1
|
Chen SD, Chu CY, Wang CB, Yang Y, Xu ZY, Qu YL, Man Y. Integrated-omics profiling unveils the disparities of host defense to ECM scaffolds during wound healing in aged individuals. Biomaterials 2024; 311:122685. [PMID: 38944969 DOI: 10.1016/j.biomaterials.2024.122685] [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: 02/05/2024] [Revised: 06/11/2024] [Accepted: 06/23/2024] [Indexed: 07/02/2024]
Abstract
Extracellular matrix (ECM) scaffold membranes have exhibited promising potential to better the outcomes of wound healing by creating a regenerative microenvironment around. However, when compared to the application in younger individuals, the performance of the same scaffold membrane in promoting re-epithelialization and collagen deposition was observed dissatisfying in aged mice. To comprehensively explore the mechanisms underlying this age-related disparity, we conducted the integrated analysis, combing single-cell RNA sequencing (scRNA-Seq) with spatial transcriptomics, and elucidated six functionally and spatially distinctive macrophage groups and lymphocytes surrounding the ECM scaffolds. Through intergroup comparative analysis and cell-cell communication, we characterized the dysfunction of Spp1+ macrophages in aged mice impeded the activation of the type Ⅱ immune response, thus inhibiting the repair ability of epidermal cells and fibroblasts around the ECM scaffolds. These findings contribute to a deeper understanding of biomaterial applications in varied physiological contexts, thereby paving the way for the development of precision-based biomaterials tailored specifically for aged individuals in future therapeutic strategies.
Collapse
Affiliation(s)
- Shuai-Dong Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chen-Yu Chu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chen-Bing Wang
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Yang Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhao-Yu Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yi-Li Qu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yi Man
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
2
|
Endothelial Heterogeneity in Development and Wound Healing. Cells 2021; 10:cells10092338. [PMID: 34571987 PMCID: PMC8469713 DOI: 10.3390/cells10092338] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/30/2021] [Accepted: 09/06/2021] [Indexed: 12/28/2022] Open
Abstract
The vasculature is comprised of endothelial cells that are heterogeneous in nature. From tissue resident progenitors to mature differentiated endothelial cells, the diversity of these populations allows for the formation, maintenance, and regeneration of the vascular system in development and disease, particularly during situations of wound healing. Additionally, the de-differentiation and plasticity of different endothelial cells, especially their capacity to undergo endothelial to mesenchymal transition, has also garnered significant interest due to its implication in disease progression, with emphasis on scarring and fibrosis. In this review, we will pinpoint the seminal discoveries defining the phenotype and mechanisms of endothelial heterogeneity in development and disease, with a specific focus only on wound healing.
Collapse
|
3
|
Frohlich J, Vinciguerra M. Candidate rejuvenating factor GDF11 and tissue fibrosis: friend or foe? GeroScience 2020; 42:1475-1498. [PMID: 33025411 PMCID: PMC7732895 DOI: 10.1007/s11357-020-00279-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Growth differentiation factor 11 (GDF11 or bone morphogenetic protein 11, BMP11) belongs to the transforming growth factor-β superfamily and is closely related to other family member-myostatin (also known as GDF8). GDF11 was firstly identified in 2004 due to its ability to rejuvenate the function of multiple organs in old mice. However, in the past few years, the heralded rejuvenating effects of GDF11 have been seriously questioned by many studies that do not support the idea that restoring levels of GDF11 in aging improves overall organ structure and function. Moreover, with increasing controversies, several other studies described the involvement of GDF11 in fibrotic processes in various organ setups. This review paper focuses on the GDF11 and its pro- or anti-fibrotic actions in major organs and tissues, with the goal to summarize our knowledge on its emerging role in regulating the progression of fibrosis in different pathological conditions, and to guide upcoming research efforts.
Collapse
Affiliation(s)
- Jan Frohlich
- International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic
| | - Manlio Vinciguerra
- International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic.
- Institute for Liver and Digestive Health, Division of Medicine, University College London (UCL), London, UK.
| |
Collapse
|
4
|
Rousselle P, Braye F, Dayan G. Re-epithelialization of adult skin wounds: Cellular mechanisms and therapeutic strategies. Adv Drug Deliv Rev 2019; 146:344-365. [PMID: 29981800 DOI: 10.1016/j.addr.2018.06.019] [Citation(s) in RCA: 318] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/28/2018] [Accepted: 06/25/2018] [Indexed: 12/21/2022]
Abstract
Cutaneous wound healing in adult mammals is a complex multi-step process involving overlapping stages of blood clot formation, inflammation, re-epithelialization, granulation tissue formation, neovascularization, and remodelling. Re-epithelialization describes the resurfacing of a wound with new epithelium. The cellular and molecular processes involved in the initiation, maintenance, and completion of epithelialization are essential for successful wound closure. A variety of modulators are involved, including growth factors, cytokines, matrix metalloproteinases, cellular receptors, and extracellular matrix components. Here, we focus on cellular mechanisms underlying keratinocyte migration and proliferation during epidermal closure. Inability to re-epithelialize is a clear indicator of chronic non-healing wounds, which fail to proceed through the normal phases of wound healing in an orderly and timely manner. This review summarizes the current knowledge regarding the management and treatment of acute and chronic wounds, with a focus on re-epithelialization, offering some insights into novel future therapies.
Collapse
|
5
|
Ayala-Peña VB, Armiento MN, Fernández Bell Fano PM, Santillán GE, Scolaro LA. Infection of Rat Osteoblasts with Junin Virus Promotes the Expression of Bone Morphogenetic Protein 6, an Osteogenic Differentiation Inducer. Intervirology 2019; 62:1-8. [PMID: 31121597 DOI: 10.1159/000499466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 02/27/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The arenavirus Junin virus (JUNV), causative agent of the argentine hemorrhagic fever, is able to modulate several signaling pathways involved in cell survival and multiplication. OBJECTIVES We aimed to characterize the infection of rat osteoblasts (OBCs) with JUNV and its consequence on the modulation of osteogenic genes expression, thus studying the ability of this virus to induce cell differentiation. In addition, we evaluated the effect of purinergic agonists on viral replication. METHOD Quantification of infectivity by plaque forming unit (PFU) assay, synthesis of viral proteins by western blot and immunofluorescence, and expression of osteogenic differentiation markers (ODM) by quantitative real-time polymerase chain reaction were employed. RESULTS Infection of OBCs with JUNV (MOI 0.01 PFU/cell) showed a peak of infectivity, reaching 1.5 × 105 PFU/mL at the second day post-infection (p.i.). A marked restriction in multiplication was detected at day 7 p.i. that did not impair the establishment of a persistent stage of infection in OBCs. Analysis of mRNAs corresponding to ODM such as alkaline phosphatase, bone sialo-protein, and bone morphogenetic proteins (BMPs) 4 and 6 revealed that only the levels of BMP-6 were significantly higher in infected cells. Treatment with the purinergic agonists ATPγS, UTP, ADP, or UDP diminished viral titer and reduced the expression of the viral nucleoprotein. Also, treatment with 10 μM ATPγS reduced the stimulation of BMP-6 expression induced by the infection. CONCLUSIONS These data demonstrate that JUNV is capable of infecting OBCs and point out BMP-6 as a key factor during this process.
Collapse
Affiliation(s)
- Victoria B Ayala-Peña
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina.,Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina
| | - María N Armiento
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina
| | - Pablo M Fernández Bell Fano
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina
| | - Graciela E Santillán
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina.,Instituto de Ciencias Biológicas y Biomédicas del Sur, Consejo Nacional de Investigaciones Científicas y Técnicas (INBIOSUR-CONICET), Bahía Blanca, Argentina
| | - Luis A Scolaro
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina,
| |
Collapse
|
6
|
Arndt S, Karrer S, Hellerbrand C, Bosserhoff AK. Bone Morphogenetic Protein-6 Inhibits Fibrogenesis in Scleroderma Offering Treatment Options for Fibrotic Skin Disease. J Invest Dermatol 2019; 139:1914-1924.e6. [PMID: 30878675 DOI: 10.1016/j.jid.2019.02.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 02/23/2019] [Accepted: 02/25/2019] [Indexed: 12/17/2022]
Abstract
BMP6 is known to be crucial for regulating embryonic skin development. This study assessed the role of BMP6 in dermal fibrosis. We detected that BMP6 is significantly increased in skin-derived fibroblasts of patients with localized scleroderma. Moreover, it was shown that BMP6 significantly impacts proliferation, migration, cytoskeletal organization, and collagen expression, as well as activity of the major pro-fibrogenic transcription factor AP-1 in dermal fibroblasts. The importance of BMP6 in dermal fibrosis was further confirmed in an in vivo model of dermal fibrosis in which BMP6-deficient mice showed significantly enhanced fibrosis compared with wild-type mice. Conversely, application of recombinant BMP6 significantly ameliorated dermal fibrosis in this preclinical bleomycin-induced sclerosis model, and herewith provided proof of concept for the successful treatment of this fibrotic skin disease.
Collapse
Affiliation(s)
- Stephanie Arndt
- Department of Dermatology, University Medical Center Regensburg, Regensburg, Germany
| | - Sigrid Karrer
- Department of Dermatology, University Medical Center Regensburg, Regensburg, Germany
| | - Claus Hellerbrand
- Institute of Biochemistry, Emil-Fischer-Center, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Anja Katrin Bosserhoff
- Institute of Biochemistry, Emil-Fischer-Center, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany.
| |
Collapse
|
7
|
Bone morphogenetic protein 6 (BMP-6) modulates lung function, pulmonary iron levels and cigarette smoke-induced inflammation. Mucosal Immunol 2019; 12:340-351. [PMID: 30542109 DOI: 10.1038/s41385-018-0116-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 10/16/2018] [Accepted: 11/20/2018] [Indexed: 02/04/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is associated with abnormal inflammatory responses and airway wall remodeling, leading to reduced lung function. An association between the bone morphogenetic protein (BMP-6) locus and forced vital capacity has been found in a genome-wide association study. However, the role of BMP-6 in the pathogenesis of COPD remains unknown. The pulmonary expression of BMP-6 was analyzed in patients with COPD and in cigarette smoke (CS)-exposed mice. We evaluated lung function and histology in BMP-6 KO mice at baseline. We exposed BMP-6 KO mice to CS for 4 weeks and measured pulmonary inflammation and iron levels. Pulmonary mRNA levels of BMP-6 were decreased in smokers with and without COPD and in CS-exposed mice. Importantly, BMP-6 expression was lowest in severe COPD. Accordingly, protein levels of BMP-6 were decreased in patients with COPD. Lung function measurements demonstrated a decreased compliance and total lung capacity in BMP-6 KO mice, whereas lung histology was normal. Furthermore, BMP-6 KO mice displayed elevated iron levels and an aggravated CS-induced inflammatory response. These results suggest that BMP-6 is important for normal lung function and that downregulation of BMP-6-as observed in patients with COPD-contributes to pulmonary inflammation after CS exposure.
Collapse
|
8
|
Kim JH, Bae HC, Kim J, Lee H, Ryu WI, Son ED, Lee TR, Jeong SH, Son SW. HIF-1α-mediated BMP6 down-regulation leads to hyperproliferation and abnormal differentiation of keratinocytes in vitro. Exp Dermatol 2018; 27:1287-1293. [PMID: 30230035 DOI: 10.1111/exd.13785] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 09/05/2018] [Accepted: 09/09/2018] [Indexed: 12/16/2022]
Abstract
Hypoxia-inducible factor-1α (HIF-1α) has been reported to be up-regulated in psoriatic epidermis, resulting in increased proliferation and abnormal differentiation of human keratinocytes (KCs). However, the role of HIF-1α in psoriatic epidermis, which is mainly composed of KCs, is poorly understood. Here, we show that morphogenic protein 6 (BMP6) is down-regulated when HIF-1α is upregulated in patients with psoriasis skin lesions. HIF-1α overexpression in primary human KCs promoted proliferation and inhibited terminal differentiation. Furthermore, HIF1-α repressed the expression of BMP6 by binding directly to the hypoxia-response element (HRE) in the BMP6 promotor region, which shows that BMP6 is a novel target gene of HIF-1α. We also found that HIF-1α-mediated BMP6 suppression could alter the proliferation status by modulating the expression levels of cell cycle regulatory proteins and also affect the early differentiation of KCs. Therefore, we suggest that HIF-1α-dependent BMP6 suppression has a critical role in the induction of hyper-proliferation and abnormal differentiation in psoriatic KCs.
Collapse
Affiliation(s)
- Jin Hee Kim
- Department of Dermatology and Division of Brain Korea 21 Project for Biomedical Science, Korea University College of Medicine, Seoul, Korea
| | - Hyun Cheol Bae
- Department of Dermatology and Division of Brain Korea 21 Project for Biomedical Science, Korea University College of Medicine, Seoul, Korea
| | - Jaehyung Kim
- Department of Dermatology and Division of Brain Korea 21 Project for Biomedical Science, Korea University College of Medicine, Seoul, Korea
| | - Hana Lee
- Department of Dermatology and Division of Brain Korea 21 Project for Biomedical Science, Korea University College of Medicine, Seoul, Korea
| | - Woo-In Ryu
- Department of Dermatology and Division of Brain Korea 21 Project for Biomedical Science, Korea University College of Medicine, Seoul, Korea
| | - Eui Dong Son
- AmorePacific Corporation/R&D Center, Yongin-si, Gyeonggi-do, Korea
| | - Tae Ryong Lee
- AmorePacific Corporation/R&D Center, Yongin-si, Gyeonggi-do, Korea
| | - Sang Hoon Jeong
- Department of Dermatology and Division of Brain Korea 21 Project for Biomedical Science, Korea University College of Medicine, Seoul, Korea
| | - Sang Wook Son
- Department of Dermatology and Division of Brain Korea 21 Project for Biomedical Science, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
9
|
Monsuur HN, van den Broek LJ, Koolwijk P, Niessen FB, Gibbs S. Endothelial cells enhance adipose mesenchymal stromal cell-mediated matrix contraction via ALK receptors and reduced follistatin: Potential role of endothelial cells in skin fibrosis. J Cell Physiol 2018; 233:6714-6722. [PMID: 29345319 PMCID: PMC6056025 DOI: 10.1002/jcp.26494] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/17/2018] [Indexed: 12/18/2022]
Abstract
Abnormal cutaneous wound healing can lead to formation of fibrotic hypertrophic scars. Although several clinical risk factors have been described, the cross‐talk between different cell types resulting in hypertrophic scar formation is still poorly understood. The aim of this in vitro study was to investigate whether endothelial cells (EC) may play a role in skin fibrosis, for example, hypertrophic scar formation after full‐thickness skin trauma. Using a collagen/elastin matrix, we developed an in vitro fibrosis model to study the interaction between EC and dermal fibroblasts or adipose tissue‐derived mesenchymal stromal cells (ASC). Tissue equivalents containing dermal fibroblasts and EC displayed a normal phenotype. In contrast, tissue equivalents containing ASC and EC displayed a fibrotic phenotype indicated by contraction of the matrix, higher gene expression of ACTA2, COL1A, COL3A, and less secretion of follistatin. The contraction was in part mediated via the TGF‐β pathway, as both inhibition of the ALK4/5/7 receptors and the addition of recombinant follistatin resulted in decreased matrix contraction (75 ± 11% and 24 ± 8%, respectively). In conclusion, our study shows that EC may play a critical role in fibrotic events, as seen in hypertrophic scars, by stimulating ASC‐mediated matrix contraction via regulation of fibrosis‐related proteins.
Collapse
Affiliation(s)
- Hanneke N Monsuur
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Lenie J van den Broek
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Pieter Koolwijk
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Frank B Niessen
- Department of Plastic, Reconstructive and Hand Surgery, VU University Medical Center, Amsterdam, The Netherlands
| | - Susan Gibbs
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands.,Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| |
Collapse
|
10
|
An Interleukin-25-Mediated Autoregulatory Circuit in Keratinocytes Plays a Pivotal Role in Psoriatic Skin Inflammation. Immunity 2018; 48:787-798.e4. [DOI: 10.1016/j.immuni.2018.03.019] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 08/31/2017] [Accepted: 03/13/2018] [Indexed: 12/11/2022]
|
11
|
Lewis CJ, Mardaryev A, Sharpe D, Botchkareva N. Inhibition of bone morphogenetic protein signalling promotes wound healing in a human ex vivo model. EUROPEAN JOURNAL OF PLASTIC SURGERY 2015. [DOI: 10.1007/s00238-014-1031-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
12
|
The effects on dermal wound healing using novel peptide modified by bone morphogenic protein-2. Tissue Eng Regen Med 2014. [DOI: 10.1007/s13770-014-0037-4] [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] Open
|
13
|
Kim JD, Lee HW, Jin SW. Diversity is in my veins: role of bone morphogenetic protein signaling during venous morphogenesis in zebrafish illustrates the heterogeneity within endothelial cells. Arterioscler Thromb Vasc Biol 2014; 34:1838-45. [PMID: 25060789 DOI: 10.1161/atvbaha.114.303219] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Endothelial cells are a highly diverse group of cells which display distinct cellular responses to exogenous stimuli. Although the aptly named vascular endothelial growth factor-A signaling pathway is hailed as the most important signaling input for endothelial cells, additional factors also participate in regulating diverse aspects of endothelial behaviors and functions. Given this heterogeneity, these additional factors seem to play a critical role in creating a custom-tailored environment to regulate behaviors and functions of distinct subgroups of endothelial cells. For instance, molecular cues that modulate morphogenesis of arterial vascular beds can be distinct from those that govern morphogenesis of venous vascular beds. Recently, we have found that bone morphogenetic protein signaling selectively promotes angiogenesis from venous vascular beds without eliciting similar responses from arterial vascular beds in zebrafish, indicating that bone morphogenetic protein signaling functions as a context-dependent regulator during vascular morphogenesis. In this review, we will provide an overview of the molecular mechanisms that underlie proangiogenic effects of bone morphogenetic protein signaling on venous vascular beds in the context of endothelial heterogeneity and suggest a more comprehensive picture of the molecular mechanisms of vascular morphogenesis during development.
Collapse
Affiliation(s)
- Jun-Dae Kim
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (J.-D.K., H.W.L., S.-W.J.) and Department of Internal Medicine (J.-D.K., H.W.L., S.-W.J.), Yale University School of Medicine, New Haven, CT; and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea (S.-W.J.)
| | - Heon-Woo Lee
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (J.-D.K., H.W.L., S.-W.J.) and Department of Internal Medicine (J.-D.K., H.W.L., S.-W.J.), Yale University School of Medicine, New Haven, CT; and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea (S.-W.J.)
| | - Suk-Won Jin
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (J.-D.K., H.W.L., S.-W.J.) and Department of Internal Medicine (J.-D.K., H.W.L., S.-W.J.), Yale University School of Medicine, New Haven, CT; and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea (S.-W.J.).
| |
Collapse
|
14
|
Bone morphogenetic protein signaling suppresses wound-induced skin repair by inhibiting keratinocyte proliferation and migration. J Invest Dermatol 2013; 134:827-837. [PMID: 24126843 PMCID: PMC3945401 DOI: 10.1038/jid.2013.419] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 09/02/2013] [Accepted: 09/16/2013] [Indexed: 12/29/2022]
Abstract
Bone morphogenetic protein (BMP) signalling plays a key role in the control of skin development and postnatal remodelling by regulating keratinocyte proliferation, differentiation and apoptosis. To study the role of BMPs in wound-induced epidermal repair, we used transgenic mice overexpressing the BMP downstream component Smad1 under the control of a K14 promoter as an in vivo model, as well as ex vivo and in vitro assays. K14-caSmad1 mice exhibited retarded wound healing associated with significant inhibition of proliferation and increased apoptosis in healing wound epithelium. Furthermore, microarray and qRT-PCR analyses revealed decreased expression of a number of cytoskeletal/cell motility-associated genes including wound-associated keratins (Krt16, Krt17) and Myo5a, in the epidermis of K14-caSmad1 mice versus wild-type controls during wound healing. BMP treatment significantly inhibited keratinocyte migration ex vivo, and primary keratinocytes of K14-caSmad1 mice showed retarded migration compared to wild-type controls. Finally, siRNA-mediated silencing of Bmpr-1B in primary mouse keratinocytes accelerated cell migration and was associated with increased expression of Krt16, Krt17 and Myo5a compared to controls. Thus, this study demonstrates that BMPs inhibit keratinocyte proliferation, cytoskeletal organization and migration in regenerating skin epithelium during wound healing, and raises a possibility for using BMP antagonists for the management of chronic wounds.
Collapse
|
15
|
Tandon N, Cimetta E, Villasante A, Kupferstein N, Southall MD, Fassih A, Xie J, Sun Y, Vunjak-Novakovic G. Galvanic microparticles increase migration of human dermal fibroblasts in a wound-healing model via reactive oxygen species pathway. Exp Cell Res 2013; 320:79-91. [PMID: 24113575 DOI: 10.1016/j.yexcr.2013.09.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 09/03/2013] [Accepted: 09/21/2013] [Indexed: 12/13/2022]
Abstract
Electrical signals have been implied in many biological mechanisms, including wound healing, which has been associated with transient electrical currents not present in intact skin. One method to generate electrical signals similar to those naturally occurring in wounds is by supplementation of galvanic particles dispersed in a cream or gel. We constructed a three-layered model of skin consisting of human dermal fibroblasts in hydrogel (mimic of dermis), a hydrogel barrier layer (mimic of epidermis) and galvanic microparticles in hydrogel (mimic of a cream containing galvanic particles applied to skin). Using this model, we investigated the effects of the properties and amounts of Cu/Zn galvanic particles on adult human dermal fibroblasts in terms of the speed of wound closing and gene expression. The collected data suggest that the effects on wound closing are due to the ROS-mediated enhancement of fibroblast migration, which is in turn mediated by the BMP/SMAD signaling pathway. These results imply that topical low-grade electric currents via microparticles could enhance wound healing.
Collapse
Affiliation(s)
- Nina Tandon
- Columbia University, Department of Biomedical Engineering, 622 West 168th Street, MC 104B, New York 10027, NY, USA; The Cooper Union for the Advancement of Science and Art, Department of Electrical Engineering, 41 Cooper Square, New York 10003, NY, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Moura J, da Silva L, Cruz MT, Carvalho E. Molecular and cellular mechanisms of bone morphogenetic proteins and activins in the skin: potential benefits for wound healing. Arch Dermatol Res 2013; 305:557-69. [PMID: 23800970 DOI: 10.1007/s00403-013-1381-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 06/05/2013] [Accepted: 06/14/2013] [Indexed: 01/13/2023]
Abstract
Bone morphogenetic proteins (BMPs) and activins are phylogenetically conserved proteins, belonging to the transforming growth factor-β superfamily, that signal through the phosphorylation of receptor-regulated Smad proteins, activating different cell responses. They are involved in various steps of skin morphogenesis and wound repair, as can be evidenced by the fact that their expression is increased in skin injuries. BMPs play not only a role in bone regeneration but are also involved in cartilage, tendon-like tissue and epithelial regeneration, maintain vascular integrity, capillary sprouting, proliferation/migration of endothelial cells and angiogenesis, promote neuron and dendrite formation, alter neuropeptide levels and are involved in immune response modulation, at least in animal models. On the other hand, activins are involved in wound repair through the regulation of skin and immune cell migration and differentiation, re-epithelialization and granulation tissue formation, and also promote the expression of collagens by fibroblasts and modulate scar formation. This review aims at enunciating the effects of BMPs and activins in the skin, namely in skin development, as well as in crucial phases of skin wound healing, such as inflammation, angiogenesis and repair, and will focus on the effects of these proteins on skin cells and their signaling pathways, exploring the potential therapeutic approach of the application of BMP-2, BMP-6 and activin A in chronic wounds, particularly diabetic foot ulcerations.
Collapse
Affiliation(s)
- J Moura
- Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | | | | | | |
Collapse
|
17
|
Sox2 in the dermal papilla niche controls hair growth by fine-tuning BMP signaling in differentiating hair shaft progenitors. Dev Cell 2013; 23:981-94. [PMID: 23153495 DOI: 10.1016/j.devcel.2012.10.013] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 09/25/2012] [Accepted: 10/16/2012] [Indexed: 12/11/2022]
Abstract
How dermal papilla (DP) niche cells regulate hair follicle progenitors to control hair growth remains unclear. Using Tbx18(Cre) to target embryonic DP precursors, we ablate the transcription factor Sox2 early and efficiently, resulting in diminished hair shaft outgrowth. We find that DP niche expression of Sox2 controls the migration speed of differentiating hair shaft progenitors. Transcriptional profiling of Sox2 null DPs reveals increased Bmp6 and decreased BMP inhibitor Sostdc1, a direct Sox2 transcriptional target. Subsequently, we identify upregulated BMP signaling in knockout hair shaft progenitors and demonstrate that Bmp6 inhibits cell migration, an effect that can be attenuated by Sostdc1. A shorter and Sox2-negative hair type lacks Sostdc1 in the DP and shows reduced migration and increased BMP activity of hair shaft progenitors. Collectively, our data identify Sox2 as a key regulator of hair growth that controls progenitor migration by fine-tuning BMP-mediated mesenchymal-epithelial crosstalk.
Collapse
|
18
|
|
19
|
Gerhart J, Hayes C, Scheinfeld V, Chernick M, Gilmour S, George-Weinstein M. Myo/Nog cells in normal, wounded and tumor-bearing skin. Exp Dermatol 2012; 21:466-8. [PMID: 22621191 DOI: 10.1111/j.1600-0625.2012.01503.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Murine and human skin were examined for the presence of Myo/Nog cells that were originally discovered in the chick embryo by their expression of MyoD mRNA, noggin and the G8 antigen. Myo/Nog cells are the primary source of noggin in telogen hair follicles. They are scarce within the interfollicular dermis and absent in the epidermis. Within 24 h following epidermal abrasion, Myo/Nog cells increase in number in the follicles and appear in the wound. Myo/Nog cells are also recruited to the stroma of tumors formed from v-Ras-transformed keratinocytes (Ker/Ras). Human squamous cell carcinomas and malignant melanomas contain significantly more Myo/Nog cells than basal cell carcinomas. Myo/Nog cells are distinct from macrophages, granulocytes and cells expressing alpha smooth muscle actin in the tumor stroma. Myo/Nog cells may be modulators of skin homoeostasis and wound healing, and potential diagnostic and therapeutic targets in skin cancer.
Collapse
|
20
|
Wise LM, Inder MK, Real NC, Stuart GS, Fleming SB, Mercer AA. The vascular endothelial growth factor (VEGF)-E encoded by orf virus regulates keratinocyte proliferation and migration and promotes epidermal regeneration. Cell Microbiol 2012; 14:1376-90. [PMID: 22507661 DOI: 10.1111/j.1462-5822.2012.01802.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2012] [Revised: 03/27/2012] [Accepted: 04/05/2012] [Indexed: 12/20/2022]
Abstract
Vascular endothelial growth factor (VEGF)-A, a key regulator of cutaneous blood vessel formation, appears to have an additional role during wound healing, enhancing re-epithelialization. Orf virus, a zoonotic parapoxvirus, induces proliferative skin lesions that initiate in wounds and are characterized by extensive blood vessel formation, epidermal hyperplasia and rete ridge formation. The vascular changes beneath the lesion are largely due to viral-expressed VEGF-E. This study investigated using mouse skin models whether VEGF-E can induce epidermal changes such as that seen in the viral lesion. Injection of VEGF-E into normal skin increased the number of endothelial cells and blood vessels within the dermis and increased epidermal thickening and keratinocyte number. Injection of VEGF-E into wounded skin, which more closely mimics orf virus lesions, increased neo-epidermal thickness and area, promoted rete ridge formation, and enhanced wound re-epithelialization. Quantitative RT-PCR analysis showed that VEGF-E did not induce expression of epidermal-specific growth factors within the wound, but did increase matrix metalloproteinase (MMP)-2 and MMP-9 expression. In cell-based assays, VEGF-E induced keratinocyte migration and proliferation, responses that were inhibited by a neutralizing antibody against VEGF receptor (VEGFR)-2. These findings demonstrate that VEGF-E, both directly and indirectly, regulates keratinocyte function, thereby promoting epidermal regeneration.
Collapse
Affiliation(s)
- Lyn M Wise
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand.
| | | | | | | | | | | |
Collapse
|
21
|
Madhyastha H, Madhyastha R, Nakajima Y, Omura S, Maruyama M. Regulation of growth factors-associated cell migration by C-phycocyanin scaffold in dermal wound healing. Clin Exp Pharmacol Physiol 2011; 39:13-9. [DOI: 10.1111/j.1440-1681.2011.05627.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
22
|
Eliasson P, Andersson T, Aspenberg P. Influence of a single loading episode on gene expression in healing rat Achilles tendons. J Appl Physiol (1985) 2011; 112:279-88. [PMID: 21998267 DOI: 10.1152/japplphysiol.00858.2011] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mechanical loading stimulates tendon healing via mechanisms that are largely unknown. Genes will be differently regulated in loaded healing tendons, compared with unloaded, just because of the fact that healing processes have been changed. To avoid such secondary effects and study the effect of loading per se, we therefore studied the gene expression response shortly after a single loading episode in otherwise unloaded healing tendons. The Achilles tendon was transected in 30 tail-suspended rats. The animals were let down from the suspension to load their tendons on a treadmill for 30 min once, 5 days after tendon transection. Gene expression was studied by Affymetrix microarray before and 3, 12, 24, and 48 h after loading. The strongest response in gene expression was seen 3 h after loading, when 150 genes were up- or downregulated (fold change ≥2, P ≤ 0.05). Twelve hours after loading, only three genes were upregulated, whereas 38 were downregulated. Fewer than seven genes were regulated after 24 and 48 h. Genes involved in the inflammatory response were strongly regulated at 3 and 12 h after loading; this included upregulation of iNOS, PGE synthase, and IL-1β. Also genes involved in wound healing/coagulation, angiogenesis, and production of reactive oxygen species were strongly regulated by loading. Microarray results were confirmed for 16 selected genes in a repeat experiment (N = 30 rats) using real-time PCR. It was also confirmed that a single loading episode on day 5 increased the strength of the healing tendon on day 12. In conclusion, the fact that there were hardly any regulated genes 24 h after loading suggests that optimal stimulation of healing requires a mechanical loading stimulus every day.
Collapse
Affiliation(s)
- Pernilla Eliasson
- Orthopaedics, Department of Clinical and Experimental Medicine, Faculty of Health Science, Linköping University, Linköping, Sweden.
| | | | | |
Collapse
|
23
|
Yang X, Teng Y, Hou N, Fan X, Cheng X, Li J, Wang L, Wang Y, Wu X, Yang X. Delayed re-epithelialization in Ppm1a gene-deficient mice is mediated by enhanced activation of Smad2. J Biol Chem 2011; 286:42267-42273. [PMID: 21990361 DOI: 10.1074/jbc.m111.292284] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Protein phosphatase magnesium-dependent 1A (PPM1A), a protein serine/threonine phosphatase, controls several signal pathways through cleavage of phosphate from its substrates. However, the in vivo function of Ppm1a in mammals remains unknown. Here we reported that mice lacking Ppm1a developed normally but were impaired in re-epithelialization process during cutaneous wound healing. Specifically, complete or keratinocyte-specific deletion of Ppm1a led to delayed re-epithelialization with reduced keratinocyte migration upon wounding. We showed that this effect was the result of an increase in Smad2/3 phosphorylation in keratinocytes. Keratinocyte-specific Smad2 deficient mice displayed accelerated re-epithelialization with enhanced keratinocyte migration. Importantly, Smad2 and Ppm1a double mutant mice also exhibited accelerated re-epithelialization, demonstrating that the effect of Ppm1a on promoting re-epithelialization is mediated by Smad2 signaling. Furthermore, the decreased expression of specific integrins and matrix metalloproteinases (MMPs) may contribute to the retarded re-epithelialization in Ppm1a mutant mice. These data indicate that Ppm1a, through suppressing Smad2 signaling, plays a critical role in re-epithelialization during wound healing.
Collapse
Affiliation(s)
- Xue Yang
- State Key Laboratory of Proteomics, Genetic Laboratory of Development and Disease, Institute of Biotechnology, Beijing 100071, P.R. China; Model Organism Division, E-institutes of Shanghai Universities, Shanghai JiaoTong University, Shanghai 200025, P.R. China
| | - Yan Teng
- State Key Laboratory of Proteomics, Genetic Laboratory of Development and Disease, Institute of Biotechnology, Beijing 100071, P.R. China.
| | - Ning Hou
- State Key Laboratory of Proteomics, Genetic Laboratory of Development and Disease, Institute of Biotechnology, Beijing 100071, P.R. China
| | - Xiongwei Fan
- State Key Laboratory of Proteomics, Genetic Laboratory of Development and Disease, Institute of Biotechnology, Beijing 100071, P.R. China; College of Life Sciences, Hunan Normal University, Changsha 410081, P.R. China
| | - Xuan Cheng
- State Key Laboratory of Proteomics, Genetic Laboratory of Development and Disease, Institute of Biotechnology, Beijing 100071, P.R. China
| | - Jun Li
- State Key Laboratory of Proteomics, Genetic Laboratory of Development and Disease, Institute of Biotechnology, Beijing 100071, P.R. China
| | - Lijuan Wang
- State Key Laboratory of Proteomics, Genetic Laboratory of Development and Disease, Institute of Biotechnology, Beijing 100071, P.R. China
| | - Youliang Wang
- State Key Laboratory of Proteomics, Genetic Laboratory of Development and Disease, Institute of Biotechnology, Beijing 100071, P.R. China
| | - Xiushan Wu
- College of Life Sciences, Hunan Normal University, Changsha 410081, P.R. China
| | - Xiao Yang
- State Key Laboratory of Proteomics, Genetic Laboratory of Development and Disease, Institute of Biotechnology, Beijing 100071, P.R. China; Model Organism Division, E-institutes of Shanghai Universities, Shanghai JiaoTong University, Shanghai 200025, P.R. China.
| |
Collapse
|
24
|
Mouse 3T3 fibroblasts under the influence of fibroblasts isolated from stroma of human basal cell carcinoma acquire properties of multipotent stem cells. Biol Cell 2011; 103:233-48. [PMID: 21355851 DOI: 10.1042/bc20100113] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND INFORMATION Multipotent mesenchymal stem cells can participate in the formation of a microenvironment stimulating the aggressive behaviour of cancer cells. Moreover, cells exhibiting pluripotent ESC (embryonic stem cell) markers (Nanog and Oct4) have been observed in many tumours. Here, we investigate the role of cancer-associated fibroblasts in the formation of stem cell supporting properties of tumour stroma. We test the influence of fibroblasts isolated from basal cell carcinoma on mouse 3T3 fibroblasts, focusing on the expression of stem cell markers and plasticity in vitro by means of microarrays, qRT-PCR (quantitative real-time PCR) and immunohistochemistry. RESULTS We demonstrate the biological activity of the cancer stromal fibroblasts by influencing the 3T3 fibroblasts to express markers such as Oct4, Nanog and Sox2 and to show differentiation potential similar to mesenchymal stem cells. The role of growth factors such as IGF2 (insulin-like growth factor 2), FGF7 (fibroblast growth factor 7), LEP (leptin), NGF (nerve growth factor) and TGFβ (transforming growth factor β), produced by the stromal fibroblasts, is established to participate in their bioactivity. Uninduced 3T3 do not express the stem cell markers and show minimal differentiation potential. CONCLUSIONS Our observations indicate the pro-stem cell activity of cancer-associated fibroblasts and underline the role of epithelial-mesenchymal interaction in tumour biology.
Collapse
|
25
|
Hirt-Burri N, Ramelet AA, Raffoul W, de Buys Roessingh A, Scaletta C, Pioletti D, Applegate LA. Biologicals and fetal cell therapy for wound and scar management. ISRN DERMATOLOGY 2011; 2011:549870. [PMID: 22363853 PMCID: PMC3262533 DOI: 10.5402/2011/549870] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 03/16/2011] [Indexed: 01/01/2023]
Abstract
Few biopharmaceutical preparations developed from biologicals are available for tissue regeneration and scar management. When developing biological treatments with cellular therapy, selection of cell types and establishment of consistent cell banks are crucial steps in whole-cell bioprocessing. Various cell types have been used in treatment of wounds to reduce scar to date including autolog and allogenic skin cells, platelets, placenta, and amniotic extracts. Experience with fetal cells show that they may provide an interesting cell choice due to facility of outscaling and known properties for wound healing without scar. Differential gene profiling has helped to point to potential indicators of repair which include cell adhesion, extracellular matrix, cytokines, growth factors, and development. Safety has been evidenced in Phase I and II clinical fetal cell use for burn and wound treatments with different cell delivery systems. We present herein that fetal cells present technical and therapeutic advantages compared to other cell types for effective cell-based therapy for wound and scar management.
Collapse
Affiliation(s)
- Nathalie Hirt-Burri
- Cellular Therapy Unit, Department of Musculoskeletal Medicine, University Hospital of Lausanne, CHUV/UNIL, PAV 03, 1011 Lausanne, Switzerland
| | - Albert-Adrien Ramelet
- Office of Dermatology and Angiology, Place Benjamin Constant 2, 1005 Lausanne, Switzerland
| | - Wassim Raffoul
- Department of Plastic and Reconstructive Surgery, University Hospital of Lausanne, CHUV/UNIL, BH 10, 1011 Lausanne, Switzerland
| | - Anthony de Buys Roessingh
- Department of Pediatric Surgery, University Hospital of Lausanne, CHUV/UNIL, BH 10, 1011 Lausanne, Switzerland
| | - Corinne Scaletta
- Cellular Therapy Unit, Department of Musculoskeletal Medicine, University Hospital of Lausanne, CHUV/UNIL, PAV 03, 1011 Lausanne, Switzerland
| | - Dominique Pioletti
- Biomechanical Orthopedics Laboratory, Swiss Federal Institute of Technology, EPFL, 1015 Lausanne, Switzerland
| | - Lee Ann Applegate
- Cellular Therapy Unit, Department of Musculoskeletal Medicine, University Hospital of Lausanne, CHUV/UNIL, PAV 03, 1011 Lausanne, Switzerland
| |
Collapse
|
26
|
Westerweel PE, van Velthoven CTJ, Nguyen TQ, den Ouden K, de Kleijn DPV, Goumans MJ, Goldschmeding R, Verhaar MC. Modulation of TGF-β/BMP-6 expression and increased levels of circulating smooth muscle progenitor cells in a type I diabetes mouse model. Cardiovasc Diabetol 2010; 9:55. [PMID: 20858224 PMCID: PMC2954908 DOI: 10.1186/1475-2840-9-55] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Accepted: 09/21/2010] [Indexed: 12/03/2022] Open
Abstract
Background Diabetic patients experience exaggerated intimal hyperplasia after endovascular procedures. Recently it has been shown that circulating smooth muscle progenitor cells (SPC) contribute to intimal hyperplasia. We hypothesized that SPC differentiation would be increased in diabetes and focused on modulation of TGF-β/BMP-6 signaling as potential underlying mechanism. Methods We isolated SPC from C57Bl/6 mice with streptozotocin-induced diabetes and controls. SPC differentiation was evaluated by immunofluorescent staining for αSMA and collagen Type I. SPC mRNA expression of TGF-β and BMP-6 was quantified using real-time PCR. Intima formation was assessed in cuffed femoral arteries. Homing of bone marrow derived cells to cuffed arterial segments was evaluated in animals transplanted with bone marrow from GFP-transgenic mice. Results We observed that SPC differentiation was accelerated and numeric outgrowth increased in diabetic animals (24.6 ± 8.8 vs 8.3 ± 1.9 per HPF after 10 days, p < 0.05). Quantitative real-time PCR showed increased expression of TGF-β and decreased expression of the BMP-6 in diabetic SPC. SPC were MAC-3 positive, indicative of monocytic lineage. Intima formation in cuffed arterial segments was increased in diabetic mice (intima/media ratio 0.68 ± 0.15 vs 0.29 ± 0.06, p < 0.05). In GFP-chimeric mice, bone marrow derived cells were observed in the neointima (4.4 ± 3.3 cells per section) and particularly in the adventitia (43.6 ± 9.3 cells per section). GFP-positive cells were in part MAC-3 positive, but rarely expressed α-SMA. Conclusions In conclusion, in a diabetic mouse model, SPC levels are increased and SPC TGF-β/BMP-6 expression is modulated. Altered TGF-β/BMP-6 expression is known to regulate smooth muscle cell differentiation and may facilitate SPC differentiation. This may contribute to exaggerated intimal hyperplasia in diabetes as bone marrow derived cells home to sites of neointima formation.
Collapse
Affiliation(s)
- Peter E Westerweel
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Different options are reviewed in the field of musculoskeletal tissue reconstruction, from the addition of biological actors (cells, growth factors, biological or artificial scaffolds) to the application of gene therapy or tissue engineering. Growth factors can enable innovative solutions to treat such disease if we can extrapolate to soft tissue the promising results obtained in bone reconstruction with bone morphogenetic proteins. However, as in bone reconstruction, soft-tissue regeneration will depend on the drug delivery carrier, the scaffold for the newly formed tissue, the dose of growth factor and the animal model, which must all be explored before extrapolation to clinical problems.
Collapse
Affiliation(s)
- L Obert
- Orthopaedic, Traumatology, Plastic and Hand Surgery Unit, University of Franche Comté, CHU Jean Minjoz, Besancon, France.
| | | | | | | |
Collapse
|
28
|
BMP signaling induces cell-type-specific changes in gene expression programs of human keratinocytes and fibroblasts. J Invest Dermatol 2009; 130:398-404. [PMID: 19710687 DOI: 10.1038/jid.2009.259] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BMP signaling has a crucial role in skin development and homeostasis, whereas molecular mechanisms underlying its involvement in regulating gene expression programs in keratinocytes and fibroblasts remain largely unknown. We show here that several BMP ligands, all BMP receptors, and BMP-associated Smad1/5/8 are expressed in human primary epidermal keratinocytes and dermal fibroblasts. Treatment of both cell types by BMP-4 resulted in the activation of the BMP-Smad, but not BMP-MAPK pathways. Global microarray analysis revealed that BMP-4 treatment induces distinct and cell type-specific changes in gene expression programs in keratinocytes and fibroblasts, which are far more complex than the effects of BMPs on cell proliferation/differentiation described earlier. Furthermore, our data suggest that the potential modulation of cell adhesion, extracellular matrix remodeling, motility, metabolism, signaling, and transcription by BMP-4 in keratinocytes and fibroblasts is likely to be achieved by the distinct and cell-type-specific sets of molecules. Thus, these data provide an important basis for delineating mechanisms that underlie the distinct effects of the BMP pathway on different cell populations in the skin, and will be helpful in further establishing molecular signaling networks regulating skin homeostasis in health and disease.
Collapse
|
29
|
Barrientos S, Stojadinovic O, Golinko MS, Brem H, Tomic-Canic M. PERSPECTIVE ARTICLE: Growth factors and cytokines in wound healing. Wound Repair Regen 2008; 16:585-601. [PMID: 19128254 DOI: 10.1111/j.1524-475x.2008.00410.x] [Citation(s) in RCA: 2498] [Impact Index Per Article: 146.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Stephan Barrientos
- University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | | | | | | | | |
Collapse
|
30
|
Hirt-Burri N, Scaletta C, Gerber S, Pioletti DP, Applegate LA. Wound-healing gene family expression differences between fetal and foreskin cells used for bioengineered skin substitutes. Artif Organs 2008; 32:509-18. [PMID: 18638304 DOI: 10.1111/j.1525-1594.2008.00578.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
For tissue engineering, several cell types and tissues have been proposed as starting material. Allogenic skin products available for therapeutic usage are mostly developed with cell culture and with foreskin tissue of young individuals. Fetal skin cells offer a valuable solution for effective and safe tissue engineering for wounds due to their rapid growth and simple cell culture. By selecting families of genes that have been reported to be implicated in wound repair and particularly for scarless fetal wound healing including transforming growth factor-beta (TGF-beta) superfamily, extracellular matrix, and nerve/angiogenesis growth factors, we have analyzed differences in their expression between fetal skin and foreskin cells, and the same passages. Of the five TGF-beta superfamily genes analyzed by real-time reverse transcription-polymerase chain reaction, three were found to be significantly different with sixfold up-regulated for TGF-beta2, and 3.8-fold for BMP-6 in fetal cells, whereas GDF-10 was 11.8-fold down-regulated. For nerve growth factors, midkine was 36-fold down-regulated in fetal cells, and pleiotrophin was 4.76-fold up-regulated. We propose that fetal cells present technical and therapeutic advantages compared to foreskin cells for effective cell-based therapy for wound management, and overall differences in gene expression could contribute to the degree of efficiency seen in clinical use with these cells.
Collapse
Affiliation(s)
- Nathalie Hirt-Burri
- Department of Pediatric Surgery, University Hospital, CHUV, Lausanne, Switzerland.
| | | | | | | | | |
Collapse
|
31
|
Badreddine RJ, Wang KK. Barrett's esophagus: pathogenesis, treatment, and prevention. Gastrointest Endosc Clin N Am 2008; 18:495-512, ix. [PMID: 18674699 DOI: 10.1016/j.giec.2008.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Esophageal adenocarcinoma is the most common type of esophageal cancer seen in the United States and Western Europe. Barrett's esophagus (BE) is a well-known risk factor for esophageal adenocarcinoma and is believed to be found in 6% to 12% of patients undergoing endoscopy for gastroesophageal reflux disease and in more than 1% of all patients undergoing endoscopy. This article focuses on the pathogenesis, treatment, and prevention of BE.
Collapse
Affiliation(s)
- Rami J Badreddine
- Barrett's Esophagus Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
32
|
Milano F, van Baal JWPM, Buttar NS, Rygiel AM, de Kort F, DeMars CJ, Rosmolen WD, Bergman JJGHM, VAn Marle J, Wang KK, Peppelenbosch MP, Krishnadath KK. Bone morphogenetic protein 4 expressed in esophagitis induces a columnar phenotype in esophageal squamous cells. Gastroenterology 2007; 132:2412-21. [PMID: 17570215 DOI: 10.1053/j.gastro.2007.03.026] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2006] [Accepted: 03/01/2007] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Barrett's esophagus (BE) is a metaplastic condition in which normal squamous esophageal epithelium is replaced by columnar epithelium. It is proposed that one of the possible mechanisms is dedifferentiation of squamous epithelium into columnar epithelium. The pathophysiology through which this metaplasia occurs is unknown. A recent study by serial analysis of gene expression showed that bone morphogenetic protein 4 (BMP-4) is uniquely expressed in BE. In this study, the role of the BMP pathway in the metaplastic transformation of normal squamous cells into columnar cells was examined. METHODS Tissues from patients with esophagitis and BE and in an esophagitis-BE rat model were examined for the activation of the BMP pathway. Short-term cultures of primary normal squamous esophageal cells were treated with BMP-4, and cell biological changes were examined by Western blot analysis, immunohistochemistry, and microarrays. RESULTS In both human and rat tissues, the BMP pathway proved to be activated in esophagitis and BE. Upon incubation of squamous cell cultures with BMP-4, the cytokeratin expression pattern showed a shift that was consistent with columnar epithelium. Involvement of the BMP pathway was suggested by up-regulation of Phosphorylated-Smad 1/5/8 (P-Smad 1/5/8) that was effectively blocked by Noggin, a BMP antagonist. Comparison of the gene expression profiles of squamous cells, BMP-4-treated squamous cells, and BE cells showed a significant shift in the profile of the BMP-4-treated squamous cells toward that of the cultured BE cells. CONCLUSIONS These results suggest that the BMP pathway could play a role in the transformation of normal esophageal squamous cells into columnar cells.
Collapse
Affiliation(s)
- Francesca Milano
- Laboratory of Experimental Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Andreev K, Zenkel M, Kruse F, Jünemann A, Schlötzer-Schrehardt U. Expression of bone morphogenetic proteins (BMPs), their receptors, and activins in normal and scarred conjunctiva: Role of BMP-6 and activin-A in conjunctival scarring? Exp Eye Res 2006; 83:1162-70. [PMID: 16879818 DOI: 10.1016/j.exer.2006.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2006] [Revised: 05/19/2006] [Accepted: 06/02/2006] [Indexed: 11/30/2022]
Abstract
Bone morphogenetic proteins (BMPs) and activins are multifunctional growth factors, which also affect wound healing and tissue fibrosis. To determine their putative role in conjunctival wound healing and scarring, we investigated the expression of various BMPs, BMP receptors, and activins in normal and scarred human conjunctival tissue and in cultured human Tenon's capsule fibroblasts on the mRNA and protein level. Messenger RNA expression of BMP-2, BMP-3, BMP-4, BMP-5, BMP-6, BMP-7, the BMP receptors type I (BMPR-IA, BMPR-IB) and II (BMPR-II), and of activin A and B was investigated by semi-quantitative RT-PCR in normal conjunctival specimens and in scarred filtering blebs as well as in cultured Tenon's capsule fibroblasts obtained from patients with primary open-angle glaucoma (POAG), pseudoexfoliation (PEX) glaucoma and cataract. Immunohistochemistry was used to study the protein expression of BMP-2, BMP-4, BMP-6, BMP-7, and activin A in normal and scarred conjunctival tissue as well as in cultured Tenon's capsule fibroblasts. BMP-2, BMP-3, BMP-4, BMP-6, BMP-7, all BMP receptors, and activin A were expressed on the mRNA and protein level in conjunctival biopsies without showing any differences between groups of patients. The mRNA and protein expression of both BMP-6 and activin A was found to be significantly increased in scar tissue compared with normal conjunctiva and could be immunolocalized to epithelial cells, vascular endothelia, stromal fibroblasts, and macrophage-like cells. However, no significant increase in receptor gene expression was observed in scar tissue. With the exception of BMP-7, all growth factors and receptors were also expressed in cultured Tenon's fibroblasts without showing any differences between cultures derived from normal and scarred conjunctival specimens. These findings suggest various BMPs and activin A as components of the conjunctival cytokine meshwork regulating tissue homeostasis and wound healing and provide evidence that alterations in the expression of BMP-6 and activin A, in particular, are associated with conjunctival scarring. Modulation of BMP/activin activities may, therefore, be explored as new approaches for managing postoperative conjunctival scarring responses in glaucoma patients.
Collapse
|
34
|
Abstract
During wound healing, cells recreate functional structures to regenerate the injured tissue. Understanding the healing process is essential for the development of new concepts and the design of novel biomimetic approaches for delivery of cells, genes and growth factors to accelerate tissue regeneration. To this end, realistic experimental models and high-throughput diagnostics are necessary to understand the molecular mechanisms of healing and reveal the genetic networks that determine tissue repair versus regeneration. Following a brief overview of the biology of wound healing, this review covers the in vitro and in vivo models that are employed at present to study the healing process. Discussion then covers the application of high-throughput genomic and proteomic technologies in epithelial development, living skin substitutes and wound healing. Finally, this review provides a perspective on novel technologies that should be developed to facilitate the understanding of wound healing complications and the design of therapeutics that target the underlying deficiencies.
Collapse
Affiliation(s)
- Stelios T Andreadis
- University at Buffalo, The State University of New York (SUNY), Bioengineering Laboratory, Department of Chemical & Biological Engineering, 908 Furnas Hall, Amherst, NY 14260-4200, USA.
| |
Collapse
|
35
|
Abstract
Injury to the skin initiates a cascade of events including inflammation, new tissue formation, and tissue remodeling, that finally lead to at least partial reconstruction of the original tissue. Historically, animal models of repair have taught us much about how this repair process is orchestrated and, over recent years, the use of genetically modified mice has helped define the roles of many key molecules. Aside from conventional knockout technology, many ingenious approaches have been adopted, allowing researchers to circumvent such problems as embryonic lethality, or to affect gene function in a tissue- or temporal-specific manner. Together, these studies provide us with a growing source of information describing, to date, the in vivo function of nearly 100 proteins in the context of wound repair. This article focuses on the studies in which genetically modified mouse models have helped elucidate the roles that many soluble mediators play during wound repair, encompassing the fibroblast growth factor (FGF) and transforming growth factor-beta (TGF-beta) families and also data on cytokines and chemokines. Finally, we include a table summarizing all of the currently published data in this rapidly growing field. For a regularly updated web archive of studies, we have constructed a Compendium of Published Wound Healing Studies on Genetically Modified Mice which is avaialble at http://icbxs.ethz.ch/members/grose/woundtransgenic/home.html.
Collapse
Affiliation(s)
- Richard Grose
- London Research Institute Lab 214, Cancer Research UK, 61 Lincoln's Inn Fields, London WC2A 3PX, UK.
| | | |
Collapse
|
36
|
Abstract
Cutaneous wound healing is a complex process involving blood clotting, inflammation, new tissue formation, and finally tissue remodeling. It is well described at the histological level, but the genes that regulate skin repair have only partially been identified. Many experimental and clinical studies have demonstrated varied, but in most cases beneficial, effects of exogenous growth factors on the healing process. However, the roles played by endogenous growth factors have remained largely unclear. Initial approaches at addressing this question focused on the expression analysis of various growth factors, cytokines, and their receptors in different wound models, with first functional data being obtained by applying neutralizing antibodies to wounds. During the past few years, the availability of genetically modified mice has allowed elucidation of the function of various genes in the healing process, and these studies have shed light onto the role of growth factors, cytokines, and their downstream effectors in wound repair. This review summarizes the results of expression studies that have been performed in rodents, pigs, and humans to localize growth factors and their receptors in skin wounds. Most importantly, we also report on genetic studies addressing the functions of endogenous growth factors in the wound repair process.
Collapse
Affiliation(s)
- Sabine Werner
- Institute of Cell Biology, ETH Zurich, Hönggerberg, HPM D42, CH-8093 Zurich, Switzerland.
| | | |
Collapse
|
37
|
Abstract
Bone morphogenetic proteins (BMP) are members of the transforming growth factor-beta superfamily regulating a large variety of biologic responses in many different cells and tissues during embryonic development and postnatal life. BMP exert their biologic effects via binding to two types of serine/threonine kinase BMP receptors, activation of which leads to phosphorylation and translocation into the nucleus of intracellular signaling molecules, including Smad1, Smad5, and Smad8 ("canonical" BMP signaling pathway). BMP effects are also mediated by activation of the mitogen-activated protein (MAP) kinase pathway ("noncanonical" BMP Signaling pathway). BMP activity is regulated by diffusible BMP antagonists that prevent BMP interactions with BMP receptors thus modulating BMP effects in tissues. During skin development, BMPs its receptors and antagonists show stringent spatiotemporal expressions patterns to achieve proper regulation of cell proliferation and differentiation in the epidermis and in the hair follicle. In normal postnatal skin, BMP are involved in the control of epidermal homeostasis, hair follicle growth, and melanogenesis. Furthermore, BMP are implicated in a variety of pathobiologic processes in skin, including wound healing, psoriasis, and carcinogenesis. Therefore, BMPs represent new important players in the molecular network regulating homeostasis in normal and diseased skin. Pharmacologic modulation of BMP signaling may be used as a new approach for managing skin and hair disorders.
Collapse
Affiliation(s)
- Vladimir A Botchkarev
- Department of Dermatology, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
38
|
Rosendahl A, Pardali E, Speletas M, Ten Dijke P, Heldin CH, Sideras P. Activation of bone morphogenetic protein/Smad signaling in bronchial epithelial cells during airway inflammation. Am J Respir Cell Mol Biol 2002; 27:160-9. [PMID: 12151307 DOI: 10.1165/ajrcmb.27.2.4779] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are pleiotropic secreted proteins, structurally related to transforming growth factor (TGF)-beta and activins. BMPs play pivotal roles in the regulation of embryonic lung development and branching of airways and have recently been considered to influence inflammatory processes in adults due to their chemotactic activity on fibroblasts, myocytes, and inflammatory cells. In this study, we have investigated the possible involvement of BMPs in a model of experimental allergic-airway inflammation in situ using antibodies that detect activated Smad proteins, and have monitored the modulation of BMP ligands during the inflammatory response. Inflamed bronchial epithelial cells and a few scattered alveolar cells expressed levels of phosphorylated Smad1 (pSmad1/5), indicative of active BMP/Smad signaling. This was in contrast to healthy epithelium, which was devoid of immunoreactivity. A mechanistic explanation for increased pSmad1/5 staining during inflammation was provided by the upregulated expression of all the BMP type I receptors, i.e., activin receptor-like kinase (ALK)2, ALK3, and ALK6, in the inflamed bronchial epithelial cells. Furthermore, the mRNA and protein profiles for BMP ligands were significantly altered during airway inflammation with induction of BMP2, BMP4, and BMP6, and downregulation of BMP5 and BMP7. Collectively, our data demonstrate for the first time active BMP/Smad signaling during airway inflammation in bronchial epithelial cells and thus raise the possibility that BMPs could play a determining role in respiratory pathophysiology.
Collapse
|
39
|
|
40
|
Zirngibl RA, Senis Y, Greer PA. Enhanced endotoxin sensitivity in fps/fes-null mice with minimal defects in hematopoietic homeostasis. Mol Cell Biol 2002; 22:2472-86. [PMID: 11909942 PMCID: PMC133716 DOI: 10.1128/mcb.22.8.2472-2486.2002] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The fps/fes proto-oncogene encodes a cytoplasmic protein tyrosine kinase implicated in growth factor and cytokine receptor signaling and thought to be essential for the survival and terminal differentiation of myeloid progenitors. Fps/Fes-null mice were healthy and fertile, displayed slightly reduced numbers of bone marrow myeloid progenitors and circulating mature myeloid cells, and were more sensitive to lipopolysaccharide (LPS). These phenotypes were rescued using a fps/fes transgene. This confirmed that Fps/Fes is involved in, but not required for, myelopoiesis and that it plays a role in regulating the innate immune response. Bone marrow-derived Fps/Fes-null macrophages showed no defects in granulocyte-macrophage colony-stimulating factor-, interleukin 6 (IL-6)-, or IL-3-induced activation of signal transducer and activator of transcription 3 (Stat3) and Stat5A or LPS-induced degradation of I kappa B or activation of p38, Jnk, Erk, or Akt.
Collapse
Affiliation(s)
- Ralph A Zirngibl
- Division of Cancer Biology and Genetics, Queen's University Cancer Research Institute, Queen's University, Kingston, Ontario K7L-3N6, Canada
| | | | | |
Collapse
|
41
|
van Zuijlen PPM, Angeles AP, Kreis RW, Bos KE, Middelkoop E. Scar assessment tools: implications for current research. Plast Reconstr Surg 2002; 109:1108-22. [PMID: 11884845 DOI: 10.1097/00006534-200203000-00052] [Citation(s) in RCA: 112] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Scarring is considered a major medical problem that leads to cosmetic and functional sequelae. Scar tissue is clinically distinguished from normal skin by an aberrant color, rough surface texture, increased thickness (hypertrophy), occurrence of contraction, and firmness. Marked histologic differences are the change in dermal architecture and the presence of cells such as the myofibroblast. Many assessment tools are available for analysis of pathologic conditions of the skin; however, there is no general agreement as to the most appropriate tools for evaluation of scar tissue. This review critically discusses currently available objective measurement tools, subjective assessment tools, and potential devices that may be available in the future for scar assessment.
Collapse
Affiliation(s)
- Paul P M van Zuijlen
- Burn Center, Department of Surgery, Red Cross Hospital, Vondellaan 13, 1942 LE Beverwijk, The Netherlands.
| | | | | | | | | |
Collapse
|
42
|
Mikus D, Sikiric P, Seiwerth S, Petricevic A, Aralica G, Druzijancic N, Rucman R, Petek M, Pigac B, Perovic D, Kolombo M, Kokic N, Mikus S, Duplancic B, Fattorini I, Turkovic B, Rotkvic I, Mise S, Prkacin I, Konjevoda P, Stambuk N, Anic T. Pentadecapeptide BPC 157 cream improves burn-wound healing and attenuates burn-gastric lesions in mice. Burns 2001; 27:817-827. [PMID: 11718984 DOI: 10.1016/s0305-4179(01)00055-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The effects of the gastric pentadecapeptide BPC 157 were investigated when administered topically or systemically in burned mice. This agent is known to have a beneficial effect in a variety of models of gastrointestinal lesions, as well as on wound or fracture healing. Deep partial skin thickness burns (1.5x1.5 cm) covering 20% of total body area, were induced under anesthesia on the back of mice by controlled burning and gastric lesions were assessed 1, 2, 3, 7, 14 and 21 days following injury. The first application of BPC 157 was immediately following burning, and thereafter, once daily, until 24 h before sacrifice. In the initial experiments, exposure to direct flame for 5 s, the BPC 157 was applied at 10 microg or 10 ng/kg b.w. intraperitoneally (i.p.) by injection or alternatively, topically, at the burn, as a thin layer of cream (50 microg of BPC 157 dissolved in 2 ml of distilled water was mixed with 50 g of commercial neutral cream (also used as local vehicle-control)), while silver sulfadiazine 1% cream was a standard agent acting locally. Others received no local medication: they were treated i.p. by injection of distilled water (distilled water-control) or left without any medication (control). In subsequent experiments involving deeper burns (direct flame for 7 s), BPC 157 creams (50 microg, 5 microg, 500 ng, 50 ng or 5 ng of BPC 157 dissolved in 2 ml of distilled water was mixed with 50 g of commercial neutral cream), or vehicle as a thin layer of cream, were applied topically, at the burn. Compared with untreated controls, in both experiments, in the BPC 157 cream-treated mice all parameters of burn healing were improved throughout the experiment: less edema was observed and inflammatory cell numbers decreased. Less necrosis was seen with an increased number of capillaries along with an advanced formation of dermal reticulin and collagen fibers. An increased number of preserved follicles were observed. Two weeks after injury, BPC 157 cream-treated mice completely reversed the otherwise poor re-epithelization ratio noted in the untreated control or mice treated with vehicle only. Tensiometry investigation showed an increased breaking strength and relative elongation of burned skin, while water content in burned skin decreased. This was, however, not the case with the vehicle or silver sulfadiazine. Relative to the control values, in silver sulfadiazine cream-treated mice, only collagen fiber formation was increased, in addition to a decreased inflammatory cell number. Relative to control values, BPC 157 given i.p. decreased the number of inflammatory cells, lowered water content in burned skin, and raised breaking strength and relative elongation of burned skin during tensiometry. Through the experimental period, gastric lesions were continuously noted in all thermally injured mice left without local medication and they were consistently attenuated only by BPC 157 treatments: either given i.p. (at either dose), or given locally (at either concentration). Other treatments (i.e. local treatment with silver sulfadiazine cream or neutral cream in mice subjected for 5 s to direct flame), led to only poor, if any attenuation. This stable gastric pentadecapeptide appears to be active and gives a stimulation to burn healing at the defect site. The agent may act by causing an upregulation of the growth factors, as well as influencing other local factors.
Collapse
Affiliation(s)
- D Mikus
- Department of Pharmacology, Medical Faculty, University of Zagreb, Salata 11, P.O. Box 916, 10000, Zagreb, Croatia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Wach S, Schirmacher P, Protschka M, Blessing M. Overexpression of bone morphogenetic protein-6 (BMP-6) in murine epidermis suppresses skin tumor formation by induction of apoptosis and downregulation of fos/jun family members. Oncogene 2001; 20:7761-9. [PMID: 11753654 DOI: 10.1038/sj.onc.1204962] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2001] [Revised: 09/01/2001] [Accepted: 09/13/2001] [Indexed: 11/09/2022]
Abstract
Bone morphogenetic protein-6 (BMP-6) is a member of the transforming growth factor-beta superfamily. In murine skin, BMP-6 is highly expressed in postmitotic keratinocytes from day 15.5 p.c. till day 6 p.p. Expression in adult skin remains at very low levels, but pathological conditions such as wounding induce the expression of BMP-6. We demonstrate that tumor promotion by TPA (12-O-tetradecanoylphorbol-13-acetate) also induces expression of BMP-6 in suprabasal keratinocytes. This induction is due to post-transcriptional regulation since the level of BMP-6 mRNA remained unchanged. We performed two-stage skin carcinogenesis experiments with transgenic mice epidermally overexpressing BMP-6. These mice display augmented mitotic indices in normal and TPA-treated epidermis when compared to controls. Despite this hyperproliferation, BMP-6 transgenics showed a delayed development and strong suppression of benign and malignant skin tumor formation. In order to resolve this paradox we determined apoptotic frequencies as well as the expression of constituents of AP-1 (activator protein-1) which is essential for tumor promotion. A higher rate of apoptotic keratinocytes was detectable in transgenic mice versus controls and a downregulation of mRNA for jun/fos family members in transgenic skin after TPA-treatment. Thus expression of BMP-6 augments apoptosis and downregulates the transcription of AP-1 family members thereby establishing tumor resistance.
Collapse
Affiliation(s)
- S Wach
- I. Medical Department, Section of Pathophysiology, Johannes Gutenberg-University, 55131 Mainz, Germany
| | | | | | | |
Collapse
|
44
|
Wankell M, Munz B, Hübner G, Hans W, Wolf E, Goppelt A, Werner S. Impaired wound healing in transgenic mice overexpressing the activin antagonist follistatin in the epidermis. EMBO J 2001; 20:5361-72. [PMID: 11574468 PMCID: PMC125651 DOI: 10.1093/emboj/20.19.5361] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recently, we demonstrated a strong upregulation of activin expression after skin injury. Furthermore, overexpression of this transforming growth factor beta family member in the skin of transgenic mice caused dermal fibrosis, epidermal hyperthickening and enhanced wound repair. However, the role of endogenous activin in wound healing has not been determined. To address this question we overexpressed the soluble activin antagonist follistatin in the epidermis of transgenic mice. These animals were born with open eyes, and the adult mice had larger ears, longer tails and reduced body weight compared with non-transgenic littermates. Their skin was characterized by a mild dermal and epidermal atrophy. After injury, a severe delay in wound healing was observed. In particular, granulation tissue formation was significantly reduced, leading to a major reduction in wound breaking strength. The wounds, however, finally healed, and the resulting scar area was smaller than in control animals. These results implicate an important function of endogenous activin in the control of wound repair and scar formation.
Collapse
Affiliation(s)
- Miriam Wankell
- Institute of Cell Biology, ETH Zürich, Hönggerberg, CH-8093 Zürich, Switzerland, Max-Planck-Institute of Biochemistry, Am Klopferspitz 18a, 82152 Martinsried, Switch-Biotech AG, Fraunhoferstrasse 10, 82152 Martinsried and Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University, D-81377 Munich, Germany Present address: Department of Molecular Pharmacology, Stanford University Medical School, 300 Pasteur Drive, Stanford, CA 94305-5332, USA Present address: Quintiles GmbH, Mühlweg 2, D-82054 Sauerlach, Germany Corresponding author at: Institute of Cell Biology, ETH Zürich, Hönggerberg, CH-8093, Zürich, Switzerland e-mail:
| | - Barbara Munz
- Institute of Cell Biology, ETH Zürich, Hönggerberg, CH-8093 Zürich, Switzerland, Max-Planck-Institute of Biochemistry, Am Klopferspitz 18a, 82152 Martinsried, Switch-Biotech AG, Fraunhoferstrasse 10, 82152 Martinsried and Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University, D-81377 Munich, Germany Present address: Department of Molecular Pharmacology, Stanford University Medical School, 300 Pasteur Drive, Stanford, CA 94305-5332, USA Present address: Quintiles GmbH, Mühlweg 2, D-82054 Sauerlach, Germany Corresponding author at: Institute of Cell Biology, ETH Zürich, Hönggerberg, CH-8093, Zürich, Switzerland e-mail:
| | - Griseldis Hübner
- Institute of Cell Biology, ETH Zürich, Hönggerberg, CH-8093 Zürich, Switzerland, Max-Planck-Institute of Biochemistry, Am Klopferspitz 18a, 82152 Martinsried, Switch-Biotech AG, Fraunhoferstrasse 10, 82152 Martinsried and Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University, D-81377 Munich, Germany Present address: Department of Molecular Pharmacology, Stanford University Medical School, 300 Pasteur Drive, Stanford, CA 94305-5332, USA Present address: Quintiles GmbH, Mühlweg 2, D-82054 Sauerlach, Germany Corresponding author at: Institute of Cell Biology, ETH Zürich, Hönggerberg, CH-8093, Zürich, Switzerland e-mail:
| | - Wolfgang Hans
- Institute of Cell Biology, ETH Zürich, Hönggerberg, CH-8093 Zürich, Switzerland, Max-Planck-Institute of Biochemistry, Am Klopferspitz 18a, 82152 Martinsried, Switch-Biotech AG, Fraunhoferstrasse 10, 82152 Martinsried and Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University, D-81377 Munich, Germany Present address: Department of Molecular Pharmacology, Stanford University Medical School, 300 Pasteur Drive, Stanford, CA 94305-5332, USA Present address: Quintiles GmbH, Mühlweg 2, D-82054 Sauerlach, Germany Corresponding author at: Institute of Cell Biology, ETH Zürich, Hönggerberg, CH-8093, Zürich, Switzerland e-mail:
| | - Eckhard Wolf
- Institute of Cell Biology, ETH Zürich, Hönggerberg, CH-8093 Zürich, Switzerland, Max-Planck-Institute of Biochemistry, Am Klopferspitz 18a, 82152 Martinsried, Switch-Biotech AG, Fraunhoferstrasse 10, 82152 Martinsried and Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University, D-81377 Munich, Germany Present address: Department of Molecular Pharmacology, Stanford University Medical School, 300 Pasteur Drive, Stanford, CA 94305-5332, USA Present address: Quintiles GmbH, Mühlweg 2, D-82054 Sauerlach, Germany Corresponding author at: Institute of Cell Biology, ETH Zürich, Hönggerberg, CH-8093, Zürich, Switzerland e-mail:
| | - Andreas Goppelt
- Institute of Cell Biology, ETH Zürich, Hönggerberg, CH-8093 Zürich, Switzerland, Max-Planck-Institute of Biochemistry, Am Klopferspitz 18a, 82152 Martinsried, Switch-Biotech AG, Fraunhoferstrasse 10, 82152 Martinsried and Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University, D-81377 Munich, Germany Present address: Department of Molecular Pharmacology, Stanford University Medical School, 300 Pasteur Drive, Stanford, CA 94305-5332, USA Present address: Quintiles GmbH, Mühlweg 2, D-82054 Sauerlach, Germany Corresponding author at: Institute of Cell Biology, ETH Zürich, Hönggerberg, CH-8093, Zürich, Switzerland e-mail:
| | - Sabine Werner
- Institute of Cell Biology, ETH Zürich, Hönggerberg, CH-8093 Zürich, Switzerland, Max-Planck-Institute of Biochemistry, Am Klopferspitz 18a, 82152 Martinsried, Switch-Biotech AG, Fraunhoferstrasse 10, 82152 Martinsried and Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University, D-81377 Munich, Germany Present address: Department of Molecular Pharmacology, Stanford University Medical School, 300 Pasteur Drive, Stanford, CA 94305-5332, USA Present address: Quintiles GmbH, Mühlweg 2, D-82054 Sauerlach, Germany Corresponding author at: Institute of Cell Biology, ETH Zürich, Hönggerberg, CH-8093, Zürich, Switzerland e-mail:
| |
Collapse
|