korea
| | - Hyorim Jeong
- The DABOM Inc., 50 Yonsei-ro, Seoul, 03722, Republic of Korea
| | - Hyunjin Lee
- The DABOM Inc., 50 Yonsei-ro, Seoul, 03722, Republic of Korea
| | - Seungyeon Hwang
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seoul, 03722, Republic of Korea
| | - Ji Yeong Yang
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seoul, 03722, Republic of Korea
- The DABOM Inc., 50 Yonsei-ro, Seoul, 03722, Republic of Korea
| | - Kyung-A Hyun
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seoul, 03722, Republic of Korea
- Korea Electronics Technology Institute (KETI), Seongnam, Gyeonggi-do, 13509, Republic of Korea
| | - Hyo-Il Jung
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seoul, 03722, Republic of Korea
- The DABOM Inc., 50 Yonsei-ro, Seoul, 03722, Republic of Korea
| | - Jae-Yol Lim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| |
Collapse
|
2
|
Longmate WM. The epidermal integrin-mediated secretome regulates the skin microenvironment during tumorigenesis and repair. Matrix Biol 2024; 134:175-183. [PMID: 39491760 PMCID: PMC11585437 DOI: 10.1016/j.matbio.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
Integrins are cellular transmembrane receptors that physically connect the cytoskeleton with the extracellular matrix. As such, they are positioned to mediate cellular responses to microenvironmental cues. Importantly, integrins also regulate their own microenvironment through secreted factors, also known as the integrin-mediated secretome. Epidermal integrins, or integrins expressed by keratinocytes of the skin epidermis, regulate the cutaneous microenvironment through the contribution of matrix components, via proteolytic matrix remodeling, or by mediating factors like cytokines and growth factors that can promote support for nearby but distinct cells of the stroma, such as immune cells, endothelial cells, and fibroblasts. This role for integrins is enhanced during both pathological and repair tissue remodeling processes, such as tumor growth and progression and wound healing. This review will discuss examples of how the epithelial integrin-mediated secretome can regulate the tissue microenvironment. Although different epithelial integrins in various contexts will be explored, emphasis will be given to epidermal integrins that regulate the secretome during wound healing and cutaneous tumor progression. Epidermal integrin α3β1 is of particular focus as well, since this integrin has been revealed as a key regulator of the keratinocyte secretome.
Collapse
Affiliation(s)
- Whitney M Longmate
- Department of Surgery, Albany Medical College, Albany, NY 12208, USA; Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
3
|
Dhulipalla S, Longmate WM. Integrin mutations in blistering skin diseases and related genetically engineered mouse models. Hum Immunol 2024; 85:111175. [PMID: 39532028 PMCID: PMC11637898 DOI: 10.1016/j.humimm.2024.111175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/30/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
As major receptors for cellular adhesion, integrins in the epidermis are critical to maintain skin integrity. Integrins α6β4 and α3β1 are among the most highly and widely expressed integrins in the skin. Perhaps not surprisingly, mutation in subunits associated with these integrins cause variations of a blistering skin disease called junctional epidermolysis bullosa (JEB), which is characterized by blisters that form between the epidermis and dermis of the skin. This review highlights how the differences in structural roles and functions for these epidermal integrins lead to distinct JEB phenotypes resulting from their absence. Additionally, much has been learned by using genetically engineered mouse models, which are featured throughout the review, as they closely resemble the disorders of human patients that harbor analogous mutations.
Collapse
Affiliation(s)
- Sanjana Dhulipalla
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Whitney M Longmate
- Department of Surgery Albany Medical College, Albany, NY 12208, USA; Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
4
|
Shlepova OV, Gornostaeva TY, Kukushkin ID, Azev VN, Bychkov ML, Shenkarev ZO, Kirpichnikov MP, Lyukmanova EN. Peptide Mimicking Loop II of the Human Epithelial Protein SLURP-2 Enhances the Viability and Migration of Skin Keratinocytes. Acta Naturae 2024; 16:86-94. [PMID: 39877008 PMCID: PMC11771840 DOI: 10.32607/actanaturae.27494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/16/2024] [Indexed: 01/31/2025] Open
Abstract
The secreted human protein SLURP-2 is a regulator of epithelial homeostasis, which enhances the viability and migration of keratinocytes. The targets of SLURP-2 in keratinocytes are nicotinic and muscarinic acetylcholine receptors. This work is devoted to the search for the SLURP-2 functional regions responsible for enhancing keratinocyte viability and migration. We produced synthetic peptides corresponding to the SLURP-2 loop regions and studied their effect on the viability and migration of HaCaT skin keratinocytes using the WST-8 test and scratch-test, respectively. The highest activity was exhibited by a loop II-mimicking peptide that enhanced the viability of keratinocytes and stimulated their migration. The peptide activity was mediated by interactions with α7- and α3β2-nAChRs and suppression of the p38 MAPK intracellular signaling pathway. Thus, we obtained new data that explain the mechanisms underlying SLURP-2 regulatory activity and indicate the promise of further research into loop II-mimicking peptides as prototypes of wound healing drugs.
Collapse
Affiliation(s)
- O. V. Shlepova
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
| | - T. Ya. Gornostaeva
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
- Moscow Center for Advanced Studies, Moscow, 123592 Russian Federation
| | - I. D. Kukushkin
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
- Moscow Center for Advanced Studies, Moscow, 123592 Russian Federation
| | - V. N. Azev
- Branch of the Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Pushchino, 142290 Russian Federation
| | - M. L. Bychkov
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
| | - Z. O. Shenkarev
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
- Moscow Center for Advanced Studies, Moscow, 123592 Russian Federation
| | - M. P. Kirpichnikov
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
- Interdisciplinary Scientific and Educational School of Moscow University “Molecular Technologies of the Living Systems and Synthetic Biology”, Department of Biology, Lomonosov Moscow State University, Moscow, 119234 Russian Federation
| | - E. N. Lyukmanova
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russian Federation
- Moscow Center for Advanced Studies, Moscow, 123592 Russian Federation
- Shenzhen MSU–BIT University, Longgang District, Shenzhen, Guangdong Province, 518172 China
- Interdisciplinary Scientific and Educational School of Moscow University “Molecular Technologies of the Living Systems and Synthetic Biology”, Department of Biology, Lomonosov Moscow State University, Moscow, 119234 Russian Federation
| |
Collapse
|
5
|
Miskin RP, DiPersio CM. Roles for epithelial integrin α3β1 in regulation of the microenvironment during normal and pathological tissue remodeling. Am J Physiol Cell Physiol 2024; 326:C1308-C1319. [PMID: 38497112 PMCID: PMC11371326 DOI: 10.1152/ajpcell.00128.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/08/2024] [Accepted: 03/08/2024] [Indexed: 03/19/2024]
Abstract
Integrin receptors for the extracellular matrix activate intracellular signaling pathways that are critical for tissue development, homeostasis, and regeneration/repair, and their loss or dysregulation contributes to many developmental defects and tissue pathologies. This review will focus on tissue remodeling roles for integrin α3β1, a receptor for laminins found in the basement membranes (BMs) that underlie epithelial cell layers. As a paradigm, we will discuss literature that supports a role for α3β1 in promoting ability of epidermal keratinocytes to modify their tissue microenvironment during skin development, wound healing, or tumorigenesis. Preclinical and clinical studies have shown that this role depends largely on ability of α3β1 to govern the keratinocyte's repertoire of secreted proteins, or the "secretome," including 1) matrix proteins and proteases involved in matrix remodeling and 2) paracrine-acting growth factors/cytokines that stimulate other cells with important tissue remodeling functions (e.g., endothelial cells, fibroblasts, inflammatory cells). Moreover, α3β1 signaling controls gene expression that helps epithelial cells carry out these functions, including genes that encode secreted matrix proteins, proteases, growth factors, or cytokines. We will review what is known about α3β1-dependent gene regulation through both transcription and posttranscriptional mRNA stability. Regarding the latter, we will discuss examples of α3β1-dependent alternative splicing (AS) or alternative polyadenylation (APA) that prevents inclusion of cis-acting mRNA sequences that would otherwise target the transcript for degradation via nonsense-mediated decay or destabilizing AU-rich elements (AREs) in the 3'-untranslated region (3'-UTR). Finally, we will discuss prospects and anticipated challenges of exploiting α3β1 as a clinical target for the treatment of cancer or wound healing.
Collapse
Affiliation(s)
| | - C Michael DiPersio
- Department of Surgery, Albany Medical College, Albany, New York, United States
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, United States
| |
Collapse
|
6
|
Longmate WM, Norton E, Duarte GA, Wu L, DiPersio MR, Lamar JM, DiPersio CM. Keratinocyte integrin α3β1 induces expression of the macrophage stimulating factor, CSF-1, through a YAP/TEAD-dependent mechanism. Matrix Biol 2024; 127:48-56. [PMID: 38340968 PMCID: PMC10923166 DOI: 10.1016/j.matbio.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/11/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
The development of wound therapy targeting integrins is hampered by inadequate understanding of integrin function in cutaneous wound healing and the wound microenvironment. Following cutaneous injury, keratinocytes migrate to restore the skin barrier, and macrophages aid in debris clearance. Thus, both keratinocytes and macrophages are critical to the coordination of tissue repair. Keratinocyte integrins have been shown to participate in this coordinated effort by regulating secreted factors, some of which crosstalk to distinct cells in the wound microenvironment. Epidermal integrin α3β1 is a receptor for laminin-332 in the cutaneous basement membrane. Here we show that wounds deficient in epidermal α3β1 express less epidermal-derived macrophage colony-stimulating factor 1 (CSF-1), the primary macrophage-stimulating growth factor. α3β1-deficient wounds also have fewer wound-proximal macrophages, suggesting that keratinocyte α3β1 may stimulate wound macrophages through the regulation of CSF-1. Indeed, using a set of immortalized keratinocytes, we demonstrate that keratinocyte-derived CSF-1 supports macrophage growth, and that α3β1 regulates Csf1 expression through Src-dependent stimulation of Yes-associated protein (YAP)-Transcriptional enhanced associate domain (TEAD)-mediated transcription. Consistently, α3β1-deficient wounds in vivo display a substantially reduced number of keratinocytes with YAP-positive nuclei. Overall, our current findings identify a novel role for epidermal integrin α3β1 in regulating the cutaneous wound microenvironment by mediating paracrine crosstalk from keratinocytes to wound macrophages, implicating α3β1 as a potential target of wound therapy.
Collapse
Affiliation(s)
- Whitney M Longmate
- Department of Surgery, Albany Medical College, Albany, NY 12208, USA; Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Emily Norton
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Giesse Albeche Duarte
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Lei Wu
- Department of Surgery, Albany Medical College, Albany, NY 12208, USA
| | - Mathieu R DiPersio
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - John M Lamar
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - C Michael DiPersio
- Department of Surgery, Albany Medical College, Albany, NY 12208, USA; Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
7
|
Alasmari BG, Alomari M, Alotaibi WN, Hommadi A, Elmugadam AA, Abdalla K, Al-Tala SM. LAD-III, a Mild Phenotype Resulting From a Novel Variant of FERMT3 Gene: A Case Report. Cureus 2023; 15:e51062. [PMID: 38269242 PMCID: PMC10806943 DOI: 10.7759/cureus.51062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/25/2023] [Indexed: 01/26/2024] Open
Abstract
Leukocyte adhesion deficiency-III (LAD-III) is a rare recessive autosomal disorder characterized by bleeding syndrome of Glanzmann-type and life-threatening infections. The main etiology of this condition is variations in the FERMT3 gene, which encodes kindlin-3, an integrin-binding protein. This protein is responsible for the activation of fibrinogen receptors and integrin-mediated hematopoietic cell adhesion. So far, only limited cases of LAD-III have been reported. This case report discusses a two-year-old male infant from the Asir region, Saudi Arabia, who was referred to the pediatric hematology service due to recurrent ecchymosis and epistaxis. He was born at full term with a history of transient tachypnea of the newborn and recurrent bronchiolitis. The patient exhibited normal platelet count and coagulation profiles alongside a familial history of bleeding disorders, including a cousin with a similar condition. The patient also presented with hypospadias and café-au-lait spots. Laboratory findings revealed anemia, microcytosis, and hypochromia indicative of iron deficiency anemia. Whole exome sequencing (WES) identified a homozygous variant of uncertain significance in the FERMT3 gene, associated with autosomal recessive LAD-III. The patient was subsequently referred to an immunology subspecialty for further investigation and bone marrow transplant preparation. This case underscores the importance of comprehensive clinical and genetic evaluations in pediatric patients with unexplained bleeding tendencies.
Collapse
Affiliation(s)
| | - Mohammed Alomari
- Pediatrics, Armed Forces Hospital Southern Region, Khamis Mushait, SAU
| | - Wejdan N Alotaibi
- Pediatrics, Armed Forces Hospital Southern Region, Khamis Mushait, SAU
| | - Ashwaq Hommadi
- Pediatrics, Armed Forces Hospital Southern Region, Khamis Mushait, SAU
| | | | - Khalid Abdalla
- Pediatric Hematology Oncology, King Abdulaziz Medical City, Jeddah, SAU
| | - Saeed M Al-Tala
- Pediatrics, Armed Forces Hospital Southern Region, Khamis Mushait, SAU
| |
Collapse
|
8
|
Liu Z, Du Y, Xu S, Li M, Lu X, Tian G, Ye J, Zhao B, Wei P, Wang Y. Histatin 1-modified SIS hydrogels enhance the sealing of peri-implant mucosa to prevent peri-implantitis. iScience 2023; 26:108212. [PMID: 37965149 PMCID: PMC10641262 DOI: 10.1016/j.isci.2023.108212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 09/15/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023] Open
Abstract
Dental implants make it possible to replace teeth in more sophisticated ways. Nevertheless, peri-implantitis is one of the leading causes of implant failure, which can be avoided with proper soft tissue sealing. The aim of this study was to achieve the promotion of the synthesis of peri-implant epithelial hemidesmosome through Histatin 1 and porcine small intestinal submucosa (SIS) hydrogel to form a good peri-implant seal. The results show that hydrogel can improve the biological barrier function around implants by combining antibacterial, promoting soft tissue healing and promoting epithelial bonding. This means that the morphology and anti-infection ability of soft tissue are enhanced, which ensures the long-term stability of the implant.SIS-Hst1 hydrogel has certain clinical application in the prevention and early treatment of peri-implantitis. In conclusion, Hst1-SIS hydrogel, as a local administration system, provides experimental evidence for the prevention of peri-implant disease.
Collapse
Affiliation(s)
- Zihao Liu
- Zhongnuo Dental Hospital, Tianjin Nankai District, Tianjin 300101, China
| | - Yaqi Du
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 30070, China
| | - Shendan Xu
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 30070, China
| | - Minting Li
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 30070, China
| | - Xuemei Lu
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 30070, China
| | - Guangjie Tian
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 30070, China
| | - Jing Ye
- Department of Stomatology, Tianjin Hospital, Tianjin 300211, China
| | - Bo Zhao
- Beijing Biosis Healing Biological Technology Co., Ltd., Beijing 102600, China
| | - Pengfei Wei
- Beijing Biosis Healing Biological Technology Co., Ltd., Beijing 102600, China
| | - Yonglan Wang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 30070, China
| |
Collapse
|
9
|
Ademi H, Michalak-Micka K, Moehrlen U, Biedermann T, Klar AS. Effects of an Adipose Mesenchymal Stem Cell-Derived Conditioned medium and TGF-β1 on Human Keratinocytes In Vitro. Int J Mol Sci 2023; 24:14726. [PMID: 37834173 PMCID: PMC10572767 DOI: 10.3390/ijms241914726] [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: 08/16/2023] [Revised: 09/06/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Human keratinocytes play a crucial role during skin wound healing and in skin replacement therapies. The secretome of adipose-derived stem cells (ASCs) has been shown to secrete pro-healing factors, among which include TGF-β1, which is essential for keratinocyte migration and the re-epithelialization of cutaneous wounds during skin wound healing. The benefits of an ASC conditioned medium (ASC-CM) are primarily orchestrated by trophic factors that mediate autocrine and paracrine effects in keratinocytes. Here, we evaluated the composition and the innate characteristics of the ASC secretome and its biological effects on keratinocyte maturation and wound healing in vitro. In particular, we detected high levels of different growth factors, such as HGF, FGFb, and VEGF, and other factors, such as TIMP1 and 4, IL8, PAI-1, uPA, and IGFBP-3, in the ASC-CM. Further, we investigated, using immunofluorescence and flow cytometry, the distinct effects of a human ASC-CM and/or synthetic TGF-β1 on human keratinocyte proliferation, migration, and cell apoptosis suppression. We demonstrated that the ASC-CM increased keratinocyte proliferation as compared to TGF-β1 treatment. Further, we found that the ASC-CM exerted cell cycle progression in keratinocytes via regulating the phases G1, S, and G2/M. In particular, cells subjected to the ASC-CM demonstrated increased DNA synthesis (S phase) compared to the TGF-β1-treated KCs, which showed a pronounced G0/G1 phase. Furthermore, both the ASC-CM and TGF-β1 conditions resulted in a decreased expression of the late differentiation marker CK10 in human keratinocytes in vitro, whereas both treatments enhanced transglutaminase 3 and loricrin expression. Interestingly, the ASC-CM promoted significantly increased numbers of keratinocytes expressing epidermal basal keratinocyte markers, such DLL1 and Jagged2 Notch ligands, whereas those ligands were significantly decreased in TGF-β1-treated keratinocytes. In conclusion, our findings suggest that the ASC-CM is a potent stimulator of human keratinocyte proliferation in vitro, particularly supporting basal keratinocytes, which are crucial for a successful skin coverage after transplantation. In contrast, TGF-β1 treatment decreased keratinocyte proliferation and specifically increased the expression of differentiation markers in vitro.
Collapse
Affiliation(s)
- Hyrije Ademi
- Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, 8952 Schlieren, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Faculty of Medicine, University of Zurich, 8032 Zurich, Switzerland
| | - Katarzyna Michalak-Micka
- Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, 8952 Schlieren, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Faculty of Medicine, University of Zurich, 8032 Zurich, Switzerland
| | - Ueli Moehrlen
- Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, 8952 Schlieren, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Faculty of Medicine, University of Zurich, 8032 Zurich, Switzerland
- Department of Surgery, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, 8952 Schlieren, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Faculty of Medicine, University of Zurich, 8032 Zurich, Switzerland
| | - Agnes S. Klar
- Tissue Biology Research Unit, Department of Surgery, University Children’s Hospital Zurich, 8952 Schlieren, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Faculty of Medicine, University of Zurich, 8032 Zurich, Switzerland
| |
Collapse
|
10
|
Salois MN, Gugger JA, Webb S, Sheldon CE, Parraga SP, Lewitt GM, Grange DK, Koch PJ, Koster MI. Effects of TP63 mutations on keratinocyte adhesion and migration. Exp Dermatol 2023; 32:1575-1581. [PMID: 37432020 PMCID: PMC10529328 DOI: 10.1111/exd.14885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/12/2023] [Accepted: 06/29/2023] [Indexed: 07/12/2023]
Abstract
The goal of this study was to investigate the molecular mechanisms responsible for the formation of skin erosions in patients affected by Ankyloblepharon-ectodermal defects-cleft lip/palate syndrome (AEC). This ectodermal dysplasia is caused by mutations in the TP63 gene, which encodes several transcription factors that control epidermal development and homeostasis. We generated induced pluripotent stem cells (iPSC) from AEC patients and corrected the TP63 mutations using genome editing tools. Three pairs of the resulting conisogenic iPSC lines were differentiated into keratinocytes (iPSC-K). We identified a significant downregulation of key components of hemidesmosomes and focal adhesions in AEC iPSC-K compared to their gene-corrected counterparts. Further, we demonstrated reduced AEC iPSC-K migration, suggesting the possibility that a process critical for cutaneous wound healing might be impaired in AEC patients. Next, we generated chimeric mice expressing a TP63-AEC transgene and confirmed a downregulation of these genes in transgene-expressing cells in vivo. Finally, we also observed these abnormalities in AEC patient skin. Our findings suggest that integrin defects in AEC patients might weaken the adhesion of keratinocytes to the basement membrane. We propose that reduced expression of extracellular matrix adhesion receptors, potentially in conjunction with previously identified desmosomal protein defects, contribute to skin erosions in AEC.
Collapse
Affiliation(s)
- Maddison N. Salois
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Jessica A. Gugger
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Saiphone Webb
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Christina E. Sheldon
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Shirley P. Parraga
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC
| | | | - Dorothy K. Grange
- Division of Genetics and Genomic Medicine, Department of Pediatrics, St. Louis Children’s Hospital, Washington University School of Medicine, St. Louis, MO
| | - Peter J. Koch
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Maranke I. Koster
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC
| |
Collapse
|
11
|
Salois MN, Gugger JA, Webb S, Sheldon CE, Parraga SP, Lewitt GM, Grange DK, Koch PJ, Koster MI. Effects of TP63 Mutations on Keratinocyte Adhesion and Migration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.04.539104. [PMID: 37205354 PMCID: PMC10187256 DOI: 10.1101/2023.05.04.539104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The goal of this study was to investigate the molecular mechanisms responsible for the formation of skin erosions in patients affected by Ankyloblepharon-ectodermal defects-cleft lip/palate syndrome (AEC). This ectodermal dysplasia is caused by mutations in the TP63 gene, which encodes several transcription factors that control epidermal development and homeostasis. We generated induced pluripotent stem cells (iPSC) from AEC patients and corrected the TP63 mutations using genome editing tools. Three pairs of the resulting conisogenic iPSC lines were differentiated into keratinocytes (iPSC-K). We identified a significant downregulation of key components of hemidesmosomes and focal adhesions in AEC iPSC-K compared to their gene-corrected counterparts. Further, we demonstrated reduced iPSC-K migration, suggesting the possibility that a process critical for cutaneous wound healing might be impaired in AEC patients. Next, we generated chimeric mice expressing a TP63-AEC transgene and confirmed a downregulation of these genes in transgene-expressing cells in vivo. Finally, we also observed these abnormalities in AEC patient skin. Our findings suggest that integrin defects in AEC patients might weaken the adhesion of keratinocytes to the basement membrane. We propose that reduced expression of extracellular matrix adhesion receptors, potentially in conjunction with previously identified desmosomal protein defects, contribute to skin erosions in AEC.
Collapse
|
12
|
Bakhshandeh B, Jahanafrooz Z, Allahdadi S, Daryani S, Dehghani Z, Sadeghi M, Pedram MS, Dehghan MM. Transcriptomic and in vivo approaches introduced human iPSC-derived microvesicles for skin rejuvenation. Sci Rep 2023; 13:9963. [PMID: 37339980 DOI: 10.1038/s41598-023-36162-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 05/30/2023] [Indexed: 06/22/2023] Open
Abstract
The skin undergoes the formation of fine lines and wrinkles through the aging process; also, burns, trauma, and other similar circumstances give rise to various forms of skin ulcers. Induced pluripotent stem cells (iPSCs) have become promising candidates for skin healing and rejuvenation due to not stimulating inflammatory responses, low probability of immune rejection, high metabolic activity, good large-scale production capacity and potentials for personalized medicine. iPSCs can secrete microvesicles (MVs) containing RNA and proteins responsible for the normal repairing process of the skin. This study aimed to evaluate the possibility, safety and effectiveness of applying iPSCs-derived MVs for skin tissue engineering and rejuvenation applications. The possibility was assessed using the evaluation of the mRNA content of iPSC-derived MVs and the behavior of fibroblasts after MV treatment. Investigating the effect of microvesicle on stemness potential of mesenchymal stem cells was performed for safety concerns. In vivo evaluation of MVs was done in order to investigate related immune response, re-epithelialization and blood vessel formation to measure effectiveness. Shedding MVs were round in shape distributed in the range from 100 to 1000 nm in diameter and positive for AQP3, COL2A, FGF2, ITGB, and SEPTIN4 mRNAs. After treating dermal fibroblasts with iPSC-derived MVs, the expressions of collagens Iα1 and III transcripts (as the main fibrous extracellular matrix (ECM) proteins) were upregulated. Meanwhile, the survival and proliferation of MV treated fibroblasts did not change significantly. Evaluation of stemness markers in MV treated MSCs showed negligible alteration. In line with in vitro results, histomorphometry and histopathology findings also confirmed the helpful effect of MVs in skin regeneration in the rat burn wound models. Conducting more investigations on hiPSCs-derived MVs may lead to produce more efficient and safer biopharmaceutics for skin regeneration in the pharmaceutical market.
Collapse
Affiliation(s)
- Behnaz Bakhshandeh
- Department of Biotechnology, College of Science, University of Tehran, P.O. Box 14155-6455, Tehran, Iran.
| | - Zohreh Jahanafrooz
- Department of Biology, Faculty of Sciences, University of Maragheh, Maragheh, Iran
| | - Shiva Allahdadi
- Department of Cellular and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Shiva Daryani
- Department of Cellular and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Zahra Dehghani
- Department of Biotechnology, College of Science, University of Tehran, P.O. Box 14155-6455, Tehran, Iran
| | - Mahya Sadeghi
- Department of Biomedical Engineering, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mir Sepehr Pedram
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
- Institute of Biomedical Research, University of Tehran, Tehran, Iran
| | - Mohammad Mehdi Dehghan
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
- Institute of Biomedical Research, University of Tehran, Tehran, Iran
| |
Collapse
|
13
|
Lee HR, Kang SU, Kim HJ, Ji EJ, Yun JH, Kim S, Jang JY, Shin YS, Kim CH. Liquid plasma as a treatment for cutaneous wound healing through regulation of redox metabolism. Cell Death Dis 2023; 14:119. [PMID: 36781835 PMCID: PMC9925775 DOI: 10.1038/s41419-023-05610-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/13/2023] [Accepted: 01/20/2023] [Indexed: 02/15/2023]
Abstract
The skin functions as the outermost protective barrier to the internal organs and major vessels; thus, delayed regeneration from acute injury could induce serious clinical complications. For rapid recovery of skin wounds, promoting re-epithelialization of the epidermis at the initial stage of injury is essential, wherein epithelial keratinocytes act as leading cells via migration. This study applied plasma technology, which has been known to enable wound healing in the medical field. Through in vitro and in vivo experiments, the study elucidated the effect and molecular mechanism of the liquid plasma (LP) manufactured by our microwave plasma system, which was found to improve the applicability of existing gas-type plasma on skin cell migration for re-epithelialization. LP treatment promoted the cytoskeletal transformation of keratinocytes and migration owing to changes in the expression of integrin-dependent focal adhesion molecules and matrix metalloproteinases (MMPs). This study also identified the role of increased levels of intracellular reactive oxygen species (ROS) as a driving force for cell migration activation, which was regulated by changes in NADPH oxidases and mitochondrial membrane potential. In an in vivo experiment using a murine dorsal full-thickness acute skin wound model, LP treatment helped improve the re-epithelialization rate, reaffirming the activation of the underlying intracellular ROS-dependent integrin-dependent signaling molecules. These findings indicate that LP could be a valuable wound management material that can improve the regeneration potential of the skin via the activation of migration-related molecular signaling within the epithelial cell itself with plasma-driven oxidative eustress.
Collapse
Affiliation(s)
- Hye Ran Lee
- Department of Otolaryngology-Head and Neck Surgery, Catholic Kwandong University International St. Mary's Hospital, Incheon, 22711, Republic of Korea
- Department of Medical Sciences, Otolaryngology, Graduate School of Ajou University, Suwon, 16499, Republic of Korea
| | - Sung Un Kang
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Haeng Jun Kim
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Eun Jong Ji
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Ju Hyun Yun
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Sungryeal Kim
- Department of Otolaryngology, College of Medicine, Inha University, Incheon, 22332, Republic of Korea
| | - Jeon Yeob Jang
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Yoo Seob Shin
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Chul-Ho Kim
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea.
| |
Collapse
|
14
|
Schmidt A, da Silva Brito WA, Singer D, Mühl M, Berner J, Saadati F, Wolff C, Miebach L, Wende K, Bekeschus S. Short- and long-term polystyrene nano- and microplastic exposure promotes oxidative stress and divergently affects skin cell architecture and Wnt/beta-catenin signaling. Part Fibre Toxicol 2023; 20:3. [PMID: 36647127 PMCID: PMC9844005 DOI: 10.1186/s12989-023-00513-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 01/05/2023] [Indexed: 01/18/2023] Open
Abstract
Nano- and microplastic particles (NMP) are strong environmental contaminants affecting marine ecosystems and human health. The negligible use of biodegradable plastics and the lack of knowledge about plastic uptake, accumulation, and functional consequences led us to investigate the short- and long-term effects in freshly isolated skin cells from mice. Using fluorescent NMP of several sizes (200 nm to 6 µm), efficient cellular uptake was observed, causing, however, only minor acute toxicity as metabolic activity and apoptosis data suggested, albeit changes in intracellular reactive species and thiol levels were observed. The internalized NMP induced an altered expression of various targets of the nuclear factor-2-related transcription factor 2 pathway and were accompanied by changed antioxidant and oxidative stress signaling responses, as suggested by altered heme oxygenase 1 and glutathione peroxide 2 levels. A highly increased beta-catenin expression under acute but not chronic NMP exposure was concomitant with a strong translocation from membrane to the nucleus and subsequent transcription activation of Wnt signaling target genes after both single-dose and chronic long-term NMP exposure. Moreover, fibroblast-to-myofibroblast transdifferentiation accompanied by an increase of α smooth muscle actin and collagen expression was observed. Together with several NMP-induced changes in junctional and adherence protein expression, our study for the first time elucidates the acute and chronic effects of NMP of different sizes in primary skin cells' signaling and functional biology, contributing to a better understanding of nano- and microplastic to health risks in higher vertebrates.
Collapse
Affiliation(s)
- Anke Schmidt
- grid.461720.60000 0000 9263 3446ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Walison Augusto da Silva Brito
- grid.461720.60000 0000 9263 3446ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany ,grid.411400.00000 0001 2193 3537Department of General Pathology, State University of Londrina, Rodovia Celso Garcia Cid, Londrina, Brazil
| | - Debora Singer
- grid.461720.60000 0000 9263 3446ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Melissa Mühl
- grid.461720.60000 0000 9263 3446ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Julia Berner
- grid.461720.60000 0000 9263 3446ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany ,grid.5603.0Department Oral, Maxillofacial, and Plastic Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., Greifswald, Germany
| | - Fariba Saadati
- grid.461720.60000 0000 9263 3446ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Christina Wolff
- grid.461720.60000 0000 9263 3446ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Lea Miebach
- grid.461720.60000 0000 9263 3446ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany ,grid.5603.0Department of General, Visceral, Thoracic, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., Greifswald, Germany
| | - Kristian Wende
- grid.461720.60000 0000 9263 3446ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Sander Bekeschus
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany.
| |
Collapse
|
15
|
Liang W, Ma JX, Van L, Vasini B, Karamichos D. Prolactin-Induced Protein facilitates corneal wound healing. Exp Eye Res 2022; 225:109300. [PMID: 36328302 DOI: 10.1016/j.exer.2022.109300] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/06/2022] [Accepted: 10/25/2022] [Indexed: 12/29/2022]
Abstract
The purpose of the study was to investigate the role of Prolactin-Induced Protein (PIP) in corneal wound healing, in vivo and in vitro. In C57BL/6J mice, corneal epithelia was removed using an ocular burr. Phosphate buffered saline (PBS) or PIP (0.5 and 1.0 μg/mL) was applied topically or subconjunctivally injected. PIP accelerated wound closure as early as 24 h. PIP treatment promoted corneal wound healing and epithelial integrity and thickness. Integrin α6, integrin β4, Thrombospondin-1, and TGF-β1 expressions were all downregulated by PIP after wound closure. In vitro, scratch assays were performed using primary human epithelial cells (HCECs) and human corneal fibroblasts (HCFs), stimulated with PIP at various dosages. PIP treatment promoted both HCECs and HCFs migration. PIP upregulated expression of integrin α6, integrin β4, and Thrombospondin-1 in HCECs. Expression of TGF-β1 in HCECs and expression of smooth muscle actin (SMA) and Type III Collagen (Col III) in HCFs were significantly downregulated at 150 ng/mL PIP. PIP exhibits noteworthy anti-fibrotic potentiality. While the mechanism of how PIP is impactful on the corneal wound healing cascade is unknown, our findings are novel and further studies are warranted in order to unravel any therapeutic potential.
Collapse
Affiliation(s)
- Wentao Liang
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jian-Xing Ma
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Lyly Van
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Brenda Vasini
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Dimitrios Karamichos
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA; Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA.
| |
Collapse
|
16
|
Frommherz LH, Sayar SB, Wang Y, Trefzer LK, He Y, Leppert J, Eßer P, Has C. Integrin α3 negative podocytes: A gene expression study. Matrix Biol Plus 2022; 16:100119. [PMID: 36060790 PMCID: PMC9429797 DOI: 10.1016/j.mbplus.2022.100119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 07/30/2022] [Accepted: 08/07/2022] [Indexed: 12/04/2022] Open
Abstract
New cell model to investigate the impact of loss of integrin α3 in podocytes. In this novel model, genes of the extracellular matrix and adhesome are mostly downregulated. Loss of integrin α3 results in changes of cell adhesion and spreading.
Integrin α3β1 is a cell adhesion receptor widely expressed in epithelial cells. Pathogenic variants in the gene encoding the integrin α3 subunit ITGA3 lead to a syndrome including interstitial lung disease, nephrotic syndrome, and epidermolysis bullosa (ILNEB). Renal involvement mainly consists of glomerular disease caused by loss of adhesion between podocytes and the glomerular basement membrane. The aim of this study was to characterize the impact of loss of integrin α3 on human podocytes. ITGA3 was stably knocked-out in the human podocyte cell line AB8/13, designated as PodoA3−, and in human proximal tubule epithelial cell line HK2 using the targeted genome editing technique CRISPR/Cas9. Cell clones were characterized by Sanger sequencing, quantitative PCR, Western Blot and immunofluorescence staining. RNASeq of integrin α3 negative cells and controls was performed to identify differential gene expression patterns. Differentiated PodoA3− did not substantially change morphology and adhesion under standard culture conditions, but displayed significantly reduced spreading and adhesion when seed on laminin 511 in serum free medium. Gene expression studies demonstrated a distinct dysregulation of the adhesion network with downregulation of most integrin α3 interaction partners. In agreement with this, biological processes such as “extracellular matrix organization” and “cell differentiation” as well as KEGG pathways such as “ECM-receptor interaction”, “focal adhesion” and the “PI3K-Akt signaling pathway” were significantly downregulated in human podocytes lacking the integrin α3 subunit.
Collapse
Affiliation(s)
- L H Frommherz
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany.,Department of Dermatology and Allergology, University Hospital, LMU Munich, Germany
| | - S B Sayar
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Y Wang
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - L K Trefzer
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Y He
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - J Leppert
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - P Eßer
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| | - C Has
- Department of Dermatology, Medical Center - University of Freiburg, Freiburg, Germany
| |
Collapse
|
17
|
Tuned Gum ghatti and pectin for green synthesis of novel wound dressing material: Engineering aspects and in vivo study. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
18
|
Lim JH, Kim DH, Noh KH, Jung CR, Kang HM. The proliferative and multipotent epidermal progenitor cells for human skin reconstruction in vitro and in vivo. Cell Prolif 2022; 55:e13284. [PMID: 35723171 PMCID: PMC9436902 DOI: 10.1111/cpr.13284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/16/2022] [Accepted: 05/23/2022] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES The skin exhibits tremendous regenerative potential, as different types of progenitor and stem cells regulate skin homeostasis and damage. However, in vitro primary keratinocytes present with several drawbacks, such as high donor variability, short lifespan, and limited donor tissue availability. Therefore, more stable primary keratinocytes are needed to generate multiple uniform in vitro and in vivo skin models. RESULTS We identified epidermal progenitor cells from primary keratinocytes using Integrin beta 1 (ITGB1) an epidermal stem cell marker markedly decreased after senescence in vitro. Epidermal progenitor cells exhibited unlimited proliferation and the potential for multipotent differentiation capacity. Moreover, they could completely differentiate to form an organotypic skin model including conversed mesenchymal cells in the dermis and could mimic the morphologic and biochemical processes of human epidermis. We also discovered that proliferation and the multipotent differentiation capacity of these cells relied on ITGB1 expression. Eventually, we examined the in vitro and in vivo wound healing capacity of these epidermal progenitor cells. CONCLUSIONS Overall, the findings suggest that these stable and reproducible cells can differentiate into multiple lineages, including human skin models. They are a potentially powerful tool for studying skin regeneration, skin diseases, and are an alternative for in vivo experiments.
Collapse
Affiliation(s)
- Jung Hwa Lim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Dae Hun Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Department of Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Kyung Hee Noh
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Cho-Rok Jung
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Department of Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Hyun Mi Kang
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| |
Collapse
|
19
|
Picker J, Lan Z, Arora S, Green M, Hahn M, Cosgriff-Hernandez E, Hook M. Prokaryotic Collagen-Like Proteins as Novel Biomaterials. Front Bioeng Biotechnol 2022; 10:840939. [PMID: 35372322 PMCID: PMC8968730 DOI: 10.3389/fbioe.2022.840939] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/10/2022] [Indexed: 12/13/2022] Open
Abstract
Collagens are the major structural component in animal extracellular matrices and are critical signaling molecules in various cell-matrix interactions. Its unique triple helical structure is enabled by tripeptide Gly-X-Y repeats. Understanding of sequence requirements for animal-derived collagen led to the discovery of prokaryotic collagen-like protein in the early 2000s. These prokaryotic collagen-like proteins are structurally similar to mammalian collagens in many ways. However, unlike the challenges associated with recombinant expression of mammalian collagens, these prokaryotic collagen-like proteins can be readily expressed in E. coli and are amenable to genetic modification. In this review article, we will first discuss the properties of mammalian collagen and provide a comparative analysis of mammalian collagen and prokaryotic collagen-like proteins. We will then review the use of prokaryotic collagen-like proteins to both study the biology of conventional collagen and develop a new biomaterial platform. Finally, we will describe the application of Scl2 protein, a streptococcal collagen-like protein, in thromboresistant coating for cardiovascular devices, scaffolds for bone regeneration, chronic wound dressing and matrices for cartilage regeneration.
Collapse
Affiliation(s)
- Jonathan Picker
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M, Houston, TX, United States
| | - Ziyang Lan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Srishtee Arora
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M, Houston, TX, United States
| | - Mykel Green
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Mariah Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| | | | - Magnus Hook
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M, Houston, TX, United States
| |
Collapse
|
20
|
Yazarlu O, Iranshahi M, Kashani HRK, Reshadat S, Habtemariam S, Iranshahy M, Hasanpour M. Perspective on the application of medicinal plants and natural products in wound healing: A mechanistic review. Pharmacol Res 2021; 174:105841. [PMID: 34419563 DOI: 10.1016/j.phrs.2021.105841] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/14/2021] [Accepted: 08/17/2021] [Indexed: 12/14/2022]
Abstract
Wound is defined as any injury to the body such as damage to the epidermis of the skin and disturbance to its normal anatomy and function. Since ancient times, the importance of wound healing has been recognized, and many efforts have been made to develop novel wound dressings made of the best material for rapid and effective wound healing. Medicinal plants play a great role in the wound healing process. In recent decades, many studies have focused on the development of novel wound dressings that incorporate medicinal plant extracts or their purified active compounds, which are potential alternatives to conventional wound dressings. Several studies have also investigated the mechanism of action of various herbal medicines in wound healing process. This paper attempts to highlight and review the mechanistic perspective of wound healing mediated by plant-based natural products. The findings showed that herbal medicines act through multiple mechanisms and are involved in various stages of wound healing. Some herbal medicines increase the expression of vascular endothelial growth factor (VEGF) and transforming growth factor-β (TGF-β) which play important role in stimulation of re-epithelialization, angiogenesis, formation of granulation tissue, and collagen fiber deposition. Some other wound dressing containing herbal medicines act as inhibitor of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and inducible nitric oxide synthase (iNOS) protein expression thereby inducing antioxidant and anti-inflammatory properties in various phases of the wound healing process. Besides the growing public interest in traditional and alternative medicine, the use of herbal medicine and natural products for wound healing has many advantages over conventional medicines, including greater effectiveness due to diverse mechanisms of action, antibacterial activity, and safety in long-term wound dressing usage.
Collapse
Affiliation(s)
- Omid Yazarlu
- Mashhad University of Medical Sciences, Department of General Surgery, Mashhad, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Sara Reshadat
- Department of Internal Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Solomon Habtemariam
- Pharmacognosy Research Laboratories and Herbal Analysis Services UK, University of Greenwich, Central Avenue, Chatham-Maritime, Kent ME4 4TB, UK
| | - Milad Iranshahy
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Maede Hasanpour
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
21
|
Lin BQ, Zhang WB, Zhao J, Zhou XH, Li YJ, Deng J, Zhao Q, Fu G, Xie CM, Xu YK, Feng GK. An Optimized Integrin α6-Targeted Magnetic Resonance Probe for Molecular Imaging of Hepatocellular Carcinoma in Mice. J Hepatocell Carcinoma 2021; 8:645-656. [PMID: 34235103 PMCID: PMC8244641 DOI: 10.2147/jhc.s312921] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/20/2021] [Indexed: 12/25/2022] Open
Abstract
Introduction Integrin α6 is an attractive diagnostic biomarker for molecular imaging of hepatocellular carcinoma (HCC) as it has an extremely high positive rate (approximately 94%) in clinical early-stage HCC. In this study, based on our previously identified integrin α6-targeted peptide, we developed an optimized integrin α6-targeted magnetic resonance (MR) probe dubbed DOTA(Gd)-ANADYWR for MR imaging of HCC in mice. Materials and Methods The longitudinal (R1) relaxivity of DOTA(Gd)-ANADYWR was measured on a 3.0 T MR system . The specific tumor enhancement of the agent was investigated in four distinct mouse models, including subcutaneous, orthotopic, genetically engineered and chemically induced HCC mice. Results The R1 relaxivity value of DOTA(Gd)-ANADYWR is 5.11 mM−1s−1 at 3.0 T, which is similar to that of the nonspecific clinical agent Gadoteridol. DOTA(Gd)-ANADYWR generated superior enhanced MR signal in HCC lesions and provided complementary enhancement MR signals to the clinically available hepatobiliary MR contrast agent gadoxetate disodium (Gd-EOB-DTPA). Importantly, DOTA(Gd)-ANADYWR could efficiently visualize small HCC lesion (approximately 1 mm) which was hardly detected by the clinical Gd-EOB-DTPA. Conclusion These findings suggest the potential application of this integrin α6-targeted MR probe for the detection of HCC, particularly for small HCC.
Collapse
Affiliation(s)
- Bing-Quan Lin
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou City, Guangdong Province, 510515, People's Republic of China
| | - Wen-Biao Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Medical Imaging, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Jing Zhao
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Xu-Hui Zhou
- Department of Radiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, People's Republic of China
| | - Yong-Jiang Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Jun Deng
- Department of Biological Products, Guangdong Institute for Drug Control, Guangzhou, 510663, People's Republic of China
| | - Qin Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Medical Imaging, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Gui Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China.,Department of Medical Imaging, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Chuan-Miao Xie
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Yi-Kai Xu
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou City, Guangdong Province, 510515, People's Republic of China
| | - Guo-Kai Feng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| |
Collapse
|
22
|
Qu H, Miao T, Wang Y, Tan L, Huang B, Zhang L, Liu X, Long M, Zhang R, Liao X, Gong X, Wang J, Xiong X, Liu J, Li X, Yu J, Yang G, Zhu Z, Zheng H, Zheng Y. Dedicator of Cytokinesis 5 Regulates Keratinocyte Function and Promotes Diabetic Wound Healing. Diabetes 2021; 70:1170-1184. [PMID: 33627322 PMCID: PMC8173801 DOI: 10.2337/db20-1008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 02/12/2021] [Indexed: 02/06/2023]
Abstract
Cutaneous wound healing is a fundamental biologic and coordinated process, and failure to maintain this process contributes to the dysfunction of tissue homeostasis, increasing the global burden of diabetic foot ulcerations. However, the factors that mediate this process are not fully understood. Here, we identify the pivotal role of dedicator of cytokinesis 5 (Dock5) in keratinocyte functions contributing to the process of skin wound healing. Specifically, Dock5 is highly upregulated during the proliferative phase of wound repair and is predominantly expressed in epidermal keratinocytes. It regulates keratinocyte adhesion, migration, and proliferation and influences the functions of extracellular matrix (ECM) deposition by facilitating the ubiquitination of transcription factor ZEB1 to activate laminin-332/integrin signaling. Genetic ablation of Dock5 in mice leads to attenuated reepithelialization and granulation tissue formation, and Dock5 overexpression-improved skin repair can be abrogated by LAMA3 knockdown. Importantly, Dock5 expression in the skin edge is reduced in patients and animal models of diabetes, further suggesting a direct correlation between its abundance and healing capability. The rescue of Dock5 expression in diabetic mice causes a significant improvement in reepithelialization, collagen deposition, ECM production, and granulation. Our study provides a potential therapeutic target for wound healing impairment during diabetes.
Collapse
Affiliation(s)
- Hua Qu
- Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, Department of Endocrinology, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Tian Miao
- Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, Department of Endocrinology, Second Affiliated Hospital of Army Medical University, Chongqing, China
- Department of Respiratory and Critical Care Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Yuren Wang
- Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, Department of Endocrinology, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Liang Tan
- Department of Neurosurgery, Southwest Hospital, Army Medical University, Chongqing, China
- Department of Electrical and Computer Engineering, Faculty of Science and Technology, University of Macau, Macau, China
| | - Bangliang Huang
- Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, Department of Endocrinology, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Linlin Zhang
- Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, Department of Endocrinology, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xiufei Liu
- Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, Department of Endocrinology, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Min Long
- Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, Department of Endocrinology, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Rui Zhang
- Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, Department of Endocrinology, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xiaoyu Liao
- Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, Department of Endocrinology, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Xiaoli Gong
- Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, Department of Endocrinology, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Ju Wang
- Department of Neurosurgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Xin Xiong
- Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, Department of Endocrinology, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Junli Liu
- Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xi Li
- Biology Science Institutes, Chongqing Medical University, Chongqing, China
| | - Jiang Yu
- Department of Outpatient, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Gangyi Yang
- Department of Endocrinology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Third Affiliated Hospital of Army Medical University, Chongqing, China
| | - Hongting Zheng
- Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, Department of Endocrinology, Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Yi Zheng
- Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, Department of Endocrinology, Second Affiliated Hospital of Army Medical University, Chongqing, China
| |
Collapse
|
23
|
Integrin α3β1 Promotes Invasive and Metastatic Properties of Breast Cancer Cells through Induction of the Brn-2 Transcription Factor. Cancers (Basel) 2021; 13:cancers13030480. [PMID: 33513758 PMCID: PMC7866210 DOI: 10.3390/cancers13030480] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Metastatic triple-negative breast cancer (TNBC) is highly lethal with limited therapy options. Integrin α3β1 is a cell surface receptor that interacts with the extracellular matrix and facilitates communication between tumor cells and their microenvironment. α3β1 is implicated in breast cancer progression and metastasis, so understanding mechanisms by which α3β1 promotes invasion and metastasis will facilitate the development of this integrin as a potential therapeutic target. Here we identify a novel role for α3β1 in promoting the expression of the transcription factor Brain-2 (Brn-2) in triple-negative breast cancer cells. We further report that Brn-2 promotes invasion and metastasis and partially restores invasion to cells in which expression of α3β1 has been suppressed. Bioinformatic analysis of patient datasets revealed a positive correlation between the expression of the genes encoding the integrin α3 subunit and Brn-2. In summary, our work identifies α3β1-mediated induction of Brn-2 as a mechanism that regulates invasive and metastatic properties of breast cancer cells. Abstract In the current study, we demonstrate that integrin α3β1 promotes invasive and metastatic traits of triple-negative breast cancer (TNBC) cells through induction of the transcription factor, Brain-2 (Brn-2). We show that RNAi-mediated suppression of α3β1 in MDA-MB-231 cells caused reduced expression of Brn-2 mRNA and protein and reduced activity of the BRN2 gene promoter. In addition, RNAi-targeting of Brn-2 in MDA-MB-231 cells decreased invasion in vitro and lung colonization in vivo, and exogenous Brn-2 expression partially restored invasion to cells in which α3β1 was suppressed. α3β1 promoted phosphorylation of Akt in MDA-MB-231 cells, and treatment of these cells with a pharmacological Akt inhibitor (MK-2206) reduced both Brn-2 expression and cell invasion, indicating that α3β1-Akt signaling contributes to Brn-2 induction. Analysis of RNAseq data from patients with invasive breast carcinoma revealed that high BRN2 expression correlates with poor survival. Moreover, high BRN2 expression positively correlates with high ITGA3 expression in basal-like breast cancer, which is consistent with our experimental findings that α3β1 induces Brn-2 in TNBC cells. Together, our study demonstrates a pro-invasive/pro-metastatic role for Brn-2 in breast cancer cells and identifies a role for integrin α3β1 in regulating Brn-2 expression, thereby revealing a novel mechanism of integrin-dependent breast cancer cell invasion.
Collapse
|
24
|
Ndoye A, Miskin RP, DiPersio CM. Integrin α3β1 Represses Reelin Expression in Breast Cancer Cells to Promote Invasion. Cancers (Basel) 2021; 13:cancers13020344. [PMID: 33477804 PMCID: PMC7832892 DOI: 10.3390/cancers13020344] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Breast cancer remains the second leading cause of cancer-related deaths in women, and about 1 in 8 women in the United States develops invasive breast cancer in her lifetime. Integrin α3β1 has been linked to breast cancer progression, but mechanisms whereby it promotes tumor invasion remain unclear. The goal of our study was to determine how α3β1 drives invasion, towards exploiting this integrin as a therapeutic target for breast cancer. We found that α3β1 represses the expression of Reelin, a secreted glycoprotein that inhibits invasion and for which loss of expression is associated with poor prognosis in breast cancer. We also show that increased Reelin expression following RNAi-mediated suppression of α3β1 causes a significant decrease in breast cancer cell invasion. Our findings demonstrate a critical role for α3β1 in promoting cell invasion through repression of Reelin, highlighting the potential value of this integrin as a therapeutic target for breast cancer. Abstract Integrin α3β1, a cell adhesion receptor for certain laminins, is known to promote breast tumor growth and invasion. Our previous gene microarray study showed that the RELN gene, which encodes the extracellular glycoprotein Reelin, was upregulated in α3β1-deficient (i.e., α3 knockdown) MDA-MB-231 cells. In breast cancer, reduced RELN expression is associated with increased invasion and poor prognosis. In this study we demonstrate that α3β1 represses RELN expression to enhance breast cancer cell invasion. RELN mRNA was significantly increased upon RNAi-mediated α3 knockdown in two triple-negative breast cancer cell lines, MDA-MB-231 and SUM159. Modulation of baseline Reelin levels altered invasive potential, where enhanced Reelin expression in MDA-MB-231 cells reduced invasion, while RNAi-mediated suppression of Reelin in SUM159 cells increased invasion. Moreover, treatment of α3β1-expressing MDA-MB-231 cells with culture medium that was conditioned by α3 knockdown MDA-MB-231 cells led to decreased invasion. RNAi-mediated suppression of Reelin in α3 knockdown MDA-MB-231 cells mitigated this effect of conditioned-medium, identifying secreted Reelin as an inhibitor of cell invasion. These results demonstrate a novel role for α3β1 in repressing Reelin in breast cancer cells to promote invasion, supporting this integrin as a potential therapeutic target.
Collapse
Affiliation(s)
- Abibatou Ndoye
- Department of Surgery, Albany Medical College, Albany, 12208 NY, USA;
| | | | - C. Michael DiPersio
- Department of Surgery, Albany Medical College, Albany, 12208 NY, USA;
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, 12208 NY, USA
- Correspondence:
| |
Collapse
|
25
|
Schmidt A, Liebelt G, Nießner F, von Woedtke T, Bekeschus S. Gas plasma-spurred wound healing is accompanied by regulation of focal adhesion, matrix remodeling, and tissue oxygenation. Redox Biol 2021; 38:101809. [PMID: 33271456 PMCID: PMC7710641 DOI: 10.1016/j.redox.2020.101809] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/05/2022] Open
Abstract
In response to injury, efficient migration of skin cells to rapidly close the wound and restore barrier function requires a range of coordinated processes in cell spreading and migration. Gas plasma technology produces therapeutic reactive species that promote skin regeneration by driving proliferation and angiogenesis. However, the underlying molecular mechanisms regulating gas plasma-aided cell adhesion and matrix remodeling essential for wound closure remain elusive. Here, we combined in vitro analyses in primary dermal fibroblasts isolated from murine skin with in vivo studies in a murine wound model to demonstrate that gas plasma treatment changed phosphorylation of signaling molecules such as focal adhesion kinase and paxillin α in adhesion-associated complexes. In addition to cell spreading and migration, gas plasma exposure affected cell surface adhesion receptors (e.g., integrinα5β1, syndecan 4), structural proteins (e.g., vinculin, talin, actin), and transcription of genes associated with differentiation markers of fibroblasts-to-myofibroblasts and epithelial-to-mesenchymal transition, cellular protrusions, fibronectin fibrillogenesis, matrix metabolism, and matrix metalloproteinase activity. Finally, we documented that gas plasma exposure increased tissue oxygenation and skin perfusion during ROS-driven wound healing. Altogether, these results provide critical insights into the molecular machinery of gas plasma-assisted wound healing mechanisms.
Collapse
Affiliation(s)
- Anke Schmidt
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany.
| | - Grit Liebelt
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Felix Nießner
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Thomas von Woedtke
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany; Department of Hygiene and Environmental Medicine, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475, Greifswald, Germany
| | - Sander Bekeschus
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany.
| |
Collapse
|
26
|
Shafaie S, Andalib S, Shafaei H, Montaseri A, Tavakolizadeh M. Differential Biological Behavior of Fibroblasts and Endothelial Cells under Aloe vera Gel Culturing. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2020; 9:234-246. [PMID: 33274186 PMCID: PMC7703660 DOI: 10.22088/ijmcm.bums.9.3.234] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/06/2020] [Indexed: 01/10/2023]
Abstract
Aloe vera is used for its large variety of biological activities such as wound healing, anti-fungal, anti-inflammatory, hypoglycemic, immunomodulatory, gastroprotective, and anti-cancer. Although the beneficial effects of Aloe vera on wound healing have been proven, little is known about its effects at the cellular level. In this study, we evaluated the angiogenic and migrative effects of Aloe vera gel on fibroblasts and endothelial cells. Fibroblasts and endothelial cells were cultured in monolayer conditions with low glucose DMEM with 10% serum and 1% penicillin-streptomycin. Fresh and mature leaves of Aloe vera were used for gel preparation. Cell proliferation and morphology were studied by an inverted microscope. The migration of fibroblasts was assessed by scratch assay. MTT assay was performed for cell viability assessment, and real-time RT-PCR was used for evaluation of PECAM-1, integrin α1 and β1 transcription. After two days, the protein level of PECAM-1 was detected by flow cytometry. Our results showed that Aloe vera has a higher proliferative effect on fibroblasts in comparison with endothelial cells. Aloe vera also induced the migration of fibroblasts. The viability of both types of cells was similar to control ones. Integrin α1, β1 and PECAM-1 gene expression increased significantly (P <0.005) in Aloe vera treated fibroblasts and endothelial cells in comparison with the control groups. However, the expression of these genes was significantly higher in fibroblasts in comparison with endothelial cells. Protein levels of PECAM-1 showed no change in both cell types upon Aloe vera treatment. Aloe vera gel induced angiogenic and cell adhesion properties in fibroblasts more than endothelial cells. Further investigations are needed to show the main role of fibroblasts rather than endothelial cells in wound healing by Aloe vera administration.
Collapse
Affiliation(s)
- Saba Shafaie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Sina Andalib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hajar Shafaei
- Stem Cell Research Center, Tabriz University of Medical Sciences , Tabriz.,Department of Anatomical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Azadeh Montaseri
- Department of Anatomical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Tavakolizadeh
- Department of Pharmacognosy, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
27
|
Meizarini A, Aryati A, Rianti D, Riawan W, Puteri A. Effectivity of zinc oxide-turmeric extract dressing in stimulating the reepithelization phase of wound healing. Vet World 2020; 13:2221-2225. [PMID: 33281360 PMCID: PMC7704322 DOI: 10.14202/vetworld.2020.2221-2225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/15/2020] [Indexed: 11/16/2022] Open
Abstract
Background and Aim Reepithelialization can be described as the resurfacing of a wound with new epithelium in the process of healing, with the overlapping step from keratinocyte migration and proliferation to the tissue contraction. Zinc oxide-turmeric extract dressing has been proven to have anti-inflammatory properties, but its effectivity in the reepithelialization process is still unknown. This study aimed to determine the effect of a wound dressing consisting of zinc oxide and turmeric extract on wound reepithelialization by assessing the expression of cytokeratin 14 (CK14), epidermal growth factor receptor (EGFR), and epithelial cadherin (E-cadherin). Materials and Methods A total of 40 Wistar rats were randomized into four control and four treatment groups (n=5 per group). On day 1, a square-shaped full-thickness skin excision measuring 6×6 mm in size was created in the dorsal thoracic area of the rats, and the wounds were either dressed with a combination of zinc oxide and turmeric extract in the treatment groups or left undressed in the control groups. Then, the rats were sequentially sacrificed on days 3, 5, 7, and 14 to obtain subepithelial excision samples, which were subsequently subjected to immunohistochemistry analysis for the expression of CK14, EGFR, and E-cadherin to ascertain wound reepithelization. The data were tabulated and analyzed using a one-way analysis of variance and least significant difference test. Results The highest expression levels of CK14, EGFR, and E-cadherin were observed on days 7 and 14 in the treatment and control groups, respectively. While the expression levels of these markers on day 7 were found to be significantly higher in the treatment than the control groups, no significant difference in the expression levels on day 14 was detected between the control and treatment groups (p<0.05). Conclusion A wound dressing consisting of zinc oxide and turmeric extract can help accelerate reepithelization in the wound healing process.
Collapse
Affiliation(s)
- Asti Meizarini
- Department of Dental Material, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Aryati Aryati
- Department of Clinical Pathology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Devi Rianti
- Department of Dental Material, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Wibi Riawan
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Astari Puteri
- Department of Oral and Maxillofacial Pathology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
28
|
Bidirectional regulation of i-type lysozyme on cutaneous wound healing. Biomed Pharmacother 2020; 131:110700. [PMID: 33152906 DOI: 10.1016/j.biopha.2020.110700] [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] [Received: 07/31/2020] [Revised: 08/21/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE This study aimed to assess the effect and mechanism of i-type lysozyme on cutaneous wound healing animal model and Multiple cell models both in vivo and in vitro. METHODS Therefore, to evaluate its regenerative efficacy on wound healing process, we daily applied i-type lysozyme on murine full-thickness excisional wounds. After sacrifice on indicated days, skin tissues around surgical defects were harvested and assessed for re-epithelialization, granulation tissue formation, neovascularization and remodeling. To elucidate the underlying mechanisms, i-type lysozyme was analyzed for its tissue regenerative potency on the proliferation, invasion, migration and tube formation against keratinocytes, fibroblasts and endothelial cells. Antioxidant and antimicrobial experiments were also conducted to elucidate protective ability of i-type lysozyme to wound bed. RESULTS It displayed excellent bi-directional regulation in wound repair, with significant acceleration of epidermal and dermal regeneration as well as the efficient attenuation of excessive collagen deposition and fibrosis in the surgical lesion. I-type lysozyme treatment augmented the proliferation and migration of HaCaT, NIH 3T3 and HUVECs, enhanced the invasion of HaCaT and HUVECs as well as accelerated tube formation of HUVECs. Additionally, it significantly recovered the proliferation of H2O2-damaged cells, whereas represented no microbicidal effect under effective concentration of wound healing. CONCLUSION Our findings demonstrate the bi-directional regulation of i-type lysozyme in wound healing process through promoting tissue regeneration while hampering scar formation, implying that it is a promising therapeutic agent for wound repair.
Collapse
|
29
|
Dabelsteen S, Pallesen EMH, Marinova IN, Nielsen MI, Adamopoulou M, Rømer TB, Levann A, Andersen MM, Ye Z, Thein D, Bennett EP, Büll C, Moons SJ, Boltje T, Clausen H, Vakhrushev SY, Bagdonaite I, Wandall HH. Essential Functions of Glycans in Human Epithelia Dissected by a CRISPR-Cas9-Engineered Human Organotypic Skin Model. Dev Cell 2020; 54:669-684.e7. [PMID: 32710848 PMCID: PMC7497784 DOI: 10.1016/j.devcel.2020.06.039] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 04/07/2020] [Accepted: 06/29/2020] [Indexed: 12/26/2022]
Abstract
The glycome undergoes characteristic changes during histogenesis and organogenesis, but our understanding of the importance of select glycan structures for tissue formation and homeostasis is incomplete. Here, we present a human organotypic platform that allows genetic dissection of cellular glycosylation capacities and systematic interrogation of the roles of distinct glycan types in tissue formation. We used CRISPR-Cas9 gene targeting to generate a library of 3D organotypic skin tissues that selectively differ in their capacity to produce glycan structures on the main types of N- and O-linked glycoproteins and glycolipids. This tissue library revealed distinct changes in skin formation associated with a loss of features for all tested glycoconjugates. The organotypic skin model provides phenotypic cues for the distinct functions of glycoconjugates and serves as a unique resource for further genetic dissection and identification of the specific structural features involved. The strategy is also applicable to other organotypic tissue models.
Collapse
Affiliation(s)
- Sally Dabelsteen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark; Department of Oral Pathology, School of Dentistry, University of Copenhagen, Denmark
| | - Emil M H Pallesen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Irina N Marinova
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Mathias I Nielsen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Maria Adamopoulou
- Department of Oral Pathology, School of Dentistry, University of Copenhagen, Denmark
| | - Troels B Rømer
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Asha Levann
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel M Andersen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Zilu Ye
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - David Thein
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Eric P Bennett
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Christian Büll
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Sam J Moons
- Institute for Molecules and Materials, Nijmegen 6525 AJ, the Netherlands
| | - Thomas Boltje
- Institute for Molecules and Materials, Nijmegen 6525 AJ, the Netherlands
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Sergey Y Vakhrushev
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ieva Bagdonaite
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Hans H Wandall
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
30
|
Kleiser S, Nyström A. Interplay between Cell-Surface Receptors and Extracellular Matrix in Skin. Biomolecules 2020; 10:E1170. [PMID: 32796709 PMCID: PMC7465455 DOI: 10.3390/biom10081170] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/01/2020] [Accepted: 08/05/2020] [Indexed: 12/12/2022] Open
Abstract
Skin consists of the epidermis and dermis, which are connected by a specialized basement membrane-the epidermal basement membrane. Both the epidermal basement membrane and the underlying interstitial extracellular matrix (ECM) created by dermal fibroblasts contain distinct network-forming macromolecules. These matrices play various roles in order to maintain skin homeostasis and integrity. Within this complex interplay of cells and matrices, cell surface receptors play essential roles not only for inside-out and outside-in signaling, but also for establishing mechanical and biochemical properties of skin. Already minor modulations of this multifactorial cross-talk can lead to severe and systemic diseases. In this review, major epidermal and dermal cell surface receptors will be addressed with respect to their interactions with matrix components as well as their roles in fibrotic, inflammatory or tumorigenic skin diseases.
Collapse
Affiliation(s)
- Svenja Kleiser
- Department of Dermatology, Faculty of Medicine and Medical Center, University of Freiburg, Hauptstraße 7, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany
| | - Alexander Nyström
- Department of Dermatology, Faculty of Medicine and Medical Center, University of Freiburg, Hauptstraße 7, 79104 Freiburg, Germany
| |
Collapse
|
31
|
Takematsu E, Spencer A, Auster J, Chen PC, Graham A, Martin P, Baker AB. Genome wide analysis of gene expression changes in skin from patients with type 2 diabetes. PLoS One 2020; 15:e0225267. [PMID: 32084158 PMCID: PMC7034863 DOI: 10.1371/journal.pone.0225267] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/31/2019] [Indexed: 12/15/2022] Open
Abstract
Non-healing chronic ulcers are a serious complication of diabetes and are a major healthcare problem. While a host of treatments have been explored to heal or prevent these ulcers from forming, these treatments have not been found to be consistently effective in clinical trials. An understanding of the changes in gene expression in the skin of diabetic patients may provide insight into the processes and mechanisms that precede the formation of non-healing ulcers. In this study, we investigated genome wide changes in gene expression in skin between patients with type 2 diabetes and non-diabetic patients using next generation sequencing. We compared the gene expression in skin samples taken from 27 patients (13 with type 2 diabetes and 14 non-diabetic). This information may be useful in identifying the causal factors and potential therapeutic targets for the prevention and treatment of diabetic related diseases.
Collapse
Affiliation(s)
- Eri Takematsu
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - Adrianne Spencer
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - Jeff Auster
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - Po-Chih Chen
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - Annette Graham
- Department of Biological and Biomedical Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Scotland, United Kingdom
| | - Patricia Martin
- Department of Biological and Biomedical Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Scotland, United Kingdom
| | - Aaron B. Baker
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX
- The Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX
- Institute for Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, TX
- * E-mail:
| |
Collapse
|
32
|
Interplay between integrins and PI4P5K Sktl is crucial for cell polarization and reepithelialisation during Drosophila wound healing. Sci Rep 2019; 9:16331. [PMID: 31704968 PMCID: PMC6842001 DOI: 10.1038/s41598-019-52743-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 10/22/2019] [Indexed: 11/08/2022] Open
Abstract
Phosphatidylinositol(4,5)-bisphosphate [PI(4,5)P2] regulates cell adhesion and actin dynamics during cell migration. PI(4,5)P2 binds various components of the cell adhesion machinery, but how these processes affect migration of the epithelial cell sheet is not well understood. Here, we report that PI(4,5)P2 and Sktl, the kinase that converts PI4P to PI(4,5)P2, are both localized to the rear side of cells during wound healing of the Drosophila larval epidermis. The Sktl localization requires JNK pathway activation and integrins, but not PVR. The sktl knockdown epidermis displays strong defects in would closure, reminiscent of the JNK-depleted epidermis, and shows severe disruption of cell polarity, as determined by myosin II localization. Sktl and βPS integrin colocalize at the rear side of cells forming the trailing edge during wound healing and the two are inter-dependent in that the absence of one severely disrupts the rear localization of the other. These results strongly suggest that the JNK pathway regulates the rear localization of Sktl and integrins and the interplay between Sktl and integrins sets up cell polarity, which is crucial for reepithelialisation during wound healing.
Collapse
|
33
|
Magne B, Dedier M, Nivet M, Coulomb B, Banzet S, Lataillade JJ, Trouillas M. IL-1β-Primed Mesenchymal Stromal Cells Improve Epidermal Substitute Engraftment and Wound Healing via Matrix Metalloproteinases and Transforming Growth Factor-β1. J Invest Dermatol 2019; 140:688-698.e21. [PMID: 31513805 DOI: 10.1016/j.jid.2019.07.721] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/18/2019] [Accepted: 07/31/2019] [Indexed: 12/16/2022]
Abstract
Since the 1980s, deep and extensive skin wounds and burns are treated with autologous split-thickness skin grafts, or cultured epidermal autografts, when donor sites are limited. However, the clinical use of cultured epidermal autografts often remains unsatisfactory because of poor engraftment rates, altered wound healing, and reduced skin functionality. In the past few decades, mesenchymal stromal cells (MSCs) have raised much attention because of their anti-inflammatory, protrophic, and pro-remodeling capacities. More specifically, gingival MSCs have been shown to possess enhanced wound healing properties compared with other tissue sources. Growing evidence also indicates that MSC priming could potentiate therapeutic effects in diverse in vitro and in vivo models of skin trauma. In this study, we found that IL-1β-primed gingival MSCs promoted cell migration, dermal-epidermal junction formation, and inflammation reduction in vitro, as well as improved epidermal substitute engraftment in vivo. IL-1β-primed gingival MSCs had different secretory profiles from naive gingival MSCs, characterized by an overexpression of transforming growth factor-β and matrix metalloproteinase (MMP) pathway agonists. Eventually, MMP-1, MMP-9, and transforming growth factor-β1 appeared to be critically involved in IL-1β-primed gingival MSC mechanisms of action.
Collapse
Affiliation(s)
- Brice Magne
- IRBA (French Armed-Forces Biomedical Research Institute), Clamart, France; INSERM UMR-1197, Villejuif, France; Scarcell Therapeutics, Paris, France
| | - Marianne Dedier
- IRBA (French Armed-Forces Biomedical Research Institute), Clamart, France
| | - Muriel Nivet
- IRBA (French Armed-Forces Biomedical Research Institute), Clamart, France; INSERM UMR-1197, Villejuif, France
| | - Bernard Coulomb
- INSERM UMR-1197, Villejuif, France; Scarcell Therapeutics, Paris, France
| | - Sébastien Banzet
- IRBA (French Armed-Forces Biomedical Research Institute), Clamart, France; INSERM UMR-1197, Villejuif, France
| | - Jean-Jacques Lataillade
- IRBA (French Armed-Forces Biomedical Research Institute), Clamart, France; INSERM UMR-1197, Villejuif, France
| | - Marina Trouillas
- IRBA (French Armed-Forces Biomedical Research Institute), Clamart, France; INSERM UMR-1197, Villejuif, France.
| |
Collapse
|
34
|
Yang R, Liu F, Wang J, Chen X, Xie J, Xiong K. Epidermal stem cells in wound healing and their clinical applications. Stem Cell Res Ther 2019; 10:229. [PMID: 31358069 PMCID: PMC6664527 DOI: 10.1186/s13287-019-1312-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The skin has important barrier, sensory, and immune functions, contributing to the health and integrity of the organism. Extensive skin injuries that threaten the entire organism require immediate and effective treatment. Wound healing is a natural response, but in severe conditions, such as burns and diabetes, this process is insufficient to achieve effective treatment. Epidermal stem cells (EPSCs) are a multipotent cell type and are committed to the formation and differentiation of the functional epidermis. As the contributions of EPSCs in wound healing and tissue regeneration have been increasingly attracting the attention of researchers, a rising number of therapies based on EPSCs are currently under development. In this paper, we review the characteristics of EPSCs and the mechanisms underlying their functions during wound healing. Applications of EPSCs are also discussed to determine the potential and feasibility of using EPSCs clinically in wound healing.
Collapse
Affiliation(s)
- Ronghua Yang
- Department of Burn Surgery, The First People’s Hospital of Foshan, Foshan, 528000 China
| | - Fengxia Liu
- Department of Human Anatomy, School of Basic Medical Science, Xinjiang Medical University, Urumqi, 830001 China
| | - Jingru Wang
- Department of Burn Surgery, The First People’s Hospital of Foshan, Foshan, 528000 China
| | - Xiaodong Chen
- Department of Burn Surgery, The First People’s Hospital of Foshan, Foshan, 528000 China
| | - Julin Xie
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 512100 China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Morphological Sciences Building, Central South University, 172 Tongzi Po Road, Changsha, 410013 Hunan China
| |
Collapse
|
35
|
Perez‐Aso M, Roca A, Bosch J, Martínez‐Teipel B. Striae reconstructed, a full thickness skin model that recapitulates the pathology behind stretch marks. Int J Cosmet Sci 2019; 41:311-319. [DOI: 10.1111/ics.12538] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/03/2019] [Indexed: 01/08/2023]
Affiliation(s)
- M. Perez‐Aso
- Provital, S.A. Gorgs Llado 200, 08210 Barbera del Valles Barcelona Spain
| | - A. Roca
- Provital, S.A. Gorgs Llado 200, 08210 Barbera del Valles Barcelona Spain
| | - J. Bosch
- Provital, S.A. Gorgs Llado 200, 08210 Barbera del Valles Barcelona Spain
| | - B. Martínez‐Teipel
- Provital, S.A. Gorgs Llado 200, 08210 Barbera del Valles Barcelona Spain
| |
Collapse
|
36
|
DiPersio CM, Van De Water L. Integrin Regulation of CAF Differentiation and Function. Cancers (Basel) 2019; 11:cancers11050715. [PMID: 31137641 PMCID: PMC6563118 DOI: 10.3390/cancers11050715] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/14/2019] [Accepted: 05/21/2019] [Indexed: 02/07/2023] Open
Abstract
Extensive remodeling of the extracellular matrix, together with paracrine communication between tumor cells and stromal cells, contribute to an “activated” tumor microenvironment that supports malignant growth and progression. These stromal cells include inflammatory cells, endothelial cells, and cancer-associated fibroblasts (CAFs). Integrins are expressed on all tumor and stromal cell types where they regulate both cell adhesion and bidirectional signal transduction across the cell membrane. In this capacity, integrins control pro-tumorigenic cell autonomous functions such as growth and survival, as well as paracrine crosstalk between tumor cells and stromal cells. The myofibroblast-like properties of cancer-associated fibroblasts (CAFs), such as robust contractility and extracellular matrix (ECM) deposition, allow them to generate both chemical and mechanical signals that support invasive tumor growth. In this review, we discuss the roles of integrins in regulating the ability of CAFs to generate and respond to extracellular cues in the tumor microenvironment. Since functions of specific integrins in CAFs are only beginning to emerge, we take advantage of a more extensive literature on how integrins regulate wound myofibroblast differentiation and function, as some of these integrin functions are likely to extrapolate to CAFs within the tumor microenvironment. In addition, we discuss the roles that integrins play in controlling paracrine signals that emanate from epithelial/tumor cells to stimulate fibroblasts/CAFs.
Collapse
|
37
|
Zheng R, Longmate WM, DeFreest L, Varney S, Wu L, DiPersio CM, Van De Water L. Keratinocyte Integrin α3β1 Promotes Secretion of IL-1α to Effect Paracrine Regulation of Fibroblast Gene Expression and Differentiation. J Invest Dermatol 2019; 139:2029-2038.e3. [PMID: 30878678 DOI: 10.1016/j.jid.2019.02.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/05/2019] [Accepted: 02/06/2019] [Indexed: 12/31/2022]
Abstract
After cutaneous injury, keratinocytes secrete paracrine factors that regulate wound cell functions; dysregulation of this signaling can lead to wound pathologies. Previously, we established that keratinocyte integrin α3β1 promotes wound angiogenesis through paracrine stimulation of endothelial cells. We hypothesize here that α3β1-dependent paracrine signaling from keratinocytes regulates the differentiation state of myofibroblasts. We report that epidermal α3-knockout mice exhibit more wound myofibroblasts and fewer cyclooxygenase 2 (Cox-2)-positive dermal cells than controls. We also found that conditioned medium from α3-expressing mouse keratinocytes (MKα3+), but not from α3-null MK cells (MKα3-), induces expression of Cox-2 in fibroblasts in a time- and dose-dependent manner and that this induction is mediated by IL-1α. Compared with MKα3- cells, MKα3+ cells secrete more IL-1α and less IL-1RA, a natural IL-1 receptor antagonist. Treatment with an IL-1α neutralizing antibody, recombinant IL-1RA, or IL-1 receptor-targeting small interfering RNA suppresses MKα3+ conditioned medium-dependent induction of Cox-2 expression in fibroblasts. Finally, active recombinant IL-1α is sufficient to induce Cox-2 in fibroblasts and to inhibit transforming growth factor-β-induced α-SMA expression. Our findings support a role for keratinocyte integrin α3β1 in controlling the secretion of IL-1α, a paracrine factor that regulates the wound myofibroblast phenotype.
Collapse
Affiliation(s)
- Rui Zheng
- Department of Surgery, Albany Medical College, Albany, New York, USA; Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York
| | | | - Lori DeFreest
- Department of Surgery, Albany Medical College, Albany, New York, USA
| | - Scott Varney
- Department of Surgery, Albany Medical College, Albany, New York, USA
| | - Lei Wu
- Department of Surgery, Albany Medical College, Albany, New York, USA
| | - C Michael DiPersio
- Department of Surgery, Albany Medical College, Albany, New York, USA; Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York
| | - Livingston Van De Water
- Department of Surgery, Albany Medical College, Albany, New York, USA; Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York.
| |
Collapse
|
38
|
Andasari V, Lü D, Swat M, Feng S, Spill F, Chen L, Luo X, Zaman M, Long M. Computational model of wound healing: EGF secreted by fibroblasts promotes delayed re-epithelialization of epithelial keratinocytes. Integr Biol (Camb) 2018; 10:605-634. [PMID: 30206629 PMCID: PMC6571173 DOI: 10.1039/c8ib00048d] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
It is widely agreed that keratinocyte migration plays a crucial role in wound re-epithelialization. Defects in this function contribute to wound reoccurrence causing significant clinical problems. Several in vitro studies have shown that the speed of migrating keratinocytes can be regulated by epidermal growth factor (EGF) which affects keratinocyte's integrin expression. The relationship between integrin expression (through cell-matrix adhesion) stimulated by EGF and keratinocyte migration speed is not linear since increased adhesion, due to increased integrin expression, has been experimentally shown to slow down cell migration due to the biphasic dependence of cell speed on adhesion. In our previous work we showed that keratinocytes that were co-cultured with EGF-enhanced fibroblasts formed an asymmetric migration pattern, where, the cumulative distances of keratinocytes migrating toward fibroblasts were smaller than those migrating away from fibroblasts. This asymmetric pattern is thought to be provoked by high EGF concentration secreted by fibroblasts. The EGF stimulates the expression of integrin receptors on the surface of keratinocytes migrating toward fibroblasts via paracrine signaling. In this paper, we present a computational model of keratinocyte migration that is controlled by EGF secreted by fibroblasts using the Cellular Potts Model (CPM). Our computational simulation results confirm the asymmetric pattern observed in experiments. These results provide a deeper insight into our understanding of the complexity of keratinocyte migration in the presence of growth factor gradients and may explain re-epithelialization failure in impaired wound healing.
Collapse
Affiliation(s)
- Vivi Andasari
- Boston University, Department of Biomedical Engineering, 44 Cummington Mall, Boston, MA 02215, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Constitutional absence of epithelial integrin α3 impacts the composition of the cellular microenvironment of ILNEB keratinocytes. Matrix Biol 2018; 74:62-76. [PMID: 30466509 DOI: 10.1016/j.matbio.2018.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 07/01/2018] [Indexed: 11/19/2022]
Abstract
Integrin α3β1, a major epidermal adhesion receptor is critical for organization of the basement membrane during development and wound healing. Integrin α3 deficiency leads to interstitial lung disease, nephrotic syndrome and epidermolysis bullosa (ILNEB), an autosomal recessive multiorgan disease characterized by basement membrane abnormalities in skin, lung and kidney. The pathogenetic chains from ITGA3 mutation to tissue abnormalities are still unclear. Although integrin α3 was reported to regulate multiple extracellular proteins, the composition of the extracellular compartment of integrin α3-negative keratinocytes has not been resolved so far. In a comprehensive approach, quantitative proteomics of deposited extracellular matrix, conditioned cultured media as well as of the intracellular compartment of keratinocytes isolated from an ILNEB patient and from normal skin were performed. By mass spectrometry-based proteomics, 167 proteins corresponding to the GO terms "extracellular" and "cell adhesion", or included in the "human matrisome" were identified in the deposited extracellular matrix, and 217 in the conditioned media of normal human keratinocytes. In the absence of integrin α3, 33% and 26% respectively were dysregulated. Dysregulated proteins were functionally related to integrin α3 or were known interaction partners. The results show that in the absence of integrin α3 ILNEB keratinocytes produce a fibronectin-rich microenvironment and make use of fibronectin-binding integrin subunits αv and α5. The most important results were validated in monolayer and organotypic coculture models. Finally, the in vivo relevance of the most dysregulated components was demonstrated by immunostainings of skin, kidney and lung samples of three ILNEB patients.
Collapse
|
40
|
Das L, Gard JMC, Prekeris R, Nagle RB, Morrissey C, Knudsen BS, Miranti CK, Cress AE. Novel Regulation of Integrin Trafficking by Rab11-FIP5 in Aggressive Prostate Cancer. Mol Cancer Res 2018; 16:1319-1331. [PMID: 29759989 DOI: 10.1158/1541-7786.mcr-17-0589] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 02/07/2018] [Accepted: 04/23/2018] [Indexed: 11/16/2022]
Abstract
The laminin-binding integrins, α3β1 and α6β1, are needed for tumor metastasis and their surface expression is regulated by endocytic recycling. β1 integrins share the Rab11 recycling machinery, but the trafficking of α3β1 and α6β1 are distinct by an unknown mechanism. Using a mouse PDX tumor model containing human metastatic prostate cancer, Rab11 family interacting protein 5 (Rab11-FIP5) was identified as a lead candidate for α6β1 trafficking. Rab11-FIP5 and its membrane-binding domain were required for α6β1 recycling, without affecting the other laminin-binding integrin (i.e., α3β1) or unrelated membrane receptors like CD44, transferrin receptor, or E-cadherin. Depletion of Rab11-FIP5 resulted in the intracellular accumulation of α6β1 in the Rab11 recycling compartment, loss of cell migration on laminin, and an unexpected loss of α6β1 recycling in cell-cell locations. Taken together, these data demonstrate that α6β1 is distinct from α3β1 via Rab11-FIP5 recycling and recycles in an unexpected cell-cell location.Implications: Rab11-FIP5-dependent α6β1 integrin recycling may be selectively targeted to limit migration of prostate cancer cells into laminin-rich tissues. Mol Cancer Res; 16(8); 1319-31. ©2018 AACR.
Collapse
Affiliation(s)
- Lipsa Das
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona
| | - Jaime M C Gard
- The University of Arizona Cancer Center, University of Arizona, Tucson, Arizona
| | - Rytis Prekeris
- University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Raymond B Nagle
- The University of Arizona Cancer Center, University of Arizona, Tucson, Arizona.,Pathology, University of Washington, Seattle, Washington
| | | | | | - Cindy K Miranti
- Cellular and Molecular Medicine, The University of Arizona Cancer Center, University of Arizona, Tucson, Arizona
| | - Anne E Cress
- The University of Arizona Cancer Center, University of Arizona, Tucson, Arizona. .,Cellular and Molecular Medicine, The University of Arizona Cancer Center, University of Arizona, Tucson, Arizona.,Molecular and Cellular Biology, The University of Arizona Cancer Center, University of Arizona, Tucson, Arizona
| |
Collapse
|
41
|
Longmate WM, Lyons SP, DeFreest L, Van De Water L, DiPersio CM. Opposing Roles of Epidermal Integrins α3β1 and α9β1 in Regulation of mTLD/BMP-1-Mediated Laminin-γ2 Processing during Wound Healing. J Invest Dermatol 2018; 138:444-451. [PMID: 28923241 PMCID: PMC5794664 DOI: 10.1016/j.jid.2017.09.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 08/25/2017] [Accepted: 09/01/2017] [Indexed: 11/27/2022]
Abstract
Proteolytic processing of the laminin-γ2 chain is a hallmark of basement membrane maturation in the skin. Integrin α3β1, a major receptor for epidermal adhesion to laminin-332, is critical for proper basement membrane organization during skin development and wound healing. Previously, we identified a role for α3β1 in promoting the processing of laminin-γ2 in cultured keratinocytes in vitro and in wound epidermis in vivo. In this study we identify the Bmp1 gene, which encodes variants of the mTLD/BMP-1 metalloproteases, as a critical regulator of α3β1-dependent laminin-γ2 processing, thereby expanding the role of this integrin in controlling the secretion by the epidermis of factors that modulate the tissue microenvironment. Because our previous studies identified another epidermal integrin, α9β1, as a suppressive regulator of α3β1-dependent wound angiogenesis, we investigated whether α9β1 has a similar cross-suppressive effect on the ability of α3β1 to promote basement membrane organization. Here, we show that, rather than a cross-suppressive role, α9β1 has an opposing role in basement membrane assembly/maturation through reduced laminin-γ2 processing via mTLD/BMP-1. Although α3β1 promotes this process during wound healing, α9β1 has an inhibitory role, suggesting that regulation of basement membrane assembly requires a complex interplay between these distinct epidermal integrins.
Collapse
Affiliation(s)
- Whitney M Longmate
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
| | - Scott P Lyons
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
| | - Lori DeFreest
- Department of Surgery, Albany Medical College, Albany, New York, USA
| | - Livingston Van De Water
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA; Department of Surgery, Albany Medical College, Albany, New York, USA
| | - C Michael DiPersio
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA; Department of Surgery, Albany Medical College, Albany, New York, USA.
| |
Collapse
|
42
|
Reimer A, He Y, Has C. Update on Genetic Conditions Affecting the Skin and the Kidneys. Front Pediatr 2018; 6:43. [PMID: 29552546 PMCID: PMC5840143 DOI: 10.3389/fped.2018.00043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/14/2018] [Indexed: 01/01/2023] Open
Abstract
Genetic conditions affecting the skin and kidney are clinically and genetically heterogeneous, and target molecular components present in both organs. The molecular pathology involves defects of cell-matrix adhesion, metabolic or signaling pathways, as well as tumor suppressor genes. This article gives a clinically oriented overview of this group of disorders, highlighting entities which have been recently described, as well as the progress made in understanding well-known entities. The genetic bases as well as molecular cell biological mechanisms are described, with therapeutic applications.
Collapse
Affiliation(s)
- Antonia Reimer
- Department of Dermatology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany.,Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Yinghong He
- Department of Dermatology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Cristina Has
- Department of Dermatology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| |
Collapse
|
43
|
Ferroni L, Gardin C, De Pieri A, Sambataro M, Seganfreddo E, Goretti C, Iacopi E, Zavan B, Piaggesi A. Treatment of diabetic foot ulcers with Therapeutic Magnetic Resonance (TMR®) improves the quality of granulation tissue. Eur J Histochem 2017; 61:2800. [PMID: 29046049 PMCID: PMC5572111 DOI: 10.4081/ejh.2017.2800] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/25/2017] [Accepted: 06/25/2017] [Indexed: 11/22/2022] Open
Abstract
Diabetic foot ulcers (DFUs) often result in severely adverse outcomes, such as serious infections, hospitalization, and lower extremity amputations. In last few years, to improve the outcome of DFUs, clinicians and researchers put their attention on the application of low intensity pulsating electro-magnetic fields through Therapeutic Magnetic Resonance (TMR®). In our study, patients with DFUs have been divided into two groups: The Sham Group treated with non-functioning TMR® device, and the Active Group treated with a functioning device. Biopsies were recovered from ulcers before and after a 15-day treatment with both kind of TMR® device. To recognize signs of inflammation or healing process, the harvested biopsies were subjected to histological and molecular analyses. The histological analysis showed a change in cell population after treatment with TMR®: an increase of fibroblasts and endothelial cells with a reduction of inflammatory cells. After TMR® application, the gene expression profile analysis revealed an improvement in extracellular matrix components such as matrix metalloproteinases, collagens and integrins, a reduction in pro-inflammatory interleukins, and an increase in growth factors expression. In conclusion, our research has identified histological and molecular features of reduced inflammation and increased cell proliferation during the wound healing process in response to TMR® application.
Collapse
|
44
|
Das L, Anderson TA, Gard JM, Sroka IC, Strautman SR, Nagle RB, Morrissey C, Knudsen BS, Cress AE. Characterization of Laminin Binding Integrin Internalization in Prostate Cancer Cells. J Cell Biochem 2017; 118:1038-1049. [PMID: 27509031 PMCID: PMC5553695 DOI: 10.1002/jcb.25673] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 08/09/2016] [Indexed: 12/27/2022]
Abstract
Laminin binding integrins α6 (CD49f) and α3 (CD49c) are persistently but differentially expressed in prostate cancer (PCa). Integrin internalization is an important determinant of their cell surface expression and function. Using flow cytometry, and first order kinetic modeling, we quantitated the intrinsic internalization rates of integrin subunits in a single cycle of internalization. In PCa cell line DU145, α6 integrin internalized with a rate constant (kactual ) of 3.25 min-1 , threefold faster than α3 integrin (1.0 min-1 ), 1.5-fold faster than the vitronectin binding αv integrin (CD51) (2.2 min-1 ), and significantly slower than the unrelated transferrin receptor (CD71) (15 min-1 ). Silencing of α3 integrin protein expression in DU145, PC3, and PC3B1 cells resulted in up to a 1.71-fold increase in kactual for α6 integrin. The internalized α6 integrin was targeted to early endosomes but not to lamp1 vesicles. Depletion of α3 integrin expression resulted in redistribution of α6β4 integrin to an observed cell-cell staining pattern that is consistent with a suprabasal distribution observed in epidermis and early PIN lesions in PCa. Depletion of α3 integrin increased cell migration by 1.8-fold, which was dependent on α6β1 integrin. Silencing of α6 integrin expression however, had no significant effect on the kactual of α3 integrin or its distribution in early endosomes. These results indicate that α3 and α6 integrins have significantly different internalization kinetics and that coordination exists between them for internalization. J. Cell. Biochem. 118: 1038-1049, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lipsa Das
- Department of Cancer Biology, University of Arizona, Tucson, AZ 85724
| | - Todd A. Anderson
- The University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724
| | - Jaime M.C. Gard
- The University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724
| | - Isis C. Sroka
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724
| | | | - Raymond B. Nagle
- Department of Pathology, University of Arizona, Tucson, AZ 85724
- The University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724
| | | | | | - Anne E. Cress
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85724
- The University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724
| |
Collapse
|
45
|
Has C, He Y. Renal-skin syndromes. Cell Tissue Res 2017; 369:63-73. [DOI: 10.1007/s00441-017-2623-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 04/03/2017] [Indexed: 12/16/2022]
|
46
|
Longmate WM, Lyons SP, Chittur SV, Pumiglia KM, Van De Water L, DiPersio CM. Suppression of integrin α3β1 by α9β1 in the epidermis controls the paracrine resolution of wound angiogenesis. J Cell Biol 2017; 216:1473-1488. [PMID: 28416479 PMCID: PMC5412555 DOI: 10.1083/jcb.201510042] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 02/10/2017] [Accepted: 03/14/2017] [Indexed: 12/02/2022] Open
Abstract
The development of novel therapies to promote wound healing is hindered by our poor understanding of how different integrins function together in the epidermis. Longmate et al. show that cross-suppression by integrins within the epidermis controls paracrine signals that regulate wound angiogenesis. Integrin α9β1 suppresses the proangiogenic functions of α3β1 during late-stage wound healing, leading to the normalization of blood vessel density in the wound bed. Development of wound therapies is hindered by poor understanding of combinatorial integrin function in the epidermis. In this study, we generated mice with epidermis-specific deletion of α3β1, α9β1, or both integrins as well as keratinocyte lines expressing these integrin combinations. Consistent with proangiogenic roles for α3β1, α3-null keratinocytes showed reduced paracrine stimulation of endothelial cell migration and survival, and wounds of epidermis-specific α3 knockout mice displayed impaired angiogenesis. Interestingly, α9β1 in keratinocytes suppressed α3β1-mediated stimulation of endothelial cells, and wounds of epidermis-specific α9 knockout mice displayed delayed vascular normalization and reduced endothelial apoptosis, indicating that α9β1 cross-suppresses α3β1 proangiogenic functions. Moreover, α9β1 inhibited α3β1 signaling downstream of focal adhesion kinase (FAK) autoactivation at the point of Src-mediated phosphorylation of FAK Y861/Y925. Finally, α9β1 cross-suppressed many α3β1-dependent genes, including the gene that encodes MMP-9, which we implicated as a regulator of integrin-dependent cross talk to endothelial cells. Our findings identify a novel physiological context for combinatorial integrin signaling, laying the foundation for therapeutic strategies that manipulate α9β1 and/or α3β1 during wound healing.
Collapse
Affiliation(s)
- Whitney M Longmate
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208
| | - Scott P Lyons
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208
| | - Sridar V Chittur
- Center for Functional Genomics, University at Albany, Rensselaer, NY 12144
| | - Kevin M Pumiglia
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208
| | - Livingston Van De Water
- Department of Surgery, Albany Medical College, Albany, NY 12208.,Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208
| | - C Michael DiPersio
- Department of Surgery, Albany Medical College, Albany, NY 12208 .,Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208
| |
Collapse
|
47
|
TMF and glycitin act synergistically on keratinocytes and fibroblasts to promote wound healing and anti-scarring activity. Exp Mol Med 2017; 49:e302. [PMID: 28303029 PMCID: PMC5382558 DOI: 10.1038/emm.2016.167] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 09/27/2016] [Accepted: 10/31/2016] [Indexed: 01/04/2023] Open
Abstract
Keratinocyte-fibroblast interactions are critical for skin repair after injury. During the proliferative phase of wound healing, proliferation, migration and differentiation of these cells are the major mechanisms leading to tissue remodeling. We have previously reported that glycitin, a major soy isoflavone, stimulates dermal fibroblast proliferation; and the phytochemical, 4′,6,7-trimethoxyisoflavone (TMF), induces migration of HaCaT keratinocyte cells. We therefore investigated whether these compounds display synergistic effects on skin cells during wound healing in vitro and in vivo. Co-treatment with TMF and glycitin synergistically promotes the proliferation and migration of both keratinocytes and dermal fibroblasts, with a 1:1 ratio of these compounds showing the greatest efficacy in our co-culture system. This keratinocyte-fibroblast interaction occurred via the secretion of TGF-β, and the induction of differentiation and proliferation was confirmed in both indirect and direct co-culture assays. In an excisional and burn wound animal model, mice treated with a 1:1 ratio of TMF and glycitin showed faster wound closure, regeneration and scar reduction than even the positive control drug. These data indicate that two isoflavones, TMF and glycitin, act synergistically to promote wound healing and anti-scarring and could potentially be developed together as a bioactive therapeutic for wound treatment.
Collapse
|
48
|
Shen C, Sun L, Zhu N, Qi F. Kindlin-1 contributes to EGF-induced re-epithelialization in skin wound healing. Int J Mol Med 2017; 39:949-959. [PMID: 28290610 PMCID: PMC5360437 DOI: 10.3892/ijmm.2017.2911] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 02/21/2017] [Indexed: 02/04/2023] Open
Abstract
The commercial use of epidermal growth factor (EGF) is extensive and has been shown to be effective for skin wound healing in clinical practice. There is evidence to indicate that the topical administration of EGF significantly accelerates re-epithelialization by promoting keratinocyte mitogenesis and migration following acute injury; however, the mechanisms involved remain to be elucidated. Thus, in this study, we focused on Kindlin-1, a four-point-one, ezrin, radixin, moesin (FERM)-domain-containing adaptor protein, and report its contribution to EGF-induced re-epithelialization in skin wound healing. In tissue samples, the expression of Kindlin-1 was induced upon EGF treatment compared to that in the natural healing group. In immortalized human keratinocytes (HaCaT cells), we further proved that Kindlin-1 was necessary for mediating EGF-induced activation signals, including integrin β1 activation, focal adhesion kinase (FAK) phosphorylation and actin re-organization, which finally led to enhanced cell proliferation and migration. These results indicate that Kindlin-1 is essential in EGF-induced re-epithelialization in skin wound healing and provide additional rationale for the clinical application of EGF in the treatment of acute wounds.
Collapse
Affiliation(s)
- Congcong Shen
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Linlin Sun
- Department of Biochemistry and Molecular Biology, Basic Medical College of Fudan University, Shanghai 200032, P.R. China
| | - Ningwen Zhu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Fazhi Qi
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
49
|
Iwanabe Y, Masaki C, Tamura A, Tsuka S, Mukaibo T, Kondo Y, Hosokawa R. The effect of low-intensity pulsed ultrasound on wound healing using scratch assay in epithelial cells. J Prosthodont Res 2016; 60:308-314. [DOI: 10.1016/j.jpor.2016.03.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/07/2016] [Accepted: 03/15/2016] [Indexed: 10/22/2022]
|
50
|
Integrin-mediated regulation of epidermal wound functions. Cell Tissue Res 2016; 365:467-82. [PMID: 27351421 DOI: 10.1007/s00441-016-2446-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 06/02/2016] [Indexed: 01/14/2023]
Abstract
During cutaneous wound healing, keratinocyte proliferation and migration are critical for re-epithelialization. In addition the epidermis secretes growth factors, cytokines, proteases, and matricellular proteins into the wound microenvironment that modify the extracellular matrix and stimulate other wound cells that control the inflammatory response, promote angiogenesis and facilitate tissue contraction and remodeling. Wound keratinocytes express at least seven different integrins-the major cell adhesion receptors for the extracellular matrix-that collectively control essential cell-autonomous functions to ensure proper re-epithelialization, including migration, proliferation, survival and basement membrane assembly. Moreover, it has become evident in recent years that some integrins can regulate paracrine signals from wound epidermis that stimulate other wound cells involved in angiogenesis, contraction and inflammation. Importantly, it is likely that abnormal integrin expression or function in the epidermis contributes to wound pathologies such as over-exuberant healing (e.g., hypertrophic scar formation) or diminished healing (e.g., chronic wounds). In this review, we discuss current knowledge of integrin function in the epidermis, which implicates them as attractive therapeutic targets to promote wound healing or treat wound pathologies. We also discuss challenges that arise from the complex roles that multiple integrins play in wound epidermis, which may be regulated through extracellular matrix remodeling that determines ligand availability. Indeed, understanding how different integrin functions are temporally coordinated in wound epidermis and which integrin functions go awry in pathological wounds, will be important to determine how best to target them clinically to achieve maximum therapeutic benefit. Graphical abstract In addition to their well-characterized roles in keratinocyte adhesion, migration and wound re-epithelialization, epidermal integrins play important roles in modifying the wound microenvironment by regulating the expression and secretion of growth factors, extracellular proteases, and matricellular proteins that stimulate other wound cells, including vascular endothelial cells and fibroblasts/myofibroblasts.
Collapse
|